Neoplasia Flashcards

1
Q

What is cancer,what are the characteristics of cancer,state the hallmarks of cancer

The Epi genetic changes may themselves stem from acquired mutations in genes that regulate such modifi­ cations.
These genetic and epigenetic changes alter the expression or function of key genes that regulate funda­ mental cellular processes, such as growth, survival, and senescence.
True or false

All tumors are clonal- explain

Darwinian selection also plays a role in the pro­ gression and recurrence of cancers.
True or false

The genetic alterations that give rise to these hallmarks of cancers are sustained and enabled by the development of genomic instability, adding fuel to the fire.Genomic instability is a characteristic of cancer
What is genomic instability?

A

• Cancer is a genetic disorder caused by DNA mutations that are acquired spontaneously or induced by environmental insults.

-In addition, cancers frequently show epigenetic changes, such as focal
increases in DNA methylation and alterations in histone modifications, -Thesegeneticalterationsareheritable,beingpassedtodaugh- ter cells upon cell division. As a result, cells harboring these alterations are subject to darwinian selection (survival of the fittest, arguably the most important scientific concept yet conceived), with cells bearing mutations that provide them with growth or survival advantages outcompeting their neighbors and thus coming to dominate the popu­ lation.

Accumulation of mutations gives rise to a set of properties that have been called hallmarks of cancer. These include (1) self­sufficiency in growth signals whereby the growth of cancers becomes autonomous and is unregulated by physiologic cues; (2) lack of response to growth inhibitory signals that control non­neoplastic cellular proliferations such as hyperplasias; (3) evasion of cell death, allowing cancer cells to survive under conditions that induce apoptosis in normal cells; (4) limitless repli­ cative potential, thus making cancer cells immortal; (5) development of angiogenesis(will form new blood vessels)to sustain the growth of cancer cells; (6) ability to invade local tissues and spread to distant sites; (7) reprogramming of metabolic pathways—specifically, a switch to aerobic glycolysis even when there is abundant oxygen; and (8) ability to evade the immune system.

The increased tendency for DNA mutations (changes) and other genetic changes to occur during cell division. Genomic instability is caused by defects in certain processes that control the way cells divide

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

What is neoplasia ,neoplasm
Why are neoplastic cells said to be transformed ,when is a tumor said to be benign or malignant

Neoplasms therefore enjoy a certain degree of autonomy and tend to increase in size regardless of their local environment. True or false

Why isn’t the autonomy of cancer cells complete?

Of note, however, benign tumors can produce more than localized lumps, and sometimes they are responsible for serious disease.
True or false

A

Neoplasia literally means “new growth.” Neoplastic cells are said to be transformed because they continue to repli­ cate, apparently oblivious to the regulatory influences that control normal cell growth.

Their autonomy is by no means complete, however. Some neoplasms require endocrine support, and such dependencies sometimes can be exploited therapeutically. All neoplasms depend on the host for their nutrition and blood supply.

a neoplasm often is referred to as a tumor, and the study of tumors is called oncology (from oncos, “tumor,” and logos, “study of”). Among tumors, the division of neoplasms into benign and malig­ nant categories is based on a judgment of a tumor’s poten­ tial clinical behavior.
• A tumor is said to be benign when its microscopic and gross characteristics are considered to be relatively inno­ cent, implying that it will remain localized and is ame­nable (easily controlled or responsive ) to local surgical removal; the patient generally survives.
• Malignant tumors are collectively referred to as cancers, derived from the Latin word for “crab”—that is, they adhere to any part that they seize in an obstinate manner, similar to a crab’s behavior. Malignant, as applied to a neoplasm, implies that the lesion can invade and destroy adjacent structures and spread to distant sites (metasta­ size) to cause death. Not all cancers pursue so deadly a course. The most aggressive are also some of the most curable, but the designation malignant constitutes a red flag.

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

What are the basic components of tumors

What is the importance of the non neoplastic stroma

Although the biologic behavior of tumors largely reflects the behavior of the parenchymal cells, there has been a growing realization that stromal cells and neoplastic cells carry on a two­way conversation that influences the growth of the tumor.
True or false

The tumor derived its name from which component?

A

All tumors, benign and malignant, have two basic com­ ponents: (1) the parenchyma, made up of transformed or neoplastic cells, and (2) the supporting, host­derived, non­ neoplastic stroma, made up of connective tissue, blood vessels, and host­derived inflammatory cells.

The paren­ chyma of the neoplasm largely determines its biologic behavior, and it is this component from which the tumor
derives its name.

The stroma is crucial to the growth of the neoplasm, since it carries the blood supply and provides support for the growth of parenchymal cells.

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

What is a fibroma and a chondroma

How is The nomenclature of benign epithelial tumors classified?

What is adenoma ,papilloma ,a polyp ,cystadenoma

Polyp is commonly is used for benign tumors, some malignant tumors also may grow as polyps, whereas other polyps (such as nasal polyps) are not neo­ plastic but inflammatory in origin. True or false

A

general, benign tumors are designated by attaching the suffix -oma to the cell type from which the tumor arises.

A benign tumor arising in fibrous tissue is a fibroma;
a benign cartilaginous tumor is a chondroma.

The nomenclature of benign epithelial tumors is more complex. They are classi­ fied sometimes on the basis of their microscopic pattern and sometimes on the basis of their macroscopic pattern. Others are classified by their cells of origin.
For instance, the term adenoma is generally applied to benign epithelial neoplasms producing gland patterns and to neoplasms derived from glands but not necessarily exhibiting glandular patterns. A benign epithelial neo­ plasm arising from renal tubule cells and growing in glandlike patterns is termed an adenoma, as is a mass of benign epithelial cells that produces no glandular patterns but has its origin in the adrenal cortex.

Papillomas are benign epithelial neoplasms, growing on any surface, that produce microscopic or macroscopic finger­like fronds.

A polyp is a mass that projects above a mucosal surface, as in the gut, to form a macroscopically visible structure .

Cystadenomas are hollow cystic masses that typically arise in the ovary.

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

What are sarcomas ,lymphomas

Sarcomas are des­ ignated by ?

What are carcinomas

Epithelia of the body are derived from all three germ cell layers true or false

Furthermore, meso­ derm may give rise to carcinomas (epithelial), sarcomas (mesenchymal), and hematolymphoid tumors (leuke­ mias and lymphomas). True or false

What is a squamous cell carcinomas ,what is a poorly differentiated or undifferentiated carcinoma

A

Malignant neoplasms arising in “solid” mesenchymal tissues or its derivatives are called sarcomas,
whereas those arising from the mesenchymal cells of the blood are called leukemias or lymphomas.

Sarcomas are des­ ignated by the cell type of which they are composed, which is presumably their cell of origin.

Thus, a cancer of fibrous tissue origin is a fibrosarcoma, and a malignant neoplasm composed of chondrocytes is a chondrosarcoma.

• While the epithelia of the body are derived from all three germ cell layers, malignant neoplasms of epithelial cells are called carcinomas regardless of the tissue of origin.

Thus, a malignant neoplasm arising in the renal tubular epithelium (mesoderm) is a carcinoma, as are the cancers arising in the skin (ectoderm) and lining epithelium of the gut (endoderm).

• Carcinomas are subdivided further. Carcinomas that grow in a glandular pattern are called adenocarcinomas, and those that produce squamous cells are called squa- mous cell carcinomas. Sometimes the tissue or organ of origin can be identified, as in the designation of renal cell adenocarcinoma. Sometimes the tumor shows little or no differentiation and must be called poorly differenti- ated or undifferentiated carcinoma.

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

The transformed cells in a neoplasm, whether benign or malignant, often resemble each other, as though all had been derived from a single progenitor, consistent with the monoclonal origin of tumors.
True or false
What are mixed tumors and give an example
Explain the types of mixed tumors

What is a teratoma
Where does it originate from

Germ cells have the capacity to differenti­ ate into any of the cell types found in the adult body; not surprisingly, therefore, they may give rise to neoplasms that mimic, in helter­skelter fashion, bits of bone, epithe­ lium, muscle, fat, nerve, and other tissues
True or false

What is hamartoma
Oh histopathological examination what is seen ?

What is choristoma

Hamartomas have tradi­ tionally been considered developmental malformations, but some genetic studies have shown the presence of acquired translocations, suggesting a neoplastic origin. True or false

A

In some unusual instances, however, the tumor cells undergo divergent differentiation, creating so­called mixed tumors. The best example is mixed tumor of salivary gland. These tumors have obvious epi­ thelial components dispersed throughout a fibromyxoid stroma, sometimes harboring islands of cartilage or bone

All of these diverse elements are thought to derive from epithelial cells or myoepithelial cells, or both, and the preferred designation for these neoplasms is pleomorphic adenoma.
Fibroadenoma of the female breast is another common mixed tumor. This benign tumor contains a mixture of proliferating ductal elements (adenoma) embedded in a loose fibrous tissue (fibroma). Although only the fibrous component is neoplastic, the term fibroad- enoma remains in common usage.

Teratoma is a special type of mixed tumor that contains recognizable mature or immature cells or tissues represen­ tative of more than one germ cell layer and sometimes all three.

Teratomas originate from totipotential germ cells such as those normally present in the ovary and testis and sometimes abnormally present in sequestered midline embryonic rests.
.
For example, the terms lymphoma, mesothelioma, melanoma, and seminoma are used for malig­ nant neoplasms. .
There are other instances of confusing terminology:
• Hamartoma is a mass of disorganized tissue indigenous to the particular site.

Histopathologic examination may show a mass of mature but disorganized hepatic cells, blood vessels, and possibly bile ducts within the liver, or a nodule in the lung containing islands of cartilage, bronchi, and blood vessels.

• Choristoma is a congenital anomaly consisting of a het­erotopic (heterotopia is the presence of a particular tissue type at a non-physiological site, but usually co-existing with original tissue in its correct anatomical location) rest of cells.

For example, a small nodule of well­developed and normally organized pancreatic tissue may be found in the submucosa of the stomach, duodenum, or small intestine.

This heterotopic rest may be replete with islets of Langerhans and exocrine glands. The designation -oma, connoting a neoplasm, imparts to the heterotopic rest a gravity far beyond its usual trivial significance.

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

State the name of a benign and malignant tumor that arises from some named tissues or origin,

There are tumors that are only malignant 
Name them (they’re seven)
A

Tissue or origin
Composed of One Parenchymal Cell Type
Connective tissue and derivatives:
Fibroma -benign

Fibrosarcoma-malignant

Lipoma-benign

Liposarcoma-malignant

Chondroma-benign

Chondrosarcoma-malignant

Osteoma-benign
Osteogenic sarcoma-malignant

Endothelial and related tissues
Blood vessels-Hemangioma(benign)-Angiosarcoma(malignant)

Lymph vessels-Lymphangioma(benign)-Lymphangiosarcoma(malignant )

Mesothelium-Mesothelioma(malignant )

Brain coverings-Meningioma(benign)-
Invasive meningioma(malignant )

Blood cells and related cells
Hematopoietic cells -Leukemias (malignant)
Lymphoid tissue -Lymphomas(malignant)

Muscle
Smooth- Leiomyoma(benign) Leiomyosarcoma (malignant)
Striated -Rhabdomyoma (benign)
Rhabdomyosarcoma(malignant)

Tumors of epithelial origin
Stratified squamous-Squamous cell papilloma(benign)

Squamous cell or epidermoid carcinoma
(Malignant)
Basal cells of skin or adnexa -Basal cell carcinoma(malignant)

Epithelial lining of glands or ducts-Adenoma(benign)
Adenocarcinoma-(malignant)

Papilloma(benign)

Papillary carcinomas(malignant)

Cystadenoma (benign)
Cystadenocarcinoma(malignant)

Respiratory passages-Bronchial adenoma(benign)

Bronchogenic carcinoma
(Malignant)

Renal epithelium-Renal tubular adenoma(benign), Renal cell carcinoma(malignant)

Liver cells-Liver cell adenoma,hepatocelluoar carcinoma
Urinary tract epithelium (transitional) -Urothelial papilloma(benign), Urothelial carcinoma
Placental epithelium-Hydatidiform mole(benign), Choriocarcinoma(malignant)

Testicular epithelium (germ cells)
-Seminoma(malignant)
Embryonal carcinoma(malignant)
   Tumors of melanocytes -Nevus (benign)
Malignant melanoma(malignant)

More Than One Neoplastic Cell Type—Mixed Tumors, Usually Derived from One Germ Cell Layer
Salivary glands -Pleomorphic adenoma (mixed tumor salivary gland )
-Malignant mixed tumor of of salivary gland(malignant)

Renal anlage -Wilms tumor(malignant)

More Than One Neoplastic Cell Type Derived from More Than One Germ Cell Layer—Teratogenous:
Totipotential cells in gonads or in embryonic rests
Mature teratoma and dermoid cyst (benign)
Immature teratoma and teratocarcinoma(malignant)

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

What are four characteristics that differentiate benign and malignant neoplasms

Explain differentiation and anaplasia
What is lipoma and chondroma made up of

benign tumors appear to be genetically “simple,” harboring fewer muta­ tions than cancers, and genetically stable, changing little in genotype over time.
The latter feature probably explains why benign tumors such as lipomas and leiomyomas transform to malignancies rarely, if at all.
True or false

The more differentiated the tumor cell, the more com­ pletely it retains the functional capabilities of its normal counterparts.
True or false
It is now known, however, that at least some cancers arise from stem cells in tissues; in these tumors, failure of differentiation, rather than dedifferentiation of specialized cells, accounts for their undifferentiated appearance. Recent studies also indicate that in some cases, dedifferentiation of apparently mature cells does occur during carcinogenesis
True or false

Anaplastic cells display what?
What are the characteristics of anaplastic cells?

In well­differentiated benign tumors, mitoses are usually rare and are of normal configuration. Between the two extremes lie tumors loosely referred to as moderately well differentiated
True or false

What about the Stroma differentiates benign from malignant

Despite exceptions, the more rapidly growing and the more anaplastic a tumor, the less likely it is to have specialized functional activity.true or false

What is dysplasia,where is it usually encountered
What are the characteristics of dysplastic cells?

In dysplastic stratified squamous epithelium, mitoses are not confined to the basal layers, where they normally occur, but may be seen at all levels and even in surface cells. There is considerable architectural anarchy true or false

What is carcinoma in situ

Where are dysplastic changes commonly found

long­term studies of cigarette smokers show that epithelial dysplasia almost invariably antedates the appear­ ance of cancer, the term dysplasia is not synonymous with cancer; mild to moderate dysplasias that do not involve the entire thickness of the epithelium sometimes regress completely, par- ticularly if inciting causes are removed.
True or false

Cancers of nonendocrine origin may produce so­called ectopic hormones. For example, certain lung carcinomas may produce adrenocor­ ticotropic hormone (ACTH), parathyroid hormone–like hormone, insulin, glucagon, and others.

True or false

A

In practice, the determination of benign versus malignant is made with remarkable accuracy using long­established clinical and anatomic .there are four fundamental features by which benign and malignant tumors can be distinguished: dif- ferentiation and anaplasia, rate of growth, local invasion, and metastasis.

Differentiation and Anaplasia:
Differentiation and anaplasia are characteristics seen only in the parenchymal cells that constitute the transformed elements of neoplasms.

The differentiation of parenchymal tumor cells refers to the extent to which they resemble their normal forebears morphologically and functionally.
• Benign neoplasms are composed of well­differentiated cells that closely resemble their normal counterparts.

A lipoma is made up of mature fat cells laden with cytoplasmic lipid vacuoles, and a chondroma is made up of mature cartilage cells that synthesize their usual cartilaginous matrix—evidence of morphologic and functional differentiation.

• Malignant neoplasms are characterized by a wide range of parenchymal cell differentiation, from sur­ prisingly well differentiated to completely undifferentiated. For example, well­differentiated ade­ nocarcinomas of the thyroid may contain normal­ appearing follicles. Such tumors sometimes may be difficult to distinguish from benign proliferations..

The stroma carrying the blood supply is crucial to the growth of tumors but does not aid in the separation of benign from malignant ones. The amount of stromal connective tissue does deter­ mine, however, the consistency of a neoplasm.

Certain cancers induce a dense, abundant fibrous stroma (des­moplasia), making them hard, so­called scirrhous tumors.
• Malignant neoplasms that are composed of undiffer­entiated cells are said to be anaplastic. Lack of cellular differen­ tiation, or anaplasia, is considered a hallmark of malignancy.
The term anaplasia literally means “back­ ward formation”—implying dedifferentiation, or loss of the structural and functional differentiation of normal cells.

.1.Anaplastic cells display marked pleomorphism (i.e., variation in size and shape) .2.Often the nuclei are extremely hyperchromatic (dark­staining) and large resulting in an increased nuclear­to­cytoplasmic ratio that may approach 1 : 1 instead of the normal 1:4 or 1:6. Giant cells that are considerably larger than their neighbors may be formed and possess either one enormous nucleus or several nuclei.

Anaplastic nuclei are variable and bizarre in size and shape. 3.The chromatin is coarse and clumped, and nucle­oli may be of astounding size.
4.More important, mitoses often are numerous and distinctly atypical; anarchic multi­ ple spindles may produce tripolar or quadripolar mitotic figures .
5.Also, anaplastic cells usually fail to develop recognizable patterns of orientation to one another (i.e., they lose normal polarity). They may grow
in sheets, with total loss of communal structures, such as glands or stratified squamous architecture.

Benign neoplasms and even well­differenti­ ated cancers of endocrine glands frequently elaborate the hormones characteristic of their origin. Well­differentiated squamous cell carcinomas produce keratin.just as well­differentiated hepatocellular carcinomas secrete bile. In other instances, unanticipated functions emerge. Some cancers may elaborate fetal proteins not produced by comparable cells in the adult.

dysplasia, refers to disorderly but non­neoplastic proliferation. Dysplasia is encountered principally in epi­ thelial lesions. It is a loss in the uniformity of individual cells and in their architectural orientation. 1.Dysplastic cells exhibit considerable pleomorphism and 2.often possess hyperchro­ matic nuclei that are abnormally large for the size of the cell. 3.Mitotic figures are more abundant than usual and frequently appear in abnormal locations within the epithe­ lium.

.. When dysplastic changes are marked and involve the entire thickness of the epithelium, the lesion is referred to as carcinoma in situ, a preinvasive stage of cancer .

Although dysplastic changes often are found adjacent to foci of malignant transforma­ tion,

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

Explain rate of growth as a characteristic of the difference between benign and malignant tumors

Give an example of benign tumors that grow rapidly that some cancers

What factors affect the growth of benign tumors

Adeno­ mas of the pituitary gland locked into the sella turcica
have been observed to shrink suddenly. Presumably, they undergo a wave of necrosis as progressive enlargement compresses their blood supply. True or false

The rate of growth of malignant tumors usually correlates inversely with? Explain this
What are the variations in the growth of malignant tumors

Rapidly growing malig­ nant tumors often contain central areas of ischemic necro­ sis, because the tumor blood supply, derived from the host, fails to keep pace with the oxygen needs of the expanding mass of cells.
True or false

Many lines of experimental and clinical evidence document that most if not all cancers take years and sometimes decades to evolve into clinically overt lesions. This is true even of “acute” childhood leukemias, which often initiate during fetal development yet manifest as full­blown cancers years later.
True or false

A

Most benign tumors grow slowly, and most cancers grow much faster, eventually spreading locally and to distant sites (metastasizing) and causing death. There are many exceptions to this generalization, however, and some benign tumors grow more rapidly than some cancers.

For example, the rate of growth of leiomyomas (benign smooth muscle tumors) of the uterus is influenced by the circulat­ ing levels of estrogens. They may increase rapidly in size during pregnancy and then cease growing, becoming largely fibrocalcific, after menopause.

Other influences, such as adequacy of blood supply or pressure constraints, also may affect the growth rate of benign tumors.

it generally is true that most benign tumors increase in size slowly over the span of months to years.

their level of differentiation.

In other words, poorly differentiated tumors tend to grow more rapidly than do well­differentiated tumors.

However, there is wide variation in the rate of growth. Some grow slowly for years and then enter a phase of rapid growth, signifying the emergence of an aggressive subclone of transformed cells. Others grow relatively slowly and steadily; in exceptional instances, growth may come almost to a standstill. Even more exceptionally, some primary tumors (particularly choriocarcinomas) may become totally necrotic, leaving only secondary metastatic implants.

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

The continued growth and maintenance of many tissues that contain short­lived cells, such as the formed elements of the blood and the epithelial cells of the gastrointestinal tract and skin, require what?

tissue stem cells divide asymmetrically to produce two types of daughter cells . What re the types?

Tissue stem cells are rare and exist in a niche created by support cells, which produce paracrine factors that sustain the stem cells. True or false

Cancers are immortal and have limitless proliferative capacity, indicat­ ing that like normal tissues, they also must contain cells with “stemlike” properties. True or false

The cancer stem cell hypothesis posits that, in analogy with normal tissues, only a special subset of cells within tumors has the capacity for self­renewal. True or false

Where do cancer stem cells arise from?

Explain local invasion as a characteristic difference. between benign and malignant
Give an example of how a benign tumor invades locally but doesn’t move and where the capsule formed is derived from

Which is the most reliable feature that distinguishes malignant from benign tumors
SUMMARY
Characteristics of Benign and Malignant Tumors
• Benign and malignant tumors can be distinguished from one another based on the degree of differentiation, rate of growth, local invasiveness, and distant spread.
• Benign tumors resemble the tissue of origin and are well differentiated; malignant tumors are poorly or completely undifferentiated (anaplastic).
• Benign tumors are slow-growing, whereas malignant tumors generally grow faster.
• Benign tumors are well circumscribed and have a capsule; malignant tumors are poorly circumscribed and invade the surrounding normal tissues.
• Benign tumors remain localized to the site of origin, whereas malignant tumors are locally invasive and metas- tasize to distant sites.

True or false

A

a resident population of tissue stem cells that are long­lived and capable of self­renewal.

—those with limited proliferative potential, which undergo terminal differentiation to form particular tissues, and those that retain stem cell potential.

Cancer stem cells could arise from normal tissue stem cells or from more differentiated cells that, as part of the transformation process, acquire the property of self­renewal.

Local Invasion
A benign neoplasm remains localized at its site of origin. It does not have the capacity to infiltrate, invade, or metas­ tasize to distant sites, as do malignant neoplasms.

For example, as adenomas slowly expand, most develop an enclosing fibrous capsule that separates them from the host tissue. This capsule probably is derived from the stroma of the host tissue as the parenchymal cells atrophy under the pressure of the expanding tumor. The stroma of the tumor itself also may contribute to the capsule .Of note, however, not all benign neoplasms are encapsulated. A few benign tumors are neither encapsulated nor discretely defined; such lack of demarcation is particularly likely to be seen in some benign vascular neoplasms of the dermis. These exceptions are pointed out only to emphasize that although encapsulation is the rule in benign tumors, the lack of a capsule does not mean that a tumor is malignant.

Cancers grow by progressive infiltration, invasion, destruc- tion, and penetration of the surrounding tissue .They do not develop well­defined capsules. There are, however, occasional instances in which a slowly growing malignant tumor deceptively appears to be encased by the stroma of the surrounding host tissue, but microscopic examination usually reveals tiny crablike feet penetrating the margin and infiltrating adjacent structures. The infiltrative mode of growth makes it necessary to remove a wide margin of surrounding normal tissue when surgical excision of a malignant tumor is attempted.

Next to the development of metastases, local invasiveness is the most reliable feature that distinguishes malignant from benign tumors.

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

What is metastasis

More than any other attribute, the property of metastasis identifies a neoplasm as malignant. Not all cancers have equivalent ability to metastasize. Give an example

In general, the more anaplastic and the larger the primary neoplasm, the more likely is metastatic spread, but as with most rules, there are exceptions. What are the exceptions?

What are the three pathways of malignant neoplasm dissemination

When does spreading by seed occur? What is this type of dissemination characteristic of?

Lymphatic spread is more typical of what kind of cancers and hematogenous spread is typical of what kind of cancers

Neoplasms of the central nervous system, such as a medulloblastoma or ependymoma, may penetrate the cerebral ventricles and be carried by the cerebrospinal fluid to reimplant on the men­ ingeal surfaces, either within the brain or in the spinal cord. True or false

There are numerous interconnections, however, between the lym­ phatic and vascular systems, so all forms of cancer may disseminate through either or both systems. True or false

The pattern of lymph node involvement depends principally on?

Lung carcinomas arising in the respi­ ratory passages metastasize first to where then to where?

What is a sentinel lymph node?
How is it identified?

Carcinoma of the breast usually arises in the upper outer quadrant and first spreads to the axillary nodes. However, medial breast lesions may drain through the chest wall to the nodes along the internal mammary artery. Thereafter, in both instances, the supraclavicular and infra­ clavicular nodes may be seeded. In some cases, the cancer cells seem to traverse the lymphatic channels within the immediately proximate nodes to be trapped in subsequent lymph nodes, producing so­called skip metastases. The cells may traverse all of the lymph nodes ultimately to reach the vascular compartment by way of the thoracic duct. True or false

although enlargement of nodes near a primary neoplasm should arouse concern for metastatic spread, it does not always imply cancerous involvement. True or false

The necrotic products of the neoplasm and tumor antigens often evoke immunologic responses in the nodes, such as?

What is the favored pathway for sarcomas

What are the frequent sites for hematogenous dissemination

As might be expected, arteries are penetrated less readily than are veins. With venous invasion, the blood­borne cells follow the venous flow draining the site of the neoplasm, with tumor cells often stopping in the first capillary bed they encounter.
skeletal muscles, although rich in capillaries, are rarely the site of secondary deposits. True or false

prostatic carcinoma preferentially spreads to bone, bronchogenic carcinomas tend to involve the adre­ nals and the brain, and neuroblastomas spread to the liver and bones. Cancers arising near the vertebral column often em­ bolize through the paravertebral plexus;
Certain carcinomas have a propensity to grow within veins. Renal cell carcinoma often invades the renal vein to grow in a snakelike fashion up the inferior vena cava, sometimes reaching the right side of the heart.
True or false

A

Metastasis
Metastases are secondary implants of a tumor that are discontinuous with the primary tumor and located in remote tissues .

At one extreme are basal cell carcinomas of the skin and most primary tumors of the central nervous system, which are highly invasive locally but rarely metastasize. At the other extreme are osteogenic (bone) sarcomas, which usually have metastasized to the lungs at the time of initial discovery.

Extremely small cancers have been known to metastasize; conversely, some large and ominous­looking lesions may not.

Malignant neoplasms disseminate by one of three path­ ways: (1) seeding within body cavities, (2) lymphatic spread, or (3) hematogenous spread.

Spread by seeding occurs when neoplasms invade a natural body cavity. This mode of dissemination is particularly characteristic of cancers of the ovary, which often cover the peritoneal sur­ faces widely. The implants literally may glaze all peritoneal surfaces and yet not invade the underlying tissues.

Lymphatic spread is more typical of carcinomas, whereas hematogenous spread is favored by sarcomas.

the site of the primary neoplasm and the natural pathways of local lymphatic drainage

. the regional bronchial lymph nodes and then to the tracheobronchial and hilar nodes.

A “sentinel lymph node” is the first regional lymph node that receives lymph flow from a primary tumor. It can be identified by injection of blue dyes or radiolabeled tracers near the primary tumor. Biopsy of sentinel lymph nodes allows determination of the extent of spread of tumor and can be used to plan treatment.

hyperplasia of the follicles (lymphadenitis) and proliferation of macro­ phages in the subcapsular sinuses (sinus histiocytosis). Thus, histopathologic verification of tumor within an enlarged lymph node is required.

Hematogenous spread is the favored pathway for sarco­ mas, but carcinomas use it as well.

Since all portal area drainage flows to the liver, and all caval blood flows to the lungs, the liver and lungs are the most frequently involved secondary sites in hematogenous dissemina- tion.

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

What is carcinogenesis

Many cancers arise from non­neoplastic precursor lesions, true or false

Name three phenotypic attributes of malignant neoplastic cells

What is tumor progression

At the molecular level, tumor progression and associated heterogeneity are most likely to result from multiple mutations that accumulate inde­ pendently in different cells, generating subclones with dif­ ferent characteristics such as ?

Some of the mutations may be lethal; others may spur cell growth by affecting proto­oncogenes or cancer suppressor genes. Thus even though most malignant tumors are monoclonal in origin, by the time they become clinically evident their con- stituent cells may be extremely heterogeneous. True or false

During progression, tumor cells are subjected to ?
And give an example

A growing tumor, therefore, tends to be enriched for subclones that “beat the odds” and are adept at survival, growth, invasion, and metastasis.
True or false

What are the two properties of cancers

A

PROCESS
Carcinogenesis is a multistep process resulting from the accumulation of multiple genetic alterations that collec­ tively give rise to the transformed phenotype.

malignant neoplasms have several phenotypic attributes, such as excessive growth, local inva­siveness, and the ability to form distant metastases.

Furthermore, it is well established that over a period of time, many tumors become more aggressive and acquire greater malignant potential. This phenomenon is referred to as tumor progression and is not represented simply by an increase in tumor size.

ability to invade, rate of growth, metastatic ability, karyotype, hormonal responsiveness, and susceptibility to antineoplastic drugs.

immune and nonimmune selection pressures.

For example, cells that are highly antigenic are destroyed by host defenses, whereas those with reduced growth factor requirements are positively selected.

Thus, genetic evolution and selection can explain two of the most pernicious properties of cancers: the tendency for cancers to become (1) more aggressive and (2) less responsive to therapy over time.

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

What are the hallmarks of cancer

ABL which is a cancer associated gene is affected where? What cancer associated gene is commonly mutated

A

Some, such as TP53, are commonly mutated; others, such as ABL, are affected only in certain leukemias.

the so­called hallmarks of cancer, which together dictate the malignant phenotype. Six of these are :
• Self­sufficiency in growth signals
• Insensitivity to growth inhibitory signals
• Evasion of cell death
• Limitless replicative potential
• Development of sustained angiogenesis
• Ability to invade and metastasize
To this list may be added two “emerging” hallmarks of cancer, reprogramming of energy metabolism and evasion of the immune system, and two enabling characteristics, genomic instability and tumor­promoting inflammation.

. Of note, by convention, gene symbols are italicized but their protein products are not (e.g., RB gene and Rb protein, TP53 and p53, MYC and MYC).

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

Explain self sufficiency in growth signals

Under normal conditions,cell proliferation occurs in five steps name them

The mechanisms that endow cancer cells with the ability to proliferate can be grouped according to their role in the growth factor–induced signal transduction cascade and cell cycle regulation. True or false

What is paracrine action

Normally, cells that produce the growth factor do not express the cognate receptor. This specificity prevents the formation of positive feedback loops within the same cell.
True or false

Name two ways cancer cells acquire self sufficiency
Give an example

What kind of proteins deliver continuous mitogenic signals to the cells

Name two ways cancer cells affect growth factor or get growth autonomy

How do cancer cells affect downstream mechanism to get growth autonomy

Name the two important signal transduction molecules or oncoproteins

RAS is the most commonly mutated proto­ oncogene in human tumors. RAS is a member of a family of small G proteins that bind guanosine nucleotides (guanosine triphosphate [GTP] and guanosine diphosphate [GDP]), similar to the larger trimolecular G proteins.
True or false

How does RAS protein work

What prevents Uncontrolled RAS activation

The activated RAS stimulates downstream regulators of proliferation by two distinct pathways that converge on the nucleus and flood it with signals for cell prolifera­ tion. True or false

that muta­ tional activation of these “messengers” to the nucleus can mimic the growth promoting effects of activated RAS. True or false

How is the RAS activated

A

Cancer cells use a number of strategies to drive their pro­ liferation and become insensitive to normal growth regula­ tors. Under physiologic conditions, cell proliferation can be readily resolved into the following steps:

  1. The binding of a growth factor to its specific receptor on the cell membrane
  2. Transient and limited activation of the growth factor receptor, which in turn activates several signal­ transducing proteins on the inner leaflet of the plasma membrane
  3. Transmission of the transduced signal across the cytosol to the nucleus by second messengers or a cascade of signal transduction molecules
  4. Induction and activation of nuclear regulatory factors that initiate and regulate DNA transcription
  5. Entry and progression of the cell into the cell cycle, resulting ultimately in cell division

Growth Factors
All normal cells require stimulation by growth factors to undergo proliferation. Most soluble growth factors are made by one cell type and act on a neighboring cell to stimulate proliferation (paracrine action).

  • Many cancer cells acquire growth self­sufficiency by acquiring the ability to synthesize the same growth factors to which they are responsive. For example, many glioblastomas secrete platelet­derived growth factor (PDGF) and express the PDGF receptor, and many sar­ comas make both transforming growth factor­α (TGF­α) and its receptor. Similar autocrine loops are fairly common in many types of cancer.
  • Another mechanism by which cancer cells acquire growth self­sufficiency is by interaction with stroma. In some cases, tumor cells send signals to activate normal cells in the supporting stroma, which in turn produce growth factors that promote tumor growth.

Growth Factor Receptors and Non-Receptor
Tyrosine Kinases
1.Mutant receptor proteins deliver continuous mitogenic signals to cells, even in the absence of the growth factor in the environment.
2.overexpression of growth factor receptors, which can render cancer cells hyperresponsive to levels of the growth factor that would not normally trigger proliferation.

By causing mutations in genes that encode various components of the signaling pathways down­ stream of growth factor receptors. These signaling proteins couple growth factor receptors to their nuclear targets. They receive signals from activated growth factor receptors and transmit them to the nucleus through activation of signal transduction molecules. (Or either through second messengers or through a cascade of phosphorylation )

Two important members in this category are RAS and ABL.

Normal RAS proteins flip back and forth between an excited signal­transmitting state and a quiescent state. RAS proteins are inactive when bound to GDP; stimulation of cells by growth factors such as EGF and PDGF leads to exchange of GDP for GTP and sub­ sequent conformational changes that generate active RAS .

This excited signal­emitting state is short­lived, however, because the intrinsic guanosine triphosphatase (GTPase) activity of RAS hydrolyzes GTP to GDP, releasing a phosphate group and returning the protein to its quiescent GDP­bound state. The GTPase activity of activated RAS protein is magnified dramatically by a family of GTPase­activating proteins (GAPs), which act as molecular brakes that prevent uncontrolled RAS activation by favoring hydrolysis of GTP to GDP.

The RAS protein most commonly is activated by point mutations in amino acid residues that are either within the GTP­binding pocket or in the enzymatic region essential for GTP hydrolysis. Both kinds of mutations interfere with GTP hydrolysis, which is essential to inactivate RAS. RAS is thus trapped in its activated, GTP­bound form, and the cell is forced into a continuously proliferating state.

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

Explain downstream signal transduction proteins

A

Downstream Signal-Transducing Proteins
A relatively common mechanism by which cancer cells acquire growth autonomy is mutations in genes that encode various components of the signaling pathways down­ stream of growth factor receptors. These signaling proteins couple growth factor receptors to their nuclear targets. They receive signals from activated growth factor receptors and transmit them to the nucleus, either through second messengers or through a cascade of phosphorylation and activation of signal transduction molecules. Two important members in this category are RAS and ABL. Each of these is discussed briefly next.
RAS Protein. RAS is the most commonly mutated proto­ oncogene in human tumors. Indeed, approximately 30% of all human tumors contain mutated versions of the RAS gene, and the frequency is even higher in some specific cancers (e.g., colon and pancreatic adenocarcinomas).
• RAS is a member of a family of small G proteins that bind guanosine nucleotides (guanosine triphosphate [GTP] and guanosine diphosphate [GDP]), similar to the larger trimolecular G proteins.
• Normal RAS proteins flip back and forth between an excited signal­transmitting state and a quiescent state. RAS proteins are inactive when bound to GDP; stimulation of cells by growth factors such as EGF and PDGF leads to exchange of GDP for GTP and sub­ sequent conformational changes that generate active RAS (Fig. 5–19). This excited signal­emitting state is short­lived, however, because the intrinsic guanosine triphosphatase (GTPase) activity of RAS hydrolyzes GTP to GDP, releasing a phosphate group and returning the protein to its quiescent GDP­bound state. The GTPase activity of activated RAS protein is magnified dramatically by a family of GTPase­activating proteins (GAPs), which act as molecular brakes that prevent uncontrolled RAS activation by favoring hydrolysis of GTP to GDP.

• The activated RAS stimulates downstream regulators of proliferation by two distinct pathways that converge on the nucleus and flood it with signals for cell prolifera­ tion. While details of the signaling cascades (some of which are illustrated in Fig. 5–19) downstream of RAS are not discussed here, an important point is that muta­ tional activation of these “messengers” to the nucleus can mimic the growth promoting effects of activated RAS. For example, BRAF, which lies in the so­called RAF/ERK/MAP kinase pathway, is mutated in more than 60% of melanomas. Mutations of PI3 kinase in the PI3K/AKT pathway also occur with high frequency in some tumor types. Indeed, it appears that activating mutations of RAS as well as its downstream signaling molecules are very common in a wide variety of tumors.
The RAS protein most commonly is activated by point mutations in amino acid residues that are either within the GTP­binding pocket or in the enzymatic region essential for GTP hydrolysis. Both kinds of mutations interfere with GTP hydrolysis, which is essential to inactivate RAS. RAS is thus trapped in its activated, GTP­bound form, and the cell is forced into a continuously proliferating state.

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

Explain nuclear transcription factors and cyclins and cyclin dependent kinases

A

Nuclear Transcription Factors
Ultimately, all signal transduction pathways enter the nucleus and have an impact on a large bank of responder genes that orchestrate the cell’s orderly advance through the mitotic cycle. Indeed, the ultimate consequence of signaling through oncoproteins such as RAS or ABL is inappropriate and continuous stimulation of nuclear tran­ scription factors that drive the expression of growth­ promoting genes. Growth autonomy may thus be a consequence of mutations affecting genes that regulate transcription of DNA. A host of oncoproteins, including products of the MYC, MYB, JUN, FOS, and REL oncogenes, function as transcription factors that regulate the
expression of growth­promoting genes, such as cyclins. Of these, the MYC gene is involved most commonly in human tumors.
The MYC protein can either activate or repress the transcription of other genes. Those activated by MYC include several growth­promoting genes, including cyclin­ dependent kinases (CDKs), whose products drive cells into the cell cycle (discussed next). Genes repressed by MYC include the CDK inhibitors (CDKIs). Thus, dysregulation of MYC promotes tumorigenesis by increasing expression of genes that promote progression through the cell cycle and repressing genes that slow or prevent progression through the cell cycle. MYC also is a key regulator of intermediate metabolism, upregulating genes that promote aerobic gly­ colysis (the so­called Warburg effect, described later) and the increased utilization of glutamine, two metabolic changes that are hallmarks of cancer cells. Dysregulation of the MYC gene resulting from a t(8;14) translocation occurs in Burkitt lymphoma, a B cell tumor. MYC also is amplified in breast, colon, lung, and many other cancers; the related NMYC and LMYC genes are amplified in neuroblastomas and small cell cancers of lung.
Cyclins and Cyclin-Dependent Kinases
The ultimate outcome of all growth­promoting stimuli is the entry of quiescent cells into the cell cycle. Cancers may become autonomous if the genes that drive the cell cycle become dysregulated by mutations or amplification. Before further consideration of this aspect of carcinogene­ sis, a brief review of the normal cell cycle is warranted

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

What happens in a normal cell cycle

What are the alterations in cell cycle proteins in cancer

A

Normal Cell Cycle
Cell proliferation is a tightly controlled process that involves a large number of molecules and interrelated pathways. The replication of cells is stimulated by growth factors or by signaling from ECM components through integrins. To achieve DNA replication and division, the cell goes through a tightly controlled sequence of events known as the cell cycle. The cell cycle consists of G1 (presynthetic), S (DNA synthesis), G2 (premitotic), and M (mitotic) phases. Quiescent cells that have not entered the cell cycle are in the G0 state. Each cell cycle phase is dependent on the proper activation and completion of the previous ones and the cycle stops at a place at which an essential gene func­ tion is deficient. Because of its central role in maintaining tissue homeostasis and regulating physiologic growth pro­ cesses such as regeneration and repair, the cell cycle has multiple checkpoints, particularly during emergence from G0 into G1 and the transition from G1 to S phase.
Cells can enter G1 either from G0 (quiescent cells) or after completing mitosis (continuously replicating cells). Quies­ cent cells must first go through the transition from G0 to G1, the first decision step, which functions as a gateway to the cell cycle. Cells in G1 progress through the cell cycle and reach a critical stage at the G1­S transition, known as a restriction point, a rate­limiting step for replication. On passing this restriction point, normal cells become irrevers­ ibly committed to DNA replication. The cell cycle is tightly controlled by activators and inhibitors.
• Progression through the cell cycle, particularly at the G1­S transition, is regulated by proteins called cyclins, so

called because of the cyclic nature of their production and degradation, and associated enzymes, the cyclin- dependent kinases (CDKs). CDKs acquire catalytic activity by binding to and forming complexes with the cyclins. The orderly progression of cells through the various phases of the cell cycle is orchestrated by CDKs, which are activated by binding to the cyclins.
• TheCDK–cyclincomplexesphosphorylatecrucialtarget proteins that drive the cell through the cell cycle. On completion of this task, cyclin levels decline rapidly. More than 15 cyclins have been identified; cyclins D, E, A, and B appear sequentially during the cell cycle and bind to one or more CDKs. The cell cycle may thus be seen as a relay race in which each leg is regulated by a distinct set of cyclins: As one set of cyclins leaves the track, the next set takes over (Fig. 5–20). Activated CDKs in these complexes drive the cell cycle by phosphorylat­ ing proteins that regulate cell cycle transitions. One such protein is the retinoblastoma protein (Rb), discussed later.
• The activity of CDK–cyclin complexes is regulated by CDK inhibitors (CDKIs), which enforce cell cycle
checkpoints. Embedded in the cell cycle are surveillance mechanisms that are geared to sensing damage to DNA and chromosomes. These quality control checks are called checkpoints; they ensure that cells with damaged DNA or chromosomes do not complete replication. The G1­S checkpoint monitors the integrity of DNA before DNA replication, whereas the G2­M checkpoint checks DNA after replication and monitors whether the cell can safely enter mitosis. When cells sense DNA damage, checkpoint activation delays the cell cycle and triggers DNA repair mechanisms. If DNA damage is too severe to be repaired, the cells are eliminated by apoptosis, or enter a nonreplicative state called senescence, primarily through p53­dependent mechanisms, discussed later on. Mutations in genes regulating these checkpoints allow cells with damaged DNA to divide, producing daughter cells carrying mutations.
• There are several families of CDKIs. One family, composed of three proteins called p21 (CDKN1A), p27 (CDKN1B), and p57 (CDKN1C), inhibits the CDKs broadly, whereas the other family of CDKIs has selective effects on cyclin CDK4 and cyclin CDK6. The four members of this family—p15 (CDKN2B), p16 (CDKN2A), p18 (CDKN2C), and p19 (CDKN2D)—are sometimes called INK4 (A to D) proteins.

Alterations in Cell Cycle Control Proteins in Cancer Cells
With this background it is easy to appreciate that muta­ tions that dysregulate the activity of cyclins and CDKs would favor cell proliferation. Indeed, all cancers appear to have genetic lesions that disable the G1­S checkpoint, causing cells to continually reenter the S phase. For unclear reasons, particular lesions vary widely in frequency across tumor types.
• Mishaps increasing the expression of cyclin D or CDK4 seem to be a common event in neoplastic transforma­ tion. The cyclin D genes are overexpressed in many cancers, including those affecting the breast, esophagus, liver, and a subset of lymphomas and plasma cell tumors. Amplification of the CDK4 gene occurs in melanomas, sarcomas, and glioblastomas. Mutations affecting cyclins B and E and other CDKs also occur, but they are much less frequent than those affecting cyclin CDK4.
• The CDKIs frequently are disabled by mutation or gene silencing in many human malignancies. Germline muta­ tions of CDKN2A are present in 25% of melanoma­prone kindreds. Somatically acquired deletion or inactivation of CDKN2A is seen in 75% of pancreatic carcinomas, 40% to 70% of glioblastomas, 50% of esophageal cancers, and 20% of non–small cell lung carcinomas, soft tissue sar­ comas, and bladder cancers.
A final consideration of importance in a discussion of growth­promoting signals is that the increased production of oncoproteins does not by itself lead to sustained prolif­ eration of cancer cells. There are two built­in mechanisms, cell senescence and apoptosis, that oppose oncogene­ mediated cell growth. As discussed later, genes that regu­ late these two braking mechanisms must be disabled to allow unopposed action of oncogenes.

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

Oncogenes That Promote Unregulated Proliferation (Self-Sufficiency in Growth Signals)
Proto-oncogenes: normal cellular genes whose products promote cell proliferation
Oncogenes: mutant or overexpressed versions of proto- oncogenes that function autonomously without a require- ment for normal growth-promoting signals
Oncoproteins promote uncontrolled cell proliferation by several mechanisms:
• Stimulus-independent expression of growth factor and
its receptor, setting up an autocrine loop of cell proliferation
 PDGF–PDGF receptor in brain tumors
• Mutations in genes encoding growth factor receptors or tyrosine kinases leading to constitutive signaling
 EGF receptor family members, including HER2/NEU
(breast, lung, and other tumors)  Fusion of ABL tyrosine kinase with BCR protein in certain leukemias generates a hybrid protein with con- stitutive kinase activity.
• Mutations in genes encoding signaling molecules
 RAS commonly is mutated in human cancers and nor-
mally flips between resting GDP-bound state and active GTP-bound state; mutations block hydrolysis of GTP to GDP, leading to unchecked signaling.
• Overproduction or unregulated activity of transcription factors
 Translocation of MYC in some lymphomas leads to over- expression and unregulated expression of its target genes controlling cell cycling and survival.
• Mutations that activate cyclin genes or inactivate negative regulators of cyclins and cyclin-dependent kinases
 Complexes of cyclins with CDKs drive the cell cycle by
phosphorylating various substrates. CDKs are con- trolled by inhibitors; mutations in genes encoding cyclins, CDKs, and CDK inhibitors result in uncon- trolled cell cycle progression. Such mutations are found in a wide variety of cancers including melanomas, brain, lung, and pancreatic cancer.

True or false

A

True

19
Q

Whereas oncogenes encode proteins that promote cell growth, the products of tumor suppressor genes apply brakes to cell proliferation. Disruption of such genes renders cells refrac­ tory to growth inhibition and mimics the growth­promoting effects of oncogenes.
True or false

Insensitivity to Growth Inhibitory Signals
• Tumor suppressor genes encode proteins that inhibit cel- lular proliferation by regulating the cell cycle. Unlike onco- genes, both copies of the gene must be dysfunctional for tumor development to occur.
• In cases with familial predisposition for development of tumors, affected persons inherit one defective (nonfunc- tional) copy of a tumor suppressor gene and lose the second one through somatic mutation. In sporadic cases, both copies are lost through somatic mutations.

True or false

RB Gene: Governor of the Cell Cycle
• Rb exerts antiproliferative effects by controlling the G1- to-S transition of the cell cycle. In its active form, Rb is hypophosphorylated and binds to E2F transcription factor. This interaction prevents transcription of genes like cyclin E that are needed for DNA replication, and so the cells are arrested in G1.
• Growth factor signaling leads to cyclin D expression, acti- vation of the cyclin D–CDK4/6 complexes, inactivation of Rb by phosphorylation, and thus release of E2F.
• Loss of cell cycle control is fundamental to malignant transformation. Almost all cancers have a disabled G1 checkpoint due to mutation of either RB or genes that
affect Rb function, such as cyclin D, CDK4, and CDKIs.
• Many oncogenic DNA viruses, like HPV, encode proteins (e.g., E7) that bind to Rb and render it nonfunctional.

True or false

A

True

True

True

20
Q

What is the function of TP53 gene

A

SUMMARY
TP53 Gene: Guardian of the Genome
• The p53 protein is the central monitor of stress in the cell and can be activated by anoxia, inappropriate onco- gene signaling, or DNA damage. Activated p53 controls the expression and activity of genes involved in cell cycle arrest, DNA repair, cellular senescence, and apoptosis.
• DNA damage leads to activation of p53 by phosphoryla- tion. Activated p53 drives transcription of CDKN1A (p21), which prevents Rb phosphorylation, thereby causing a G1-S block in the cell cycle. This pause allows the cells to repair DNA damage.
• If DNA damage cannot be repaired, p53 induces cellular senescence or apoptosis.
• Of human tumors, 70% demonstrate biallelic loss of TP53. Patients with the rare Li-Fraumeni syndrome inherit one

defective copy in the germ line and lose the second one in somatic tissues; such persons develop a variety of tumors.
• As with Rb, p53 can be incapacitated by binding to pro- teins encoded by oncogenic DNA viruses such as HPV.

21
Q

Whata re the Transforming Growth Factor-β and APC–β-Catenin Pathways

A

Pathways
• TGF-β inhibits proliferation of many cell types by activa- tion of growth-inhibiting genes such as CDKIs and sup- pression of growth-promoting genes such as MYC and those encoding cyclins.

  • TGF-β function is compromised in many tumors by muta- tions in its receptors (colon, stomach, endometrium) or by mutational inactivation of SMAD genes that transduce TGF-β signaling (pancreas).
  • E-cadherin maintains contact inhibition, which is lost in malignant cells.
  • APC gene exerts antiproliferative actions by regulating the destruction of the cytoplasmic protein β-catenin. With a loss of APC, β-catenin is not destroyed, and it translocates to the nucleus, where it acts as a growth-promoting trans- cription factor.
  • In familial adenomatous polyposis syndrome, inheritance of a germ line mutation in the APC gene and sporadic loss of the sole normal allele causes the development of hun- dreds of colonic polyps at a young age. Inevitably, one or more of these polyps evolves into a colonic cancer. Somatic loss of both alleles of the APC gene is seen in approximately 70% of sporadic colon cancers.
22
Q

cells. It is now well established that accumulation of neoplastic cells may result not only from activation of growth- promoting oncogenes or inactivation of growth-suppressing tumor suppressor genes but also from mutations in the genes that regulate apoptosis.
True or false

Evasion of Apoptosis
• Apoptosis can be initiated through extrinsic or intrinsic pathways.
• Both pathways result in the activation of a proteolytic cascade of caspases that destroys the cell.
• Mitochondrial outer membrane permeabilization is regu- lated by the balance between pro-apoptotic (e.g., BAX, BAK) and anti-apoptotic molecules (BCL2, BCL-XL). BH- 3–only molecules activate apoptosis by tilting the balance in favor of the pro-apoptotic molecules.
• In 85% of follicular B cell lymphomas, the anti-apoptotic gene BCL2 is activated by the t(14;18) translocation.
• Stress may also induce cells to consume their components in a process called autophagy. Cancer cells may accumu- late mutations to avoid autophagy, or may corrupt the process to provide parts for continued growth.

True or false

A

True

23
Q

Limitless Replicative Potential
• In normal cells, which lack expression of telomerase, the shortened telomeres generated by cell division eventually activate cell cycle checkpoints, leading to senescence and placing a limit on the number of divisions a cell may undergo.
• In cells that have disabled checkpoints, DNA repair pathways are inappropriately activated by shortened telomeres, leading to massive chromosomal instability and mitotic crisis.
• Tumor cells reactivate telomerase, thus staving off mitotic catastrophe and achieving immortality.

True or false

A

True

24
Q

Explain development of sustained angiogenesis as a hallmark of cancer

A

Development of Sustained Angiogenesis
• Vascularization of tumors is essential for their growth and is controlled by the balance between angiogenic and anti- angiogenic factors that are produced by tumor and stromal cells.
• Hypoxia triggers angiogenesis through the actions of HIF- 1α on the transcription of the pro-angiogenic factor VEGF. Because of its ability to degrade HIF-1α and thereby prevent angiogenesis, VHL acts as a tumor suppressor. Inheritance of germ line mutations of VHL causes VHL syndrome, characterized by the development of a variety of tumors.
• Many other factors regulate angiogenesis; for example, p53 induces synthesis of the angiogenesis inhibitor TSP-1.

25
Q

Explain ability to invade and metastise as a hallmark of cancer

A

SUMMARY
Invasion and Metastasis
• Ability to invade tissues, a hallmark of malignancy, occurs in four steps: loosening of cell–cell contacts, degradation of ECM, attachment to novel ECM components, and migration of tumor cells.
• Cell–cell contacts are lost by the inactivation of E-cadherin through a variety of pathways.
• Basement membrane and interstitial matrix degradation is mediated by proteolytic enzymes secreted by tumor cells and stromal cells, such as MMPs and cathepsins.
• Proteolytic enzymes also release growth factors seques- tered in the ECM and generate chemotactic and angio- genic fragments from cleavage of ECM glycoproteins.
• The metastatic site of many tumors can be predicted by the location of the primary tumor. Many tumors arrest in the first capillary bed they encounter (lung and liver, most commonly).
• Some tumors show organ tropism, probably due to activa- tion of adhesion or chemokine receptors whose ligands are expressed by endothelial cells at the metastatic site.

26
Q

Explain reprogramming energy metabolism as a hallmark of cancer and state the two
Types

Explain Tumor-Promoting Inflammation as Enabler of Malignancy

A

Reprogramming of energy metabolism is so common to tumors that it is now considered a hallmark of cancer. Even in the presence of ample oxygen, cancer cells shift their glucose metabolism away from the oxygen­hungry but effi­ cient mitochondria to glycolysis. This phenomenon, called the Warburg effect and also known as aerobic glycolysis,

Genomic Instability as Enabler of Malignancy
• Persons with inherited mutations of genes involved in DNA repair systems are at greatly increased risk for the development of cancer.
• Patients with HNPCC syndrome have defects in the mis- match repair system, leading to development of carcino- mas of the colon. These patients’ genomes show MSI, characterized by changes in length of short tandem repeating sequences throughout the genome.
• Patients with xeroderma pigmentosum have a defect in the nucleotide excision repair pathway and are at increased risk for the development of cancers of the skin exposed to UV light, because of an inability to repair pyrimidine dimers.
• Syndromes involving defects in the homologous recombi- nation DNA repair system constitute a group of disorders—Bloom syndrome, ataxia-telangiectasia, and Fanconi anemia—that are characterized by hypersensitiv- ity to DNA-damaging agents, such as ionizing radiation. BRC A1 and BRC A2, which are mutated in familial breast cancers, are involved in DNA repair.
• Mutations incurred in lymphoid cells expressing gene products that induce genomic instability (RAG1, RAG2, AID) are important causes of lymphoid neoplasms.

Accumulating evidence suggests that inflammation, often thought of as a protective response against tumors, can paradoxically also enable malignancy. This occurs in two different settings:
• Persistent chronic inflammation in response to microbial infections or as part of an autoimmune reaction. This is exemplified by the increased risk of cancer in patients affected by a variety of chronic inflammatory diseases of the gastrointestinal tract. These include Barrett esoph­ agus, ulcerative colitis, H. pylori gastritis, hepatitis B and C, and chronic pancreatitis. As with any cause of chronic tissue injury, there is a compensatory proliferation of

 198 CHAPTER 5 Neoplasia cells in an attempt to repair the damage. This regenera­ tive process is aided and abetted by a plethora of growth factors, cytokines, chemokines, and other bioactive sub­ stances produced by activated immune cells collected at the site. Persistent cell replication and reduced apoptosis under these conditions place the cells at risk of acquiring mutations in one or more of the genes involved in car­ cinogenesis. In addition, inflammatory cells such as neu­ trophils can contribute to carcinogenesis by secretion of reactive oxygen species, which in turn can inflict addi­ tional DNA damage in rapidly dividing cells. • When inflammation occurs in response to tumors. Patholo­ gists have known for quite some time that many tumors are infiltrated by leukocytes. The degree of inflamma­ tion varies, but virtually every tumor contains cells of the adaptive and innate components of the immune system. The conventional wisdom has been that the inflammatory reaction is protective since it represents an attempt by the host to destroy the tumor. Indeed, that may well be the purpose of the inflammatory reaction, but these cells can exert tumor­promoting activity by producing growth factors and inflicting additional DNA damage as described above. Whatever the precise mechanism, the link between inflam­ mation and cancer has practical implications. For instance, expression of the enzyme cyclooxygenase­2 (COX­2), which brings about the conversion of arachidonic acid into prostaglandins (Chapter 2), is induced by inflammatory stimuli and is increased in colon cancers and other tumors. The use of COX­2 inhibitors for cancer prevention and treatment is an active area of research. Important clinical considerations emerge from the prin­ ciples presented in the foregoing discussion of the hall­ marks of cancer: These hallmarks provide a road map for the development of new therapeutic agents for the treat­ ment of cancer
27
Q

Multistep Carcinogenesis and Cancer Progression
As described earlier, the acquisition of several fundamental abnormalities is a prerequisite to development of malig­ nancy. It follows, then, that each cancer must result from accumulation of multiple mutations. A dramatic example of incremental acquisition of the malignant phenotype is doc­ umented by the study of colon carcinoma. These lesions are believed to evolve through a series of morphologically identifiable stages: colon epithelial hyperplasia followed by formation of adenomas that progressively enlarge and ultimately undergo malignant transformation (Chapter 14). The proposed molecular correlates of this adenoma­ carcinoma sequence are illustrated in Figure 5–30. Accord­ ing to this scheme, inactivation of the APC tumor suppressor gene occurs first, followed by activation of RAS and, ulti­ mately, loss of a tumor suppressor gene on 18q and loss of TP53. The precise temporal sequence of mutations may be different in different tumors.

True or false

A

True

28
Q

(Aetiology of cancer (carcinogenic agents)Three classes of carcinogenic agents have been identified: name them

How do chemical agents cause cancer

And give six examples of chem agents that chase cancer

A

chemicals, (2) radiant energy, and (3) microbial agents.

Direct-Acting Agents
Direct­acting agents require no metabolic conversion to become carcinogenic. They are in general weak carcino­ gens but are important because some of them are cancer chemotherapy drugs (e.g., alkylating agents) used in regi­ mens that may cure certain types of cancer (e.g., Hodgkin lymphoma), only to evoke a subsequent, second form of cancer, usually leukemia. This situation is even more tragic when the initial use of such agents has been for non­ neoplastic disorders, such as rheumatoid arthritis or Wegener granulomatosis. The associated risk of induced cancer is low, but its existence dictates judicious use of such agents.
Indirect-Acting Agents
The designation indirect-acting refers to chemicals that require metabolic conversion to an ultimate carcinogen. Some of the most potent indirect chemical carcinogens are polycyclic hydrocarbons, present in fossil fuels. For example, benzo[a]pyrene and other carcinogens are formed in the high­temperature combustion of tobacco in cigarette smoking. These products are implicated in the causation of lung cancer in cigarette smokers. Polycyclic hydrocarbons also may be produced from animal fats during the process of broiling meats and are present in smoked meats and fish.

Chemical Carcinogens
• Chemical carcinogens have highly reactive electrophile groups that directly damage DNA, leading to mutations and eventually cancer.
• Direct-acting agents do not require metabolic conversion to become carcinogenic, while indirect-acting agents are not active until converted to an ultimate carcinogen by endogenous metabolic pathways. Hence, polymorphisms of endogenous enzymes such as cytochrome P-450 may influence carcinogenesis.
• After exposure of a cell to a mutagen or an initiator, tumorigenesis can be enhanced by exposure to promot- ers, which stimulate proliferation of the mutated cells.
• Examples of human carcinogens are direct-acting agents (e.g., alkylating agents used for chemotherapy), indirect- acting agents (e.g., benzopyrene, azo dyes, aflatoxin), and promoters or agents that cause hyperplasia of endome- trium or regenerative activity in the liver.

Direct-Acting Carcinogens
Alkylating Agents
β-Propiolactone
Dimethyl sulfate
Diepoxybutane
Anticancer drugs (cyclophosphamide, chlorambucil, nitrosoureas, and
others)
Acylating Agents
1-Acetyl-imidazole Dimethylcarbamyl chloride
Procarcinogens That Require Metabolic Activation
Polycyclic and Heterocyclic Aromatic Hydrocarbons
Benz(a)anthracene Benzo(a)pyrene Dibenz(a,h)anthracene 3-Methylcholanthrene
7, 12-Dimethylbenz(a)anthracene
Aromatic Amines, Amides, Azo Dyes
2-Naphthylamine (β-naphthylamine) Benzidine
2-Acetylaminofluorene Dimethylaminoazobenzene (butter yellow)
Natural Plant and Microbial Products
Aflatoxin B1 Griseofulvin Cycasin Safrole Betel nuts
Others
Nitrosamine and amides
Vinyl chloride, nickel, chromium Insecticides, fungicides Polychlorinated biphenyls

29
Q

How does radiation cause cancer

How do some microbiological agents cause cancer

A

Radiation Carcinogenesis
• Ionizing radiation causes chromosome breakage, translo- cations, and, less frequently, point mutations, leading to genetic damage and carcinogenesis.
• UV rays induce the formation of pyrimidine dimers within DNA, leading to mutations. Therefore, UV rays can give rise to squamous cell carcinomas and melanomas of the skin.

Oncogenic RNA Viruses
• HTLV-1 causes a T cell leukemia that is endemic in Japan and the Caribbean.
• The HTLV-1 genome encodes a viral TAX protein, which turns on genes for cytokines and their receptors in infected T cells. This sets up autocrine and paracrine sig- naling loops that stimulate T cell proliferation. Although this proliferation initially is polyclonal, the proliferating T cells are at increased risk for secondary mutations that lead to the outgrowth of a monoclonal leukemia.

Hepatitis B and Hepatitis C Viruses
• Between 70% and 85% of hepatocellular carcinomas worldwide are due to infection with HBV or HCV.
• The oncogenic effects of HBV and HCV are multifactorial, but the dominant effect seems to be immunologically mediated chronic inflammation, with hepatocellular injury, stimulation of hepatocyte proliferation, and production of reactive oxygen species that can damage DNA.
• The HBx protein of HBV and the HCV core protein can activate a variety of signal transduction pathways that also may contribute to carcinogenesis

Helicobacter pylori
• H. pylori infection has been implicated in both gastric adenocarcinoma and MALT lymphoma.
• The mechanism of H. pylori–induced gastric cancers is multifactorial, including immunologically mediated chronic inflammation, stimulation of gastric cell proliferation, and production of reactive oxygen species that damage DNA. H. pylori pathogenicity genes, such as CagA, also may con- tribute by stimulating growth factor pathways.
• It is thought that H. pylori infection leads to polyclonal B cell proliferations and that eventually a monoclonal B cell tumor (MALT lymphoma) emerges as a result of accumu- lation of mutations.

30
Q

Explain the host defense against tumors or tumor immunity

A

Tumor Antigens
Antigens that elicit an immune response have been dem­ onstrated in many experimentally induced tumors and in some human cancers. Initially, they were broadly classified into two categories based on their patterns of expression: tumor-specific antigens, which are present only on tumor cells and not on any normal cells, and tumor-associated anti- gens, which are present on tumor cells and also on some normal cells. This classification, however, is imperfect, because many antigens thought to be tumor­specific turned out to be expressed by some normal cells as well. The modern classification of tumor antigens is based on their molecular structure and source.
An important advance in the field of tumor immunology was the development of techniques for identifying tumor antigens that were recognized by cytotoxic T lymphocytes (CTLs), because CTLs are responsible for the major immune defense mechanism against tumors. As described in Chapter 4, CTLs recognize peptides derived from cytoplas­ mic proteins that are displayed bound to class I major histocompatibility complex (MHC) molecules.
Described next are the main classes of tumor antigens (Fig. 5–32).
Products of Mutated Oncogenes and Tumor
Suppressor Genes
Neoplastic transformation, as discussed, results from genetic alterations, some of which may lead to the expres­ sion of cell surface antigens that are seen as non­self by the immune system. Antigens in this category are derived from mutant oncoproteins and tumor suppressor proteins. Unique tumor antigens arise from β­catenin, RAS, p53, and CDK4, for which the encoding genes frequently are mutated in tumors. Because the mutant genes are present only in tumors, their peptides are expressed only in tumor cells. Since many tumors may carry the same mutation, such antigens are shared by different tumors. Although CTLs can be induced against such antigens, they do not appear to elicit protective responses in vivo. In some cases, unmu­ tated oncogenes are overexpressed in tumors. The best

example is that of the HER2/NEU oncogene, whose product is highly expressed in a subset of breast cancers. Antibodies targeted against Her2/Neu protein are used clinically for the treatment of breast cancers.
Products of Other Mutated Genes
Because of the genetic instability of tumor cells, many genes are mutated in these cells, including genes whose products are not related to the transformed phenotype and have no known function. Products of these mutated genes are potential tumor antigens. These antigens are extremely diverse, because the carcinogens that induce the tumors may randomly mutagenize virtually any host gene. Mutated cellular proteins are found more frequently in chemical carcinogen­ or radiation­induced animal tumors than in spontaneous human cancers. They can be targeted by the immune system, since there is no self­tolerance against them.
Overexpressed or Aberrantly Expressed Cellular Proteins
Tumor antigens may be normal cellular proteins that are abnormally expressed in tumor cells and elicit immune responses. In a subset of human melanomas, some tumor antigens are structurally normal proteins that are produced at low levels in normal cells and overexpressed in tumor cells. One such antigen is tyrosinase, an enzyme involved
in melanin biosynthesis that is expressed only in normal melanocytes and melanomas. T cells from patients with melanoma recognize peptides derived from tyrosinase, raising the possibility that tyrosinase vaccines may stimu­ late such responses to melanomas; clinical trials with these vaccines are ongoing. It is somewhat surprising that these patients are able to respond to a normal self­antigen. The probable explanation is that tyrosinase normally is pro­ duced in such small amounts and in so few cells that it is not recognized by the immune system and fails to induce tolerance.
Another group, the so­called cancer­testis antigens, are encoded by genes that are silent in all normal adult tissues except the testis, and are deregulated in cancer cells— hence their name. Although the protein is present in the testis, it is not expressed on the cell surface in an antigenic form, because sperm do not express MHC class I mole­ cules. Thus, for all practical purposes, these antigens are tumor­specific. Prototypical of this group is the MAGE (melanoma antigen gene) family of genes. Although they are tumor­specific, MAGE antigens are not unique for indi­ vidual tumors. MAGE­1 is expressed on 37% of melanomas and a variable number of lung, liver, stomach, and esopha­ geal carcinomas. Similar antigens called GAGE, BAGE, and RAGE have been detected in other tumors. Several anti­ gens from this category are now being used in tumor vaccine trials. Tumor Antigens Produced by Oncogenic Viruses
As discussed earlier, some viruses are associated with cancers. Not surprisingly, these viruses produce proteins that are recognized as foreign by the immune system. The most potent of these antigens are proteins produced by latent DNA viruses; examples in humans are HPV and EBV. There is abundant evidence that CTLs recognize anti­ gens of these viruses and that a competent immune system plays a role in surveillance against virus­induced tumors because of its ability to recognize and kill virus­infected cells. Indeed, vaccines against HPV antigens have been found to be effective in prevention of cervical cancers in girls and young women.
Oncofetal Antigens
Oncofetal antigens or embryonic antigens, such as carcino­ embryonic antigen (CEA) and alpha fetoprotein, are expressed during embryogenesis but not in normal adult tissues. Derepression of the genes that encode these anti­ gens causes their reexpression in colon and liver cancers. Antibodies can be raised against these antigens and are useful for detection of oncofetal antigens. Although, as dis­ cussed later, they are not entirely tumor­specific, they can serve as serum markers for cancer.
Altered Cell Surface Glycolipids and Glycoproteins
Most human and experimental tumors express higher than normal levels and/or abnormal forms of surface glycopro­ teins and glycolipids, which may be diagnostic markers and targets for therapy. These altered molecules include gangliosides, blood group antigens, and mucins. Although most of the epitopes recognized by antibodies raised against such antigens are not specifically expressed on tumors, they are present at higher levels on cancer cells than on normal cells. This class of antigens is a target for cancer therapy with specific antibodies.
Several mucins are of special interest and have been the focus of diagnostic and therapeutic studies. These include CA­125 and CA­19­9, expressed on ovarian carcinomas, and MUC­1, expressed on breast carcinomas. Unlike many other types of mucins, MUC­1 is an integral membrane protein that normally is expressed only on the apical surface of breast ductal epithelium, a site that is relatively sequestered from the immune system. In ductal carcino­ mas of the breast, however, the molecule is expressed in an unpolarized fashion and contains new, tumor­specific car­ bohydrate and peptide epitopes. These epitopes induce both antibody and T cell responses in cancer patients and are therefore candidates for tumor vaccines.
Cell Type–Specific Differentiation Antigens
Tumors express molecules that normally are present on the cells of origin. These antigens are called differentiation anti- gens, because they are specific for particular lineages or differentiation stages of various cell types. Their impor­ tance is as potential targets for immunotherapy and in identifying the tissue of origin of tumors. For example, lymphomas may be diagnosed as B cell–derived tumors by the detection of surface markers characteristic of this lineage, such as CD20. Antibodies against CD20 are used for immunotherapy of certain B cell lymphomas. These differentiation antigens typically are normal self­antigens,
so they do not induce immune responses in tumor­bearing hosts.

31
Q

How do anti tumor effector mechanism help in tumor immunity

A

Antitumor Effector Mechanisms
Cell­mediated immunity is the dominant antitumor mech­ anism in vivo. Although antibodies can be made against tumors, there is no evidence that they play a protective role under physiologic conditions. The cellular effectors that mediate immunity are discussed fully in Chapter 4, so they are characterized only briefly here.
Cytotoxic T Lymphocytes
The role of specifically sensitized cytotoxic T lymphocytes (CTLs) in experimentally induced tumors is well estab­ lished. In humans, they seem to play a protective role, chiefly against virus­associated neoplasms (e.g., EBV­ induced Burkitt lymphoma, HPV­induced tumors). The presence of MHC­restricted CD8+ cells that can kill autolo­ gous tumor cells within human tumors suggests that the role of T cells in immunity against human tumors may be broader than was previously suspected. In some cases, such CD8+ T cells do not develop spontaneously in vivo but can be generated by immunization with tumor antigen– pulsed dendritic cells.
Natural Killer Cells
NK cells are lymphocytes that are capable of destroying tumor cells without previous sensitization; they may provide the first line of defense against tumor cells. After activation with IL­2, NK cells can lyse a wide range of human tumors, including many that seem to be nonim­ munogenic for T cells. T cells and NK cells apparently provide complementary antitumor mechanisms. Tumors that fail to express MHC class I antigens cannot be recog­ nized by T cells, but these tumors may trigger NK cells because the latter are inhibited by recognition of normal autologous class I molecules (Chapter 4). Thus, tumors may downregulate MHC class I molecules to avoid recog­ nition by T cells, which then makes them prime targets for NK cells. The triggering receptors on NK cells are extremely diverse and belong to several gene families. NKG2D pro­ teins expressed on NK cells and some T cells are important activating receptors. They recognize stress­induced anti­ gens that are expressed on tumor cells and on cells that have incurred DNA damage and are at risk for neoplastic transformation.
Macrophages
Classically activated macrophages of the M1 type (Chapter 2) exhibit cytotoxicity against tumor cells in vitro. T cells, NK cells, and macrophages may collaborate in antitumor reactivity, because interferon­γ, a cytokine secreted by T cells and NK cells, is a potent activator of macrophages. Activated macrophages may kill tumors by mechanisms similar to those used to kill microbes (e.g., production of reactive oxygen metabolites) (Chapter 2) or by secretion of tumor necrosis factor (TNF).
Humoral Mechanisms
Although there is no evidence for the protective effects of antitumor antibodies against spontaneous tumors,

administration of monoclonal antibodies against tumor cells can be therapeutically effective. A monoclonal anti­ body against CD20, a B cell surface antigen, is widely used for treatment of certain non­Hodgkin lymphomas.

32
Q

How does immune surveillance help in tumor immunity

A

Immune Surveillance
• Tumor cells can be recognized by the immune system as non-self and destroyed.
• Antitumor activity is mediated by predominantly cell- mediated mechanisms. Tumor antigens are presented on the cell surface by MHC class I molecules and are recog- nized by CD8+ CTLs.
• The different classes of tumor antigens include products of mutated proto-oncogenes, tumor suppressor genes, overexpressed or aberrantly expressed proteins, tumor antigens produced by oncogenic viruses, oncofetal anti- gens, altered glycolipids and glycoproteins, and cell type– specific differentiation antigens.
• Immunosuppressed patients have an increased risk for development of cancer.
• In immunocompetent patients, tumors may avoid the immune system by several mechanisms, including selective outgrowth of antigen-negative variants, loss or reduced expression of histocompatibility antigens, and immuno- suppression mediated by secretion of factors (e.g., TGF-β) from the tumor.

33
Q

What will make both malignant and benign tumors cause clinical problems

A

Indeed, both malignant and benign tumors may cause problems because of (1) location and impingement on adja­ cent structures, (2) functional activity such as hormone synthesis or the development of paraneoplastic syndromes, (3) bleeding and infections when the tumor ulcerates through adjacent surfaces, (4) symptoms that result from rupture or infarction, and (5) cachexia or wasting. The fol­ lowing discussion considers the effects of a tumor on the host, the grading and clinical staging of cancer, and the laboratory diagnosis of neoplasms.

34
Q

Explain how location can make a tumor cause a problem

A

Location is crucial in both benign and malignant tumors. A small (1­cm) pituitary adenoma can compress and destroy the surrounding normal gland, giving rise to hypo­ pituitarism. A 0.5­cm leiomyoma in the wall of the renal artery may encroach on the blood supply, leading to renal ischemia and hypertension. A comparably small carcinoma within the common bile duct may induce fatal biliary tract obstruction.

208
CHAPTER 5
Neoplasia
Hormone production is seen with benign and malignant neoplasms arising in endocrine glands. Adenomas and car­ cinomas arising in the beta cells of the pancreatic islets of Langerhans can produce hyperinsulinism, sometimes fatal. By analogy, some adenomas and carcinomas of the adrenal cortex elaborate corticosteroids that affect the patient (e.g., aldosterone, which induces sodium retention, hyperten­ sion, and hypokalemia). Such hormonal activity is more likely with a well­differentiated benign tumor than with a corresponding carcinoma.
A tumor may ulcerate through a surface, with con­ sequent bleeding or secondary infection. Benign or malig­ nant neoplasms that protrude into the gut lumen may become caught in the peristaltic pull of the gut, causing intussusception (Chapter 14) and intestinal obstruction or infarction.

35
Q

Explain how cancer cachexia can make a tumor cause a problem

A

Cancer Cachexia
Many cancer patients suffer progressive loss of body fat and lean body mass, accompanied by profound weakness, anorexia, and anemia—a condition referred to as cachexia. There is some correlation between the size and extent of spread of the cancer and the severity of the cachexia. However, cachexia is not caused by the nutritional demands of the tumor. Although patients with cancer often are anorexic, current evidence indicates that cachexia results from the action of soluble factors such as cytokines pro­ duced by the tumor and the host, rather than reduced food intake. In patients with cancer, calorie expenditure remains high, and basal metabolic rate is increased, despite reduced food intake. This is in contrast with the lower metabolic rate that occurs as an adaptive response in starvation. The basis of these metabolic abnormalities is not fully under­ stood. It is suspected that TNF produced by macrophages in response to tumor cells or by the tumor cells themselves mediates cachexia. TNF suppresses appetite and inhibits the action of lipoprotein lipase, inhibiting the release of free fatty acids from lipoproteins. Additionally, a protein­ mobilizing factor called proteolysis­inducing factor, which causes breakdown of skeletal muscle proteins by the ubiquitin­proteosome pathway, has been detected in the serum of cancer patients. Other molecules with lipolytic action also have been found. There is no satisfactory treat­ ment for cancer cachexia other than removal of the under­ lying cause, the tumor.

36
Q

Explain how paraneoplastic syndrome can make a tumor cause a problem

A

Paraneoplastic Syndromes
Symptom complexes that occur in patients with cancer and that cannot be readily explained by local or distant spread of the tumor or by the elaboration of hormones not indig­ enous to the tissue of origin of the tumor are referred to as paraneoplastic syndromes. They appear in 10% to 15% of patients with cancer, and their clinical recognition is impor­ tant for several reasons:
• Such syndromes may represent the earliest manifesta­ tion of an occult neoplasm.
• Inaffectedpatients,thepathologicchangesmaybeasso­ ciated with significant clinical illness and may even be lethal.
• The symptom complex may mimic metastatic disease, thereby confounding treatment.
The paraneoplastic syndromes are diverse and are associ­ ated with many different tumors (Table 5–5). The most common such syndromes are hypercalcemia, Cushing syndrome, and nonbacterial thrombotic endocarditis; the neoplasms most often associated with these and other syndromes are lung and breast cancers and hematologic malignancies. Hyper­ calcemia in cancer patients is multifactorial, but the most important mechanism is the synthesis of a parathyroid hormone–related protein (PTHrP) by tumor cells. Also implicated are other tumor­derived factors, such as TGF­α, a polypeptide factor that activates osteoclasts, and the active form of vitamin D. Another possible mechanism for hypercalcemia is widespread osteolytic metastatic disease of bone; of note, however, hypercalcemia resulting from skel- etal metastases is not a paraneoplastic syndrome. Cushing syn­ drome arising as a paraneoplastic phenomenon usually is related to ectopic production of ACTH or ACTH­like poly­ peptides by cancer cells, as occurs in small cell cancers of the lung. Sometimes one tumor induces several syndromes concurrently. For example, bronchogenic carcinomas may elaborate products identical to or having the effects of ACTH, antidiuretic hormone, parathyroid hormone, sero­ tonin, human chorionic gonadotropin, and other bioactive substances.
Paraneoplastic syndromes also may manifest as hyper­ coagulability, leading to venous thrombosis and non­ bacterial thrombotic endocarditis (Chapter 10). Other manifestations are clubbing of the fingers and hypertrophic osteoarthropathy in patients with lung carcinomas (Chapter 12). Still others are discussed in the consideration of cancers of the various organs of the body

37
Q

How is cancer graded and staged

A

Methods to quantify the probable clinical aggressiveness of a given neoplasm and its apparent extent and spread in the individual patient are necessary for making an accu­ rate prognosis and for comparing end results of various treatment protocols. For instance, the results of treating extremely small, highly differentiated thyroid adenocarci­ nomas that are localized to the thyroid gland are likely to be different from those obtained from treating highly anaplastic thyroid cancers that have invaded the neck organs.
The grading of a cancer attempts to establish some esti­ mate of its aggressiveness or level of malignancy based on the cytologic differentiation of tumor cells and the number of mitoses within the tumor. The cancer may be classified as grade I, II, III, or IV, in order of increasing anaplasia. Criteria for the individual grades vary with each form of neoplasia and are not detailed here. Difficulties in establishing clear­cut criteria have led in some instances to descriptive characterizations (e.g., “well­differentiated adenocarcinoma with no evidence of vascular or lymphatic invasion” or “highly anaplastic sarcoma with extensive vascular invasion”).
Staging of cancers is based on the size of the primary lesion, its extent of spread to regional lymph nodes, and the presence or absence of metastases. This assessment usually is based on clinical and radiographic examination (computed tomography and magnetic resonance imaging) and in some cases surgical exploration. Two methods of

staging are currently in use: the TNM system (T, primary tumor; N, regional lymph node involvement; M, metasta­ ses) and the AJC (American Joint Committee) system. In the TNM system, T1, T2, T3, and T4 describe the increasing size of the primary lesion; N0, N1, N2, and N3 indicate progressively advancing node involvement; and M0 and M1 reflect the absence and presence, respectively, of distant metastases. In the AJC method, the cancers are divided into stages 0 to IV, incorporating the size of primary lesions and the presence of nodal spread and of distant metastases. Examples of the application of these two staging systems are cited in subsequent chapters. Of note, when compared with grading, staging has proved to be of greater clinical value.

38
Q

Clinical Aspects of Tumors
• Cachexia, defined as progressive loss of body fat and lean body mass, accompanied by profound weakness, anorexia, and anemia, is caused by release of cytokines by the tumor or host.
• Paraneoplastic syndromes, defined as systemic symptoms that cannot be explained by tumor spread or by hor- mones appropriate to the tissue, are caused by the ectopic production and secretion of bioactive substances such as ACTH, PTHrP, or TGF-α.

210
CHAPTER 5
Neoplasia
• Grading of tumors is determined by cytologic appearance and is based on the idea that behavior and differentiation are related, with poorly differentiated tumors having more aggressive behavior.
• Staging, determined by surgical exploration or imaging, is based on size, local and regional lymph node spread, and distant metastases. Staging is of greater clinical value than grading

True or false

A

True

39
Q

State some diseases in Paraneoplastic Syndromes,the major form of neoplasia and the causal mechanism

A

Clinical Syndrome
Major Forms of Neoplasia
Causal Mechanism(s)/Agent(s)

Endocrinopathies
   Cushing syndrome 
Small cell carcinoma of lung
 Pancreatic carcinoma
Neural tumors
Causal mechanism: ACTH or ACTH-like substance

Syndrome of inappropriate antidiuretic hormone secretion
Small cell carcinoma of lung; intracranial neoplasms
Causal:Antidiuretic hormone or atrial natriuretic hormones

Hypercalcemia :
Squamous cell carcinoma of lung 
Breast carcinoma
Renal carcinoma
Adult T cell leukemia/lymphoma Ovarian carcinoma

Causal:Parathyroid hormone–related protein, TGF-α, TNF, IL-1

Hypoglycemia :
Fibrosarcoma
Other mesenchymal sarcomas
Hepatocellular carcinoma

Causal:Insulin or insulin-like substance

Carcinoid syndrome:
Bronchial adenoma (carcinoid)
Pancreatic carcinoma
Gastric carcinoma

Causal:Serotonin, bradykinin

Polycythemia :
Renal carcinoma
Cerebellar hemangioma
Hepatocellular carcinoma

Causal: Erythropoietin

Nerve and Muscle Syndrome
Myasthenia ;
Bronchogenic carcinoma, thymoma causal:Immunologic

Disorders of the central and peripheral nervous systems

Breast carcinoma, teratoma

Dermatologic Disorders

Acanthosis nigricans :
Gastric carcinoma
Lung carcinoma
Uterine carcinoma

Causal: Immunologic; secretion of epidermal growth

Dermatomyositis :Bronchogenic and breast carcinoma
Immunologic

Osseous, Articular, and Soft Tissue Changes
Hypertrophic osteoarthropathy and Unknown clubbing of the fingers:
Bronchogenic carcinoma

   Vascular and Hematologic Changes
   Venous thrombosis (Trousseau phenomenon):
Pancreatic carcinoma 
Bronchogenic carcinoma
Other cancers

Causal: Tumor products (mucins that activate clotting))

Nonbacterial thrombotic endocarditis :Advanced cancers
Causal:Hypercoagulability

Anemia :Thymoma
Causal:Immunologic

Others
Nephrotic syndrome Various cancers
Causal:Tumor antigens, immune complexes

40
Q

How is cancer diagnosed in the lab

A

Laboratory Diagnosis of Cancer
• Several sampling approaches exist for the diagnosis of tumors, including excision, biopsy, fine-needle aspiration, and cytologic smears.
• Immunohistochemistry and flow cytometry studies help in the diagnosis and classification of tumors, because distinct protein expression patterns define different entities.
• Proteins released by tumors into the serum, such as PSA, can be used to screen populations for cancer and to monitor for recurrence after treatment.
• Molecular analyses are used to determine diagnosis, prog- nosis, the detection of minimal residual disease, and the diagnosis of hereditary predisposition to cancer.
• Molecular profiling of tumors by cDNA arrays and sequencing can determine expression of large segments of the genome and catalog all of the mutations in the tumor genome and thus may be useful in molecular strati- fication of otherwise identical tumors or those of distinct histogenesis that share a mutation for the purpose of treatment and prognostication.

41
Q

State four targets of genetic damage in malignant cells and give one example each

What are the differences between governors and guardians

What is mutator phenotype

A

.
• Four classes of normal regulatory genes—growth-promoting proto-oncogenes, growth-inhibiting tumor suppressor genes(TP53 which is a guardian and RB which is a governor gene), genes that regulate programmed cell death (i.e., apoptosis), and genes involved in DNA repair—are the principal targets of genetic damage.

Tumor suppressor genes are usefully placed into two general groups, “governors” and “guardians.” “Gover­ nors” are classic tumor suppressor genes, such as RB, where mutation of the gene leads to transformation by removing an important brake on cellular proliferation. “Guardian” genes are responsible for sensing genomic damage. Some of these genes initiate and choreograph a complex “damage control response.” This response leads to the cessation of proliferation or, if the damage is too great to be repaired, the induction of apoptosis. TP53, the so­called “guardian of the genome,” is a pro­ totypic tumor suppressor gene of this type. Other guard­ ian genes are directly involved in recognizing and repairing specific kinds of DNA damage; these are the genes that are mutated in the autosomal recessive syndromes of DNA repair.

This increase in mutation rate is often referred to as a mutator phenotype.
• Genes that regulate apoptosis and DNA repair may act like proto­oncogenes (loss of one copy is sufficient) or tumor suppressor genes (loss of both copies)

42
Q

State the difference between a. Primary and secondary malignant cancer

A

Primary cancer is defined as the original site (organ or tissue) where cancer began. In contrast, a second or secondary cancer may be defined in a few ways; as either a new primary cancer in another region of the body or as metastasis (spread) of the original primary cancer to another region of the body.

43
Q

What will allow cancer to move from its primary site

What three features of cancer cells allow them to be metastatic

A

Through the circulatory (blood) system (hematogenous)
Through the lymphatic system.
Through the body wall into the abdominal and chest cavities (transcoelomic).

However in very basic terms, the acquisition of metastasis requires several fundamental steps including (1) loss of cell-cell adhesion, (2) acquisition of motility and (3) the ability to digest through the basement membrane to enter the circulation.