• Shuffle
    Toggle On
    Toggle Off
  • Alphabetize
    Toggle On
    Toggle Off
  • Front First
    Toggle On
    Toggle Off
  • Both Sides
    Toggle On
    Toggle Off
  • Read
    Toggle On
    Toggle Off
Reading...
Front

Card Range To Study

through

image

Play button

image

Play button

image

Progress

1/523

Click to flip

Use LEFT and RIGHT arrow keys to navigate between flashcards;

Use UP and DOWN arrow keys to flip the card;

H to show hint;

A reads text to speech;

523 Cards in this Set

  • Front
  • Back
Neoplasia/neoplasm
"new cell growth"
-physical evidence of pathologic new cell growth
-solid mass lesion/tumor or a diffuse process like leukemia
Tumor
-solid mass (subcategory of a tumor)
Cancer
-reserved for MALIGNANT tumors only
Clinical presentation of tumors (general)
-often present as occult disease (hidden) with systemic symptoms or metabolic alterations
-mass lesion present
-fever
-anemia
-anorexia, cachexia
-ectopic hormone production
-pathologic bone fracture

**must exclude tumor as an explanation for odd physical or lab results
Diagnosing/treating tumors
-may be able to treat surgically
-in hollow organs, lesions can cause pain, bleeding, and obstruction; dx with biopsy
-radiographs can find symptomatic or asymptomatic mass lesions (ex: density in lung fields in a smoker is suspicious for lung cancer, but if its in a patient who doesnt smoke but had prior cancer, indicates metastasis to lungs)
Most important factor in determining the treatment and prognosis of a tumor
-histologic diagnosis!
-made by a surgical pathologist
-benign or malignant
Benign (and 4 characteristics)
1) differentiation/anaplasia
2) rate of growth
3) local invasion
4) metastasis
-tumor does not metastasize
1) well differentiated tissue, usually typical of the tissue of origin
2) progressive, slow growth; may stop or regress..**rare, normal mitotic figures
3) well demarcated, cohesive, expansile masses that DO NOT invade surrounding tissue
4) NO METASTASIS)
Malignant (and 4 characteristics)
1) differentiation/anaplasia
2) rate of growth
3) local invasion
4) metastasis
-tumor is capable of metastasis
1) often lacks some differentiation = anaplasia; tissue may be atypical of origin tissue
2) erratic growth, slow or rapid...*numerous, abnormal mitotic figures
3) invades local surrounding tissue, although it may appear cohesive or expansile
4) metastasis is frequently present, depends on how large and undifferentiated the primary tumor is!!
Uterine leiomyomas
-fibroids
-most common benign tumors in women
-can be large or multiple or symptomatic; can cause discomfort, morbidity and even death, but does not metastasize
-like benign tumors, can recur if not completely metastasized
Malignant melanoma
-primary skin tumor that can metastasize; it is malignant
-even if primary is very small, can give rise to large, multiple, distant metastases
Carcinoma
-malignant EPITHELIAL tumor
Epithelial tumors: Papilloma
-benign
-origin: squamous epithelium
-forms masses (Benign proliferation)
Epithelial tumors: Squamous cell carcinoma
-malignant
-origin: squamous epithelium
-forms masses

ex: in cervix
Epithelial tumors: Adenoma
-benign
-origin: glandular epithelium

ex: colonic adenomas (detected by colonoscopy .. important in prevention of adenocarcinoma)

-grow as exophytic polyps, can be biopsied to look for early malignant changes
Epithelial tumors: Adenocarcinoma
-malignant
-origin: glandular epithelium

-can result from untreated adenomas of the colon
-grow in ulcerated pattern
*malignant glands have INVADED wall of colon, can now metastasize
Sarcoma
-malignant mesenchymal (connective tissue) tumor
-uncommon, associated with specific inherited cancer syndromes or increased susceptibility
Mesenchymal tumors: Lipoma
-benign
-common in adults
-origin: adipose tissue

-differentiated from liposarcomas by histology and by location: usually benign if superficial or in subQ of adipose tissue
Mesenchymal tumors: Liposarcoma
-malignant
-origin: adipose tissue

-usually malignant if deep in the adipose tissue of the extremities or retroperitoneum
-many are indolent
Mesenchymal tumors: Leiomyoma
-benign
-origin: smooth muscle
Mesenchymal tumors: Leiomyosarcoma
-malignant
-origin: smooth muscle
Mesenchymal tumors: Rhabdomyoma
-benign
-origin: skeletal muscle
Mesenchymal tumors: Rhabdomyosarcoma
-malignant
-origin: skeletal muscle
Mesenchymal tumors: Angioma
-benign
-origin: blood vessels
Mesenchymal tumors: Angiosarcoma
-malignant
-origin: blood vessels
Mesenchymal tumors: Osteoma
-benign
-origin: bone
Mesenchymal tumors: Osteosarcoma
-malignant
-origin: bone

-usually associated with bony skeleton but can be extraskeletal
Teratoma
**rarely malignant
-have mature tissues from more than one germ layer, and often all 3: ectoderm, endoderm, mesoderm

*most common location: ovary
Mature cystic teratoma (dermoid cyst)
-in ovary of young adult women
-most common type of teratoma

-hair, skin, teeth, brain, fat are common components, but occasionally some are monodermal (ex: thyroid tissue)
Hamartoma
-mass of normal but disorganized tissues indigenous to the site of origin

*most common location: lung (incidental finding on x-ray)
Heterotopia
-ectopic rest of normal tissue
Classification: CNS tumors, leukemias, lymphomas
-own nomenclature system DIFFERENT from the solid tumors listed above
Cancer incidence rate: reasons for increase
-can be reflective of prevalence, but is influenced by screening tests with increased sensitivity for the tumor (more diagnoses picked up on, but not truly new cases)

Also: increased life span (also related to more potential exposures to carcinogens and weakening immunity)

Can be a true increased prevalence for certain cancer due to exposure to a known carcinogen

Note: family history and lifestyle choices also influence risk, but not necessarily influence
Cancer survival
-proportion of patients alive at some point after cancer diagnosis
More breast and prostate cancer?
Not true increase in incidence
-reflection of effective screening programs for PSA and mammography
More lung cancer?
-may be a true increase in incidence due to the introduction of cigarettes

ex: more women began dying of lung cancer in the 60s b/c they started smoking = true increase in prevalence

**some cancers can have a long latency period b4 cancers begin to show up
Pediatric tumors
-cancer incidence is not distributed evenly, specific types depend on age
-carcinomas present in middle/older age
-children get leukemias and neural, mesenchymal origin tissue tumors
*for kids, strong component of inherited susceptibility, because haven't lived long enough to get enough carcinogen exposure
Hereditary Neoplasia
-hereditary tumors have a more evenly distributed incidence, present evenly throughout all ages, versus sporadic ones that are uncommon before middle age

-inherited traits make ppl more susceptible to cancer at earlier ages, get multiple tumors that are often bilateral in paired organs; some even have mendelian inheritence

BUT the same genes involved in hereditary tumors are also involved in the sporadic versions

Note: development of cancer at a younger age = strong association with inherited factors; if at old age, less of an association exists
Genetic damage role in causing cancer
-occurs as non-lethal damage in tumor cells **meaning it can be transmitted to future generations of cancer cells
-acquired from environment or inherited in the germ line
Genetic instability
-exhibited by most tumors
-leads to accumulation and propagation of more genetic mutations
-leads to tumor progression

*carcinogenesis is a multistep processes in both genotypic and phenotypic levels = tumor progression
Gatekeeper genes
-regulate entry of cells into tumorigenic pathway

ex: tumor suppressor genes (inhibit growth)

**if germ line mutation of these, much higher risk of cancer, even MORE risk than if mutation inherited in caretaker gene
Caretaker genes
-do not directly control tumor growth
-BUT affect genomic instability

ex: DNA mismatch repair genes, other repair genes
-loss leads to increased mutation in all genes that INCLUDES GATEKEEPER genes
**if germ line mutation of these, higher risk of cancer but LESS risk than if mutation inherited in gatekeeper gene
Tumor suppressor genes
-their protein products inhibit cell proliferation
-loss of function leads to uninhibited cell growth and is a key event if many tumors

*physiologic goal is to regulate cell growth, NOT prevent tumor formation
Knudson hypothesis
"two hit" hypothesis
-to have loss of function of the TSGs, must have deletion or silencing of BOTH ALLELES at a TSG locus - this results in the tumor phenotype

*presence of one normal allele is enough to preserve normal cell function
-however, if inherit one normal allele and one altered to begin with, increases susceptibility to particular cancers because it only takes one more mutation instead of two
Retinoblastoma genetics
-BOTH normal alleles of the RB locus must be inactivated (two hits) in order for Retinoblastoma to develop
Leukocoria
-sign of retinoblastoma
-pupil reflects incident light due to the tumor in the vitreous chamber
-pupil is not black as it should be
Hereditary (familial) Retinoblastoma
-one normal copy and one defective copy of RB are inherited

-if other copy mutates during lifetime by somatic mutation, retinal cell has lost both copies and gives rise to cancer

*these patients also at increased risk for osteosarcoma and soft tissue sarcomas
Sporadic Retinoblastoma
-two normal RB alleles are inherited, but both RB alleles are lost by somatic mutation during lifetime

-as with the familial type, once both normal copies have been lost in the retinal cell, gives rise to cancer

**note: these patients do not have same risk factors for other cancers as do the familial RB patients
Loss of heterozygosity
-cancer occurs when a cell becomes homozygous for a mutant allele, aka it loses its heterozygosity for the normal RB gene

*this occurs in individuals who inherit one mutant TSG allele (ex: one mutant RB allele)
Note: in these individuals, all of their somatic cells are normal, except that they are at an increased risk of cancer (if any cells lose heterozygosity for the normal allele)
Oncogenes
-genes that promote autonomous cell growth in cancer cells, even in the absence of normal mitotic signals
*mutated forms of their normal cellular counterparts, protooncogenes
Protooncogenes
-NORMAL cell components
-regulate cell proliferation and differentiation (promote growth)
-if mutated, turn into oncogenes
Oncoproteins
-similar to the normal protein products of protooncogenes, but lacking regulatory elements so they become CONSTITUTIVELY expressed

**gain of function mutation
Gain of function mutation
-associated w/ oncogenes (like loss of heterozygosity for TSGs)

**only ONE allele needs to be mutated to create a phenotypic effect - an abnormal protein that becomes dominant, promotes its own cell growth (autonomous)
How are oncogenes activated? 2 ways
-reduplication and amplification of their DNA sequences
-leads to overexpression

ex: HER2/neu amplification in 25% of breast cancers

---------------------------------------------
**Also may occur by chromosomal translocations (expecially if involving TKs)
-Philadelphia chromosome translocation creates the Bcr-Abl oncoprotein
Genetic alterations of solid tumors
-much more complex, hard to isolate targets for drugs in order to treat
-in comparison to sarcomas and hematopoietic cancers, which are due to translocations, etc. as discussed before
Mismatch repair
-normally caretaker genes
-damage to pathways lead to inability to correct errors in replication
-allow development of somatic mutations

*defects in this system can be inherited or can be somatic

-not themselves oncogenic, but allow mutations in protooncogenes..

*if inherited, usually autosomal recessive (xeroderma pigmentosum, ataxia-telangectasia, Bloom syndrome)
Hereditary Nonpolyposis Colon Cancer
-autosomal dominant
-caused by inactivation of a DNA mismatch repair gene

***most common cancer predisposition syndrome
Methylation
-methylate promotor sequences (CpG islands)
-normal epigenetic regulation process that silences genes but not loss of genes

*if hypermethylate TSGs, will silence them even though the gene itself is not mutated
Steps in carcinogenesis: initiation
-permanent, nonlethal DNA damage that is IRREVERSIBLE, can be transmitted to the cell's progeny
-mutated DNA template is replicated and becomes heritable
Steps in carcinogenesis: promotion
-REVERSIBLE biologic processes that can favor neoplasm development (stimulate proliferation of initated cells)
-do NOT cause genetic damage
Steps in carcinogenesis: latency
-time from exposure to appearance of neoplasm
-due to interplay between initiation and promotion after exposure to carcinogen
Carcinogen
-agent that increases incidence of neoplasm
Neoplastic Cell growth vs. Normal growth
-normal growth is at a steady state between loss and creation of cells
-tumors have autonomy of cell division = continued growth
Benign vs. Malignant neoplastic cell growth
-benign = limited by adjacent normal tissue, rate of enlargement is slow

-malignant = nonstop growth, but limited by cell loss due to degenerative changes in the tumor??
Tumor growth fraction
-fraction of tumor cells in the replicative pool
-determines rate of growth
**major effect on susceptibility to chemo (ex: aggressive tumors have a larger fraction, very responsive to chemo)
Tumor doubling time
-growth fraction cells are dividing, but not necessarily finishing cell cycle faster than normal cells; may be longer

*if larger growth fraction, FASTER doubling time

**most carcinomas double in 2-3 months
How many doublings before the tumor is clinically detectable?
-30 doublings, or 10^9 cells
=1 gram

*Note: only 10 more doublings needed before it becomes 10^12 cells and 1kg aka maximal size compatible with life!!
-therefore, by the time the cancer is detected, tumor has already completed most of its life cycle with very heterogenous mix of cells (result of many independent mutations from genetic instability ... each cell type has different growth rate, metastatic abilities, etc) ..remember that tumor progression is acquired incrementally!
Tumor progression in colon cancer
-like certain cancers, has a more predictable pattern with specific alterations from the incremental accumulation of mutations
(but still, heterogeneity occurs far before morphologic changes are clinically evident)
Steps in tumor invasion for malignant neoplasms
- tumor cells adhere to ECM components; must break through basement membrane of epithelial cells to get to the stroma
-BM sends positive and negative growth signals to the tumor cells, regulates angiogenesis
Angiogenesis
-blood vessel growth stimulated by tumors
-essential for supplying nutrients to tumor
-without vascularization, can't enlarge beyond 1-2mm in diameter (max distance that nutrients/O2 can diffuse across tissue)

**angiogenesis also required for metastasis

MOA: recruit endothelial cell precursors via existing capillaries
-tumor blood vessels are more tortuous and leaky due to increased production of VEG-F
-tumor cells produce angiogenic factors like VEGF and bFGF
Anti-angiogenic therapy
new field aimed primarily at blocking VEGF-A/VEGFR2 signaling in order to treat cancers
Metastasis
-tumor lesions discontinuous with the primary tumor
*primary tumors have preferential locations for metastasis, but not exact
-lets cancers penetrate, vessels, lymph, body cavities

*risk factors: more aggressive, more rapidly growing, larger the primary neoplasm
"Seed and Soil" idea of metastasis
-solid tumors need interaction between malignant cells as well as the stromal cells it has invaded at both the primary and metastatic sites
Pathways of Metastasis (3)
1) Direct seeding of body cavities/surfaces
2) Lymphatic spread
3) Hematogenous spread
Pathways of Metastasis: Direct seeding of body cavities/surfaces
-extension beyond organ of origin directly into adjacent structures
ex: cervical carcinoma
-penetrates wall of bladder and rectum
-makes surgery difficult, not curative (probably has metastasized elsewhere)

**seeding of body cavities occurs with certain carcinomas and melanomas
Pathways of Metastasis: Lymphatic spread
**most common pathway for initial metastasis of CARCINOMAS

-ex: breast carcinoma in upper outer quadrant disseminate to axillary LNs
Pathways of Metastasis: Hematogenous spread
**most common pathway for SARCOMAS
-venous invasion, cells travel via vein draining the site of the neoplasm

**peripheral sarcomas tend to metastasize to the lungs
Liver metastasis
-common site for tumors of the abdominal organs (like colon)
-also occurs via hematogenous metastasis
Bony Metastases
**most commonly from Prostate, Thyroid, Breast, Lung, Kidney
Peritoneal Seeding
-natural open space; other body cavities (pleural, pericardial, joint space, etc) can also be affected

*ovarian carcinoma, appendix carcinoma, melanoma (direct seeding)
Metaplasia
-reversible substitution of one adult cell type for another adult cell type
-may be an adaptation to stressors (cell types better able to withstand the environment due to inflammation, vit deficiency, etc)
*not preneoplastic and is reversible, but malignant transformation can arise if
Most common epithelial metaplasia
-columnar to squamous
-ex: in the Uterine cervix
-also in respiratory tracts of smokers (ciliated columnar replaced by stratified squamous in trachea and bronchi - **lose mucus secretion function!!)
Anaplasia
-lack of differentiation
-may exhibit pleomorphism (abnormal nuclei, size and shape), abnormal nuclear morphology, atypical mitotic figures
Dysplasia
*preneoplastic condition but does not imply it
**in GI, GU, respiratory epithelia
-may revert back to normal if irritant removed if low grade (CIN 1)
**HPV causes cervical, uterine, endocervix dysplasia

-disordered growth and pathological hyperplasia, esp in epithelium

-architectural abnormalities, loss in uniformity of individual cells, abnormal nuclear changes - increased mitotic figures, abnormal basement membrane, nuclear hyperchromasia
Atypical hyperplasia
*preneoplastic condition like dysplasia
**in breast and endometrium


-increased pathological cell proliferation
-disordered growth, esp in epithelium
-architectural abnormalities, loss in uniformity of individual cells, abnormal nuclear changes
Carcinoma in situ (grade CIN 3)
-dysplastic changes envolving entire thickness of the epithelium (ex: severe cervical dysplasia)
**confined to epithelium, NO basement membrane penetration
-high risk for progressing to invasive cancer
Invasive squamous cell carcinoma of the cervix
-from carcinoma in situ
-tumor penetrated epithelial basement membrane
-tumor cells have invaded subepithelial soft tissue
-tumor now has access to lymphatic channels, can metastasize
Ductal carcinoma in situ (DCIS)
-results from breast preneoplasia (atypical hyperplasia)
-duct filled with atypical cells that remain in the basement membrane
Invasive ductal carcinoma
-tumor cells have penetrated and stimulate formation of abundant collagenous tumor stroma = desmoplasia
-makes the tumor hard (scirrhous)
Grading Malignang Neoplasms
-based on degree of differentiation and # of mitosis
-indicates aggressiveness
-Grades I - IV .. relate to increasing anaplasia, although criteria vary
*allows for prognosis and treatment efficacy

ex: squamous cell carcinoma - low grade is well differentiated (intracellular bridges; stratified, flattened cells, cytologically okay) versus moderately differentiated (more cellular, less organized, less flattened, loss of stratification)
-ex: adenocarcinoma, low grade has recognizable glandss and moderate nuclear pleomorphism, whereas high grade (moderately differentiated) has poorly formed glands, pleomorphism and abnormal mitotic figures
Immunohistochemistry
-ancillary technique to categorize undifferentiated malignant tumors
-helps determine site of origin of a hidden primary tumor
-used to classify leukemia, lymphoma
*prognostic and therapeutic molecules that can be target

*uses monoclonal antibodies
-formalin fixed, paraffin embedded tissue
-localizes specific proteins in the tissue
Cancer of unknown origin
-metastasis with no obvious primary site, which is important to identify for therapy and prognosis
-can do immunohistochemistry!
-helps subclassify carcinomas, lymphomas and sarcomas and leukemias that may otherwise look similar
Review p. 261
-markers
Breast Cancer and Immunohistochemistry
-detects receptors in the nucleus
-if ER or PR or HER2/neu positive, can be offered hormonal therapy, better prognosis than women (anti-estrogens if ER+, trastuzumab if HER2/neu positive)
Sentinel Lymph node
-first node in a regional lymphatic basin that receives lymph flow from the primary tumor

*determining involvement of axillary LNs is important in staging
Fluorescent in situ Hybridization
-evaluates gene copy number in certain tumors
-detects translocations, etc. in leukemias, lymphomas
*can assess status of a single gene
-diagnostic, prognostic uses

**used to detect HER2/neu on chrom 17q21 (normal cell has 2 copies of gene; tumor cells with amplification have over 20)
Tumor markers
-cell surface antigens, cytoplasmic proteins, enzymes, hormones
**NOT diagnostic of cancer!! can be elevated in reactive or inflammatory conditions (ex: PSA)

*indicators of the recurrence of a tumor, or support diagnosis of cancer; can be used to monitor response to therapy (PSA for prostate, CEA for colonic adenocarcinoma, AFP for hepatocellular carcinoma and germ cell tumors, or CA-125 for ovarian carcinoma)

-can be detected in plasma or body fluids
Staging: TNM system
-T = primary tumor size
-N = regional lymph node involvement
-M = metastatic

*Stage IV reserved for distant metastasis (once this is present, cure unlikely, but long term remission and palliation are possible, esp. if tumor is hormone responsive)
AJCC Breast Cancer Stages
-I = tumor <2cm, confined to breast, no nodes involved
-II = tumor > 2 cm, metastasis in axillary nodes but not distant metastasis
-III = tumor of any size, extends into chest wall or skin, no distant metastasis
-IV = tumor of any size, with or w/o chest wall involvement or nodal metastases, but distant metastasis present
Most important prognostic factor for breast cancer
**Axillary lymph node status!!
-most important prognostic factor for invasive carcinoma in absence of distant metastasis
2nd most important prognostic factor
*tumor size
-independent of lymph node status (although risk of axillary LN status increases with size of carcinoma)
Gompertzian curve
-mathematical model describing tumor cell growth
-tumor cells grow exponentially in the early stage
*tumor takes constant amount of time to double
-Growth fraction is high
**as tumor grows, doubling time and growth fraction slowed due to lack of nutrients, etc.
Best time to attack cancer cells
-early growth phase
-more susceptible to chemo when doubling time is short
Immunotherapy
-used when tumor burden is below 10^5 cells
-use of biologic agents
Log-kill hypothesis
-fraction of cells killed with each cycle of chemo is constant, regardless of tumor burden (assumes homogenous sensitivity to chemo)
How many logs are killed by one round of chemo?
-3 logs
ex: 10^10 --> 10^7

**regrowth occurs between cycles!
Goldie-Coldman Hypothesis
-best strategy for treatment is initiate early
-combine drugs with non-overlapping toxicities

**Goal: prevent tumor resistance!!
Types of treatment (6)
-single agent
-combo chemo (many agents)
-combo chemo/radiation together
-sequential therapy (combination of chemo followed by another single or combo of chemo)
ex: AC--> Docetaxel
-immunotherapy
-hormonal therapy
-other combinations (radiation and surgery)
Treatment of liquid tumors (3)
-induction
-consolidation/adjuvant
-maintenance
Treatment of solid tumors (3)
-adjuvant therapy
-neoadjuvant therapy
-palliation
Complete response
**REMISSION
-disappearance of all evidence of tumor
Partial response
-reduction of at least 50% of the diameter of all lesions
Stable disease
-decrease of less than 50% or increase of less than 25% in diameter of lesion
Progressive disease
-increase of more than 25% in diameter of any lesion
Induction
-initial treatment to get patient into COMPLETE REMISSION
Consolidation/Adjuvants
-used to eradicate micrometastatic disease
Maintenance
-prolonged therapy

**Used especially for ALL
Neoadjuvant
-used to reduce tumor burden before surgery
Chemo cycles
-3 days chemo, 18 days rest, then repeat (21 days total - lets WBCs replete)
Tumor burden and chemo response
-high initial burden lowers responsiveness of tumor to chemo

(Note: if tumor is heterogeneous, more likely to have chemo resistance)
General toxicity: Nausea and vomiting
-causes electrolyte imbalances, decreased nutrition, dehydration

*N/V receives input from GI tract (vagal, sympathetic afferents from stomach, serotonin from small intestine), cerebral cortex, vestibular apparatus, chemoreceptor trigger zone and is coordinated by the vomiting center in the meulla, sends efferents to salivary/respiratory and GI tracts
-many NTs involved: sub P, GABA, Ach, serotonin, dopamine, histamine, endorphins
Acute chemo-induced N/V
-in first 24 hours after chemo, peaks at 3-4 hours later
-treat with serotonin and nk-1 antagonists, corticosteroids
Delayed chemo-induced N/V
-3-5 days after chemo
-tx. with dopamine antagonists, corticosterois, nk-1 antagonists

*associated with cisplatin, cyclophosphamide and doxorubicin
Anticipatory chemo-induced N/V
-before chemo
-treat with benzodiazepenes
N/V Drugs: Serotonin antagonists
-bind serotonin receptors in small intestine and in the CTZ of brain
-odansetron, granisetron, dolasetron .. given IV or oral

**treat acute CINV, not delayed

Side effects: headache, hiccups, QT prolongation, constipation
Palonosetron
-newest 5-HT3 antagonoist
-one dose per cycle of chemo (long half life, can be dosed every 5-7 days, whereas others only last a few hours)
Aprepitant
-substance P action (via neurokinin-1 receptor aka nk-1); NK1 antagonist
-for both acute and delayed

Side effects: *multiple drug interactions (CYP34A - if use with dexamethasone, reduce dexa dose), hiccups, fatigue
N/V Drugs: Dopamine antagonists
-bind dopamine D1 and 2 in CTZ
-prochloperazine, metoclopramide, perphenazine
-for acute and delayed N/V

Side effects: **extrapyramidal side effects (if mild treat with benadryl), sedation

(Prokinetic agents are like these drugs but cause diarrhea also as a side effect)
Dexamethasone
--corticosteroid
-mechanism unknown for antiemetic

Side effects: glucose intolerance, insomnia, nervousness
Olanzapine
-atypical antipsychotic
-in trials
-great for acute, less so for delayed

*for refractory cases; antagonist of multiple dopamine, serotonin, histamine receptors...etc. can cause sedation and weakness
Erythropoietin (Epo)
30-50% of patients respond

*treats anemia from chemo ONLY IF THERE ARE ALREADY ADEQUATE IRON STORES (ex: cisplatin effects on kidney reduce erythropoietin production)
-do iron studies before tx, if iron low, replete iron first

*Side effects: HT, rash, arthralgia

**At 12 Hgb, stop treatment until Hgb drops below 12 again (above 12 can cause death)

-Procrit (short acting, every 1-2 weeks, but works faster to increase HgB) or Darbepoetin (long acting, every 2-3 weeks)
-reassess every 4-6 weeks (should have >1Hgb increase)

Note: **this is ONLY used for chemo patients, not all cancer patients w/ anemia
Neutropenia
-absolute neutrophil count <500 (some say <1000)

-when ANC <1000, at high risk for infection, which is also related to the length of the neutropenia
Myeloid growth factors
-enhance prolif/differentiation of myeloid cell lines, aka neutrophils
**to treat neutropenia in chemo patients

-Filgrastim = GCSF, short acting, Pegfilgrastim, long acting GCSF, Sargramostim = GMCSF, short acting)
**interchangeable
**can be used as primary prophylaxis - before chemo for neutropenia (with drugs having a high incidence of it); secondary prophylaxis, which is right after chemo for curative intent; or for established neutropenia

***treat until ANC >1000 (will drop by half after you discontinue it)
Pegfilgrastim
-polyethylene glycol attached, higher MW, which decreases clearance
-longer half life, but clearance increases as neutrophils recover (self regulating)

**only given once after chemo!
Side effects: fever, bone pain, increases in uric acid, LDH, alkaline phosphatase, rash

Downside: EXTREMELY EXPENSIVE
Origin of all cells in the blood, immune system in an adult
-BONE MARROW
Leukemia versus Lymphoma
Leukemia: malignant cells circulate in blood
**can be seen on a blood smear

Lymphoma: malignant cells found mainly within lymphoid organs
B cell lymphomas mimic what cell types?
-mimic stages of B cell differentiation that take place in the LYMPH NODES
(follicular dendritic cells, mantle zone cells, marginal zone cells, germinal center cells, etc, ...)
B-lymphoid acute leukemias mimic what cell types?
-immature B-lymphoid stages that take place in the BONE MARROW
Biopsy site for leukemia
-bone marrow (even though the tumor cells are circulating)
Biopsy site for lymphoma
-enlarged lymph node or extranodal mass
Myeloperoxidase
-histochemistry can differentiate similar leukemias if morphology isnt good enough

**ALL is myeloperoxidase NEGATIVE whereas AML is POSITIVE; even though they appear similar
Which antigens are expressed in AML? (2)
CD 13 and CD 33
Which antigens are expressed in ALL? (2)
CD 10 and CD 20
How do you detect cell surface antigens (CDs)?
Flow cytometry
What is the chromosomal abnormality in Burkitt's lymphoma?
-translocation of long arms of 8 and 14

t(8;14)

**example of cytogenetics
Explain how this translocation causes Burkitt's lymphoma.
-t(8;14) brings the c-MYC gene (normally tightly regulated) on 8 next to an Ig heavy chain gene promotor on 14 (highly active in B cells
-overexpression of c-MYC can be deteted by immunohistochemical staining
-MYC is a transciption factor, binds DNA, activates cellular proliferation that leads to this disease
(No net loss or gain of genetic info!!)
-
Microarrays
-future technique for diagnosing cancers
-analyzing gene expression
-sequence person's genome, add RNA and see if it is highly expressed (red) or low expressed (green) or not (black)
acute vs. chronic leukemia
-chronic: overproduction of mature myeloid or lymphoid cell types; they are either post-mitotic (granulocytes, erythrocytes) or quiescent, rarely dividing ( mature B and T lymphocytes)

-acute: malignant cells are immature, not usually seen outside the bone marrow

*both have an excess of WBCs
How do you tell the difference between CML and infection?
-both cause leukocytosis and neutrophilia
-normally differentiated myeloid cells (just too many of them)

**CLONALITY - in CML, all malignant cells are descended from single progenitor that acquired the Philadelphia chrom translocation
How do you test for clonality?
-look at X inactivation patterns in women with X-linked polymorphisms
-only one active X in each somatic cell
-if you can distinguish paternal from maternal X chromosomes, and active from inactive X, can assess clonality

ex: philadelphia chromosome has clonality (same mutation) in all offspring
What are the 4 major features that blood cancers must acquire?
-autonomous proliferation
-insensitivity to negative regulatory signals
-increased survival
-limitless replicative potential

(for cancer in general, the other two features are angiogenesis and tissue invasion, but this is less important for blood cancer)
How does a tyrosine kinase mutation affect cells?
-gain a mutation in tyrosine kinase (ex: the JAK kinases are receptors with associated TKs) that causes increased or unregulated kinase activity in the absence of the growth factor stimulus

*provides autonomous proliferation and increased survival (normally cells are stimulated only by hematopoietic growth factors)

-ex: BCR-ABL --> CML
Mutation causes overproduction of maturing myeloid cells
What is the pathogenesis of acute leukemia?
-block the normal differentiation pattern of progenitors

*mutations in transcription factors that regulate genes involved in myeloid differentiation

==overgrowth of immature cells
What is the pathogenesis/how can you treat certain types of acute promyelocytic leukemia?
-subtype of AML
-t(15:17) causes a gene (often PML) to bind the retinoic acid receptor alpha (RARa) gene on 17

-normally RARa is a nuclear hormone receptor, binds retinoids **required for normal myeloid differentiation

*when fusion of the genes occurs, the fused gene binds the target genes and BLOCKS their transcription (instead of activating them)

*Treatment: Give high concentrations of all-trans retinoic acid (ATRA - vitamin A, an RARa ligand); the fusion protein binds the ligand instead of DNA, so the normal RARa protein can induce transcruption .. this allows leukemic blasts to continue differentiating, cause remission!
Can leukemia cells self renew?
-Yes .. this is a fundamental property that leukemic cells need (must be able to sustain proliferation indefinitely) BUT not ALL of them have this capability (heterogeneity)
What markers/properties do leukemia stem cells have?
-ex: in AML, only small subpopulation of leukemic cells are self renewing, or "leukemia-initiating"
-more quiescent and resistant to chemo, so may be responsible for RELAPSE following a remission

*CD34+ CD38-

Note: these also exist in solid tumors like breast cancer; probably only 10% of the population

(SCID leukemia initiating cell)
Remission
-no leukemic cells are detectable by pathology or cytogenetics
NOT the same as "cured"
Three properties of the acute leukemic cell
1) survival and proliferation
2) impaired differentiation
3) self-renewal (possibly via the Wnt signalling pathway)
Discussion review p. 276
leukemia/lymphoma symptoms
How does a tyrosine kinase mutation affect cells?
-gain a mutation in tyrosine kinase (ex: the JAK kinases are receptors with associated TKs) that causes increased or unregulated kinase activity in the absence of the growth factor stimulus

*provides autonomous proliferation and increased survival (normally cells are stimulated only by hematopoietic growth factors)

-ex: BCR-ABL --> CML
Mutation causes overproduction of maturing myeloid cells
What is the pathogenesis of acute leukemia?
-block the normal differentiation pattern of progenitors

*mutations in transcription factors that regulate genes involved in myeloid differentiation

==overgrowth of immature cells
What is the pathogenesis/how can you treat certain types of acute promyelocytic leukemia?
-subtype of AML
-t(15:17) causes a gene (often PML) to bind the retinoic acid receptor alpha (RARa) gene on 17

-normally RARa is a nuclear hormone receptor, binds retinoids **required for normal myeloid differentiation

*when fusion of the genes occurs, the fused gene binds the target genes and BLOCKS their transcription (instead of activating them)

*Treatment: Give high concentrations of all-trans retinoic acid (ATRA - vitamin A, an RARa ligand); the fusion protein binds the ligand instead of DNA, so the normal RARa protein can induce transcruption .. this allows leukemic blasts to continue differentiating, cause remission!
Can leukemia cells self renew?
-Yes .. this is a fundamental property that leukemic cells need (must be able to sustain proliferation indefinitely) BUT not ALL of them have this capability (heterogeneity)
What markers/properties do leukemia stem cells have?
-ex: in AML, only small subpopulation of leukemic cells are self renewing, or "leukemia-initiating"
-more quiescent and resistant to chemo, so may be responsible for RELAPSE following a remission

*CD34+ CD38-

Note: these also exist in solid tumors like breast cancer; probably only 10% of the population

(SCID leukemia initiating cell)
Remission
-no leukemic cells are detectable by pathology or cytogenetics
NOT the same as "cured"
Three properties of the acute leukemic cell
1) survival and proliferation
2) impaired differentiation
3) self-renewal (possibly via the Wnt signalling pathway)
Discussion review p. 276
leukemia/lymphoma symptoms
What is the most common cause of hematological cancer?
Non-hodgkin's lymphoma
-5th leading cause of cancer overall
-2nd fastest growing cancer
80% of lymphomas in US
Cell origin in NHL?
-85% B cells (remainder T cell ... very few NK cell)
What age group is affected most by NHL?
-elderly patients (older than 60)
Symptoms of NHL
-fevers, night sweats, weight loss, fatigue (constitutional systemic sx)
-swollen lymph nodes
-pain due to compression/infiltration of LNs (pressing on a nerve, etc)
-organ failure/compromise due to obstruction

**only with aggressive lymphomas (with indolent versions of NHL, often asymptomatic, tumor found incidentally)
What chromosomal abnormalities are involved in NHL?
proto-oncogenes translocated onto Ig heavy-chain locus at 14q32
-bcl-1 (MCL) - 11:14
-bcl-2 (FCL) - 14:18
-c-MYc (Burkitt's lymphoma) - 8:14
Median survival of NHL?
-ranges from 2.5 yrs (MCL) to 8 years (FCL)
Which cancer expresses CD-5 and cyclin D1?
-Mantle cell lymphoma (subtype of NHL)
-particularly poor prognosis
(morphologically, looks just like SLL)
Which cancer expresses CD-5 and CD-23?
Small lymphocytic lymphoma
Which cancer lacks CD10 (and CD5)?
Marginal zone lymphoma
Low Grade NHL
-Small lymphocytic
-Follicular, small
-Follicular mixed, small and large

*indolent
Intermediate Grade NHL
-Follicular large
-Diffuse small
-Diffuse mixed, small and large

*aggressive
High Grade NHL
-diffuse large, immunoblastic
-lymphoblastic
-small non-cleaved (Burkitt's)

**highly aggressive
Indolent types of NHL (4)
*1/2 of NHL tumors
-slow progression, median survival of up to 10 yrs

-follicular lymphoma
-marginal zone B cell lymphoma
-SLL
-lymphoplasmacytic lymphoma
Aggressive types of NHL (8)
**1/2 of NHL tumors (slightly more common)
-diffuse large B cell lymphoma
-mantle cell lymphoma
-peripheral T cell lymphoma
-primary mediastinal large B cell lymphoma
-anaplastic large cell lymphoma
-Lymphoblastic lymphoma
-Burkitt like lymphoma
-Burkitt's lymphoma
Composite lymphoma
-more than one subtype of lymphoma in a tumor
-often due to evolution of tumor from indolent to aggressive
NHL Diagnosis: Biopsy
-from lymph node, extramedullary mass, bone marrow

**fine needle aspiration NOT sufficient (although this is the case for most other malignancies) - use the open excision core needle biopsy which cuts out more tissue
NHL diagnosis: Histology
-some types are organized in groups similar to germinal lymph nodes b/c retained innate programming (architecture is nodular - MORE LIKELY TO BE INDOLENT)
-others are growing in sheets, diffuse - MORE LIKELY to be AGGRESSIVE

*can't see this architecture with a fine needle aspiration

-Also, look at cell size (small = indolent, less replication/transcription going on vs. large cells means more mitotic, etc = aggressive, less mature phenotype)
NHL diagnosis: Immunohistochemistry of T cells vs. B cells
-T cells have CD2,3,4,8,5

-B cells have 19,20,22,79a, Ig

-can also look for BCL2, BCL6 (regulate apoptosis) or Cyclin D (cycle checkpoint protein)
NHL Staging, I-IV and e
-I = single LN group
-II = two LN groups on same side of diaphragm (I and II further classified as bulky, if >10cm or 1/3 thoracic diameter; worse prognosis b/c blood supply doesn't reach center, can't bring drugs to kill it)
*stage I and II pts have limited disease, may just need radiation
-III = two+ LN groups on both sides of diaphragm
-IV - extramedullary disease (includes BM)
*these stages III and IV not amenable to radiation, mainstay is chemo

-"e" is added to any stage if there is just 1 isolated extramedullary node involved
NHL Staging, A or B
-A = patient is asymptomatic
-B = one or more of the following: fever > 38 without source, night sweats, or >10% weight loss
-B has worse prognosis
-classify in addition to I - IV
What is the imaging study of choice for NHL?
-CT scan of chest, abdomen and pelvis
When would you do a bone marrow biopsy for NHL?
-to check for stage IV extramedullary disease during diagnosis phase
International Prognostic Index
-predicts outcome, high and low risk for death

-Age>60
-Performance status of 2 or worse (pt. can't get through day without napping)
-LDH elevated
-2+ extranodal sites (bone marrow counts too)
-Stage (I,II vs. III,IV)

**DOES A BETTER JOB WITH HIGH RISK PATIENTS
Revision: Follicular Lymphoma IPI
-determined that Hgb of <12 and number of total nodal sites >5 were more important factors than performance status and extranodal sites

**DOES A BETTER JOB WITH VERY LOW RISK PATIENTS
What is the goal of treatment for an indolent NHL?
-palliative; grows slowly but cannot be cured!!
-symptom control and Q of life
"WATCH AND WAIT" - only treat if SHOWING symptoms, can only make it worse by treating a patient who has no sx
-eventually all patients die over about 10 years
What is the goal of treatment for an aggressive NHL?
-cure!
-however, get sick very quickly, high burden of disease
*those who survive the first couple of years are likely to be cured, rarely die of lymphoma (attributed to complications from aggressive therapy, and the aggressive tumor itself)
**Begin treatment promptly!!
What is the role of surgery in treating NHL?
-rarely used, no significant role b/c usually tumor has already dispersed, surgery will only delay time until medical treatment

-respond much more quickly to chemo and radiation

**DO use surgical tx to relieve obstruction and other complications by the tumor!!
What is the role of radiation in treating NHL?
-very effective and maybe curative in patients with limited disease
-can be treated without exposing too much normal tissue
**good for discrete problems like a bone lesion or airway compromise (that way you dont need to affect whole body by giving chemo)
What is the role of chemo in treating NHL?
**mainstay of NHL treatment
-alkylators, anthracyclines, and high doses of steroids
-single doses
-combinations shown to be very effectives
*cure can be achieved by high dose combination therapy + stem cell transplant
Rituximab and other new treatments
-monoclonal Ab against lymphoma antigens

(2 others that are also linked to radioactive isotopes to kill lymphoma)

-Ontak (IL2 agonist bound to diptheria toxin) and bortezemib (proteosome inhibitor) are other MoAbs

*attempting to use anti-idiotype, dendritic cells as vaccines to teach patient cells to destroy their own lymphoma
How do you treat indolent NHL lymphoma that has disseminated?
WATCH AND WAIT is preferred if no symptoms

-if sx, can do single agent chemo or XRT; can do a chemo combo with anthracycline
-transplantation
-antibody-based therapy
What is the MOA for rituximab?
-attacks CD20 via ADCC, complement-dependent cytotoxicity, and apoptosis
-Fab binds to CD20
-Fc portion recruits immune effector functions that trigger B cell lysis
What is the effect of rituximab on survival/response?
-dramatic improvement IF COMBINED WITH CHEMOTHERAPY
-high rate of complete response, doubles time to relapse
*if continued after chemo, prolongs time to recurrence even if it is discontinued 2 years after

**NOT USEFUL FOR INDOLENT LYMPHOMA
What is the MOA for radioimmunotherapy (MoAbs linked to radionuclides)?
-crossfire effect!!

-Radiolabeled MoAbs deliver low dose, site specific radiation to lymphoma cells
-lymphoma cells w/ low Ab binding are killed by radiation from the labeled MoAbs bound to ADJACENT (crossfire) tumor cells

**great for indolent lymphoma, but expensive (similar results to chemo+rituximab)
How do you treat indolent NHL lymphoma that has is localized?
WATCH AND WAIT if no sx
-use just XRT
-or 3-4 cycles chemo + XRT
-or 6-8 cycles chemo
-immunotherapy

**Note: this type of disease is rare; by the time it presents, it has usually disseminated
How do you treat aggressive NHL lymphoma that has is localized?
-determined by who can tolerate tx

-3-4 cycles chemo+XRT
-or 6-8 cycles chemotherapy (if tolerated, sufficient if not over-treatment of disease)

-or XRT (rare)
Which is better for NHL, chemo or chemo+radiation?
-about comparable
-depends on risks of each for particular patient
How do you treat aggressive, disseminated NHL lymphoma ?
-stage III and IV

-6 months of chemo, systemic

-if young with higher risk of relapse, try high dose chemo and stem cell transplant

*older patients who can't tolerate chemo can receive radiation aimed at relieving symptoms (localized palliation)
What is CHOP?
-cyclophosphamide
-doxorubicin
-vincristine
-prednisone

*most successful, least toxic chemo regimen for NHL!!
Review cases
p.293
What is SVC syndrome and what causes it?
-tumor presses on SVC and blocks outflow
-pt gets face and neck swelling
-tumors of mediastinum = Terrible T's (teratoma, thymoma, testicular/germ cell tumor, thyroid tumor, terrible lymphoma)
What are the symptoms of Hodgkin's Lymphoma?
VERY similar to NHL, although B symptoms are more prominent in HL
-Progressive lymphadenopathy (usually in neck or mediastinum
-night sweats, fevers, weight loss, Pel-Ebstein fevers
-pruritis
**occasionally, pain at the site of tumor with ingestion of alcohol
How does HL spread, and how fast?
-slow, orderly growth
-sequential fashion from one lymph node group to the next
How does this differ from NHL?
-NHL can spread to and from any LN at any time
How common is HL?
-6% of new hematologic malignancies
-far less common than HL
What ages does HL strike?
-BIMODAL
-early 20s
-late 60s
Name 3 of the epidemiologic risk factors for HL
-high socio-economic status (sheltered kids not exposed to germs enough, overprimed immune system)
-immunosuppression (HIV, transplant) - can't recognize cells with viral antigens/chronic infection
-genetics
What virus is associated with HL and why?
-Epstein Barr
-2 proteins coded by EBV genome mimic B cell surface proteins
-when expressed on the cell surface, provide activation (viral LMP1 protein) and survival signals (viral LMP2 protein)
-activation keeps a B cell alive and causes a nonproductive Ig rearrangement; once the Ig is expressed, the survival signal keeps the Ig expressed (normally, B cells that don't make a productive Ig are apoptosed)

**3-4 fold increased risk of getting HL for 2 years after EBV infection
How do you diagnose HL histologically?
-Reed Sternberg cells
What is a Reed Sternberg cell?
-CD30+, CD15+, eosinophil
(smiley face cell!)
*CD20- ... this differentiates HD from NHL which is CD20+!!

-Malignant B lymphocyte with an IgH rearrangement
-multinucleated giant cells that make up a small proportion of the total cells (rest of the B lymphocytes are normal, active)
-much larger than normal lymphocytes
What type of biopsy would you do for HL?
open biopsy
-need a large piece of tissue because RS cells are uncommon
-FNA is not sufficient ever
Which stage of B cell is overproduced in classical HL?
-Mature B cells from the germinal center that have undergone IgH rearrangement
What are the 4 types of Classical HL?
-Nodular sclerosis (most common)
-Lymphocyte rich (this has overlap with NHL, but unlike NHL the lymphocytes are polyclonal and reactive)
-Mixed Cellularity
-Lymphocyte depleted (rare, aggressive)

**treatment is the SAME for all types
What is the most common presentation of nodular sclerosis, and what is it?
-younger patients, especially women with mediastinal mass
-has the RS cells
-fibrosis surrounding the nodular lymphoid structure
What does mixed cellularity type look like?
-2nd most common type
-RS cells + inflammatory cells (B and T cells, plasma cells, eosinophils)
*older, male patients
What is the 1 subtype of Non-classical HL?
-Nodular lymphocyte predominance
-young men, neck or groin

*expresses CD20, NOT CD30 or CD15!
-BUT these are still RS cells
-responds to therapy, but recurs, exactly like indolent forms of NHL
How do you stage HL?
-Ann Arbor system
-same criteria as NHL
-A/B indicates whether the B symptoms are present
-M = bulky (worse prognosis)
-E = extranodal involvement (infiltration of another organ) ..all of these are also used to describe NHL

ex: IIIB, IAE, IVAE
Why do you image with PET or Gallium for NHL and HL?
-to see if there is occult disease on the other side of the diaphragm

(in addition to phys. exam, CT scan and BM biopsy)
What is the goal of tx for classical HL?
-aggressive
*Goal: cure (unless they cannot tolerate therapy, then aim for palliation)
How do you treat limited stage (I,II), non bulky HL?
-radiation alone or short course chemo (3-4 cycles) + reduced dose radiation, or chemo alone
How do you treat extensive stage (III,IV) HL?
-chemo .. not really amenable to XRT if mass is >5cm
Types of Radiation: Mantle
-the part a belly shirt would cover on your body
-targets the involved lymph nodes and the next set of contiguous nodes (anticipates pattern of spread)
Types of Radiation: Extended Mantle
-adds in the para-aortic nodes and the spleen
(covers the rest of the stomach)
Types of Radiation: Sub-total nodal
-adds in the part shorts would cover, kills in all lymph nodes but very toxic
What is the Chemotherapy regimen for HL? (4)
ABVD
-adriamycin = cardiotoxic
-bleomycin = pulmonary toxic
-vinblastine = BM toxicity
-dacarbizine = BM toxicity

*better than the old MOPP regimen (nitrogen mustard, risk of leukemia and infertility)
What if treatment doesn't bring about a cure by 15 years?
-more likely for pts to die from therapy than from disease, or from 2ndary effects of the therapy (leukemia, **breast cancer in young women with HL, etc)
-must balance efficacy vs. toxicity
What are the long term complications of radiation and chemo?
-Radiation: secondary malignancies, esp. breast and lung; cardiac disease; pulmonary fibrosis

-Chemo: secondary leukemia and infertility
What are 5 factors that predict for treatment failure with local XRT therapy?
-presence of B symptoms
-extranodal disease
-bulky disease
-3+ sites of nodal involvement
-SED rate >50

*if any one of these factors is present, and pt failed XRT therapy, use chemo as primary therapy
Treatment for early stage, favorable disease for HL?
-I and IIA
-most are cured
-extended field radiation or 2-4 cycles chemo plus involved field XRT (improves relapse and survival)
-for younger patients, XRT has long term risks, better to do chemo alone
-in elderly, XRT alone may work and only requires 1 month treatment
Treatment for early stage, unfavorable disease for HL?
-I and IIB
-same cure rate
-4 cycles chemo + involved field XRT, or 4-6 cycles chemo
International Prognostic Score for HL
-stages III and IV only!!
-age>45
-WBC>15000
-Albumin < normal
-Lymphocyte count <8%
-Hgb <10.5
-male
-stage IV

*0-2 = low risk, 3+ = high risk, but survival still good (still 55% vs. NHL high risk where survival is only 20%)
How do you treat advanced stage, favorable disease?
-Stages III and IVA
-75% cure
-6-8 cycles chemo
-or 6-8 cycles + involved field XRT to bulky disease
How do you treat advanced stage, unfavorable disease?
-III and IV B
-50-65% cure
-6-8 cycles of ABVD, or clinical trial
Review cases
p.309
What is acute leukemia and what is the the typical presentation?
-malignant accumulation of transformed hematopoietic progenitor cells
-clonal expansion of leukemic blasts that are self-renewing but unable to differentiate
-when it infiltrates the bone marrow, impairs normal hematopoiesis leading to BM failure

**Patients usually present with symptoms of cytopenias (anemia, bleeding, infection, etc)
What are the epidemiologic differences between AML and ALL?
-AML: incidence increases with age, median age 65, only 20% in kids
-ALL: slightly less common, but incidence DECREASES with age, median age 3-5 yrs; 75% occurs under age 15; genetics plays a role

**Note: both are slightly more likely in men, and risk is increased in ppl having chromosomal abnormalities (Down's, Klinefelter's) or other hematologic disorders like MDS or CML
What is the most common childhood cancer?
ALL
What are some causes of acute leukemia? (3)
-ionizing radiation, AML 5-7 yrs later, or ALL with prenatal exposure
-benzene/chemicals (also cause MDS and aplastic anemia) .. smoking, petroleum, paints, embalming fluids, herbicides, pesticides increase risk of AML
-anti-cancer drugs = therapy related AML (alkylating agents cause defects in chrom. 5 and 7; topo II inhibitors cause defect in 11); radiation+chemo increases risk further

**Note: most patients have no known exposure or predisposing condition
What are the symptoms of acute leukemia?
-non specific constitutional sx
-bone marrow failure
-present for weeks to months (fatigue, weakness from either anemia or hypermetabolic statet, anorexia, weight loss, fever with no obvious infection; infections may occur, or bleeding/bruising)
-if bone marrow is infiltrated, may have bone pain
What is leukostasis?
-while some patients present with leukopenia and have few blasts in peripheral blood, patients can also present with leukocytosis resulting in stasis of blood flow in pulmonary and cerebral circulation
-blast count above 50,000

cerebral leukostasis = headache, visual disturbance, confusion, progresses to coma/stroke

pulmonary leukostasis = dyspnea, tachypnea, crackles on physical exam, infiltrates

**Much more common in AML b/c blasts are larger and have adhesion molecules
How does DIC relate to acute leukemia?
-can be caused by chemotherapy
-more common in AML, especially in APL (acute promyelocytic leukemia)
-can cause intracranial or GI hemorrhage) .. elevated PT, PTT, low fibrinogen
What are manifestations of extramedullary leukemia in ALL?
-lymphadenopathy
-splenomegaly
*infiltration of lymphoid organs

-in T cell ALL, can present with large mediastinal mass

-can also have leukemic meningitis in relapse (or testicular involvement)
What are manifestations of extramedullary leukemia in AML?
-leukemia cutis (skin infiltration)
-gingival hypertrophy

-rarely develop solid tumors w/ no infolvement of bone marrow or blood = chloromas (blast tumors, respond well to radiation and chemo combo)
What is tumor lysis syndrome?
-before or during treatment, especially in ALL due to cell death
-hyperuricemia leads to nephropathy
-hyperphosphatemia

..cause renal failure and hyperkalemia
What are the metabolic abnormalities of AML? other general metabolic problems of the two?
-hypokalemia due to renal tubular injury by the lysozyme released from myeloid blasts

-in ALL especially, LDH elevaeed
-both types may have hepatic enzymes elevated, or artificial hypoglycemia after phlebotomy draw
What is the French-American British criteria for diagnosing leukemia? vs. WHO's criteria?
>30% blasts in bone marrow

WHO says only 20% needed
AML/M0
Myeloblastic leukemia without differentiation

-CD13 and 33+
-peroxidase negative
-mostly cytoplasm, nongranular
AML/ML
Myeloblastic leukemia without maturation

-Auer rods
-1+ nucleoli
-mostly cytoplasm, nongranular
What are Auer rods?
-linear aggregates of primary granules in MYELOBLASTS only
AML/M2
Myeloblastic leukemia with maturation

*all stages of granulocytes present; granular blasts

*t(8,21)
**AML/M3
Promyelocytic leukemia
**MOST IMPORTANT TO RECOGNIZE because it has UNIQUE TX

*bilobed nucleus
-abnormal promyelocytes
-Auer rods in BUNDLES
-large granules
-can cause DIC

*t(15,17)
AML/M4
-Myelomonocytic leukemia
-mostly promonocytes

*inv16
AML/M5
Monocytic leukemia

-poorly differentiated vs. differentiated
-large blasts, rare granules
*extra hematopoietic manifestations
AML/M6
-Erythroleukemia

>50% erythroblasts in bone marrow
AML/M7
Megakaryocytic leukemia
ALL/L1
-small, homogeneous cells

-round nuclei, fine chromatin, no nucleoli
-little cytoplasm
ALL/L2
-large, heterogeneous cells

-folded nucleus
-coarse chromatin
-1+ nucleoli
-moderate cytoplasm

*same tx as ALL/L1
*ALL/L3
**BURKITT's LYMPHOMA .. dif tx than ALL 1 and 2

-large, homogeneous cells
-oval or round nuclei
-dense chromatin
-abundant cytoplasm
What are the histochemical characteristics of AML?
-myeloperoxidase (in primary granules of granulocytic precursors)
-lysozyme

-non specific esterase in the monocytic subtype (M5)
What is a histochemical characteristic of ALL?
-terminal deoxynucelotidyl transferase (TdT) positive

-expressed in nucleus of lymphoblasts

**Note: lymphoblast subtypes also have more nucleus than cytoplasm, nucleoli not as prominent
What are the flow cytometry (immunophenotyping) results for AML? ALL?
-AML - CD13, CD33, CD117

-ALL - most are B precursor cell type = CD10, CD19, CD22; those of precursor T type have 1a,2,3,4,5,7,8

**BUT 5% of ALL has myeloid markers CD13 and CD33
What is the WHO classification system?
-4 subtypes of AML based on cytogenetics

AML with genetic abnormalities (see p. 323)
AML with multilineage dysplasia
AML, therapy related
AML, not otherwise categorized

-ALL uses immunophenotyping
Precursor B cell ALL
Precursor T cell ALL

(Burkitt's is classified as a lymphoma, not with this system for the WHO)
Can you treat acute leukemia with blood transfusions?
-yes - required for effective treatment
-for RBC transfusion, depends on patient's status (younger patients tolerate a lower Hgb level)

-for platelets, transfuse if fewer than 10,000/uL platelets (prophylactically for bleeding)

*make sure you give leukodepleted blood products to prevent CMV transmission, decrease risk of alloimmunization (Abs against transfused platelets); irradiate blood products to prevent GvHD especially in BMT
How do you treat leukostasis?
medical emergency!
-IV hydration and leukapheresis to reduce blast count
-hydroxyurea
-steroids if ALL
-diagnose acute leukemia, then start chemo

**avoid RBC transfusion in any leukocytotic patient because it will raise blood viscosity, can start leukostasis
What is hydroxyurea?
ribonucleotide reductase inhibitor
-can treat leukostasis by reducing blast burden
How do you manage DIC?
-replace clotting factors with FFP
-replace fibrinogen with cryoprecipitate
-platelet transfusion
How do you manage tumor lysis syndrome?
-IV hydration for diuresis
-urine alkalinization - adding bicarbonate to hydration
-allopurinol

*renal failure may still develop - give hemodialysis if this occurs
Bicarbonate role?
increases solubility of uric acid, prevents urate nephropathy in tumor lysis syndrome
Allopurinol role?
-prevents accumulation of uric acid in tumor lysis syndrome
How do you manage infection in pts with acute leukemia?
-prophylax with antibiotics, anti-fungal, anti-viral meds
-all acute leukemia patients are at risk
-sepsis, sinusitis, bronchitis, pneumonia, UTI

*be sure to look for infection any time patient presents with a fever (though it could be due to the leukemia itself)
What are some prognostic factors in AML?
-advanced age (over 60) is negative
-some cytogenetic abnormalities have favorable prognosis, or can be unfavorable; those with no abnormalities are at intermediate phenotype
-patients with an MDS or MPS or aplastic anemia disorder that develop AML (secondary leukemia) have poor prognosis
-therapy related AML from alkylating agents or topo II inhibitors have unfavorable prognosis
Which cytogenetic abnormalities have a favorable prognosis?
-genes affecting Core Binding Factor transcription
ex: t(8;21), an AML1-ETO gene fusion inAML/M2
also inv16, an AML1 smooth muscle myosin heavy chain fusion .. AML/M4

these patients have CBF AML and have a 90% prob of achieving remission

**the t(15,17) in APL M3 subtype is from a fusion of the PML gene with RARa gene on 17 .. can be treted, 85% remission
Which cytogenetic abnormalities have an unfavorable prognosis?
-del5, del7 or monosomy5 or 7 (associated with alkylating agents)
-deletions or translocations in 11q23 (associated with use of topoII inhibitors) .. what subtypes??
-inv(3)

OR multiple abnormalities
How do you treat AML?
-induction chemo to get <5% blasts in the bone marrow, no blasts in periphery, and restoration of normal hematopoiesis (remission); use 3+7 regimen

-next, do consolidation therapy to prevent relapse; eradicate residual cells; induction lowers blasts 1 log from original 10^12
-2-4 monthly cycles of 2+5, or high dose cytarabine alone; can go into long term remission; can also do an intensified version

**Failure to achieve remission, or a relapse after remission = poor prognosis

-40-70% in long term remission = Cure!
3+7 regimen
-3 days anthracycline plus cytarabine for 7 days
-standard induction therapy

(but high %age of death from toxicity - WATCH FOR INFECTION, myelosuppression lasts 21-28 days)
2+5 regimen
-anthracycline 2 days
-cytarabine 5 days
What is the intensified post remission therapy?
-in place of regular consolidation therapy
-cures more patients
-either high dose cytarabine, HSCT (autologous) or HSCT (allogeneic - therapy of choice with unfavorable disease)

**Note: HSCT (allogeneic) can salvage some patients who failed other regimens
How do you treat APL?
-acute promyelocytic leukemia often present with leukopenia and DIC
-median age is 30-40 years
-blasts have primary granules and Auer rods
**t(15,17) cytogenetic abnormality (PML-RARa fusion)

*treat with ATRA!! induces complete remission in 85% w/o blast lysis or marrow aplasia seen in chemo .. simply induces differentiation
Complication of ATRA?
Capillary leak syndrome/APL differentiation syndrome
- weight gain, edema, fever, hypoxia, pulmonary infiltrates
-few days to weeks of ATRA initiation
-due to a rapidly rising WBC count

**TREAT WITH STEROIDS or else rapidly fatal
-incidence is decreased by giving ATRA with anthracycline
Arsenic Trioxide
-can also be used to treat AP
-ATO causes apoptosis in APL cells with 85% remission

*QT prolongation, AV block and toursade de pointes; can also cause APL differentiation syndrome

**use anthracycline+ATRA as induction, maintain with ATRA and an oral anti-metabolite; if relapse, THEN use ATO
What are the prognostic factors in ALL?
-excellent prognosis in kids, poor if >60
*infants also have a poor prognosis
-if philadelphia chromosome present, poor prognosis (more common in adults)
-if MLL gene of 11q23 involved, high rate of relapse (seen in infants and adults, poor prognosis)
-t(12,21) from fusion of TEL and AML1 genes has excellent prognosis, usually seen in kids

*precursor B cell ALL has better prognosis than precursor T cell

-if high WBC count, failure to achieve copmlete remission in 1 month, high LDH are all poor prognostic factors
Treatment for ALL?
-kids usually cured with standard chemo; adults have less response
-anti-metabolites, vincristine, cytarabine, cyclophosphamide, L-asparaginease, etoposide, glucocorticoids

-2-3 years
Stages of ALL treatment
-induction therapy with 4-5 drugs (anthracycline, vincristine, glucocorticoids)

-intensification: need to target CNS, which is a great place for leukemic blasts to live .. likely site of relapse! Prophylax with intrathecal administration of cytarabine and methotrexate with or without cranial irradiation

-Maintenance - extends disease free survival esp in kids, high relapse in adults: give daily oral plus weekly chemo for 2 yrs
What about patients with high risk for relapse?
-cytogenetic abnormalities, failure to achieve remission

-allogeneic HSCT! especially if a sibling matches as a donor
What is the most common leukemia in the western world?
CLL
-long median survival, so there is a high prevalence
What is the median age of diagnosis for CLL?
-60
What is CLL?
-too many mature B cells (naive B lymphocytes, low density Ig)
-smudge cells
What are smudge cells?
-seen in CLL
-cells have fragile membranes, often rupture during slide preparations
-rupture during flow through cell counting machine, difficult to get a count
How do you diagnose CLL?
-lymphocytosis >5000 / uL
-30% lymphocytes in BM
-<50% immature lymphocytes in peripheral blood (> would instead lead to dx of prolymphocytic leukemia diagnosis)
-clonal expansion of abnormal B cells

*Note: some ppl have CLL cells under 5000 count (analogous to MGUS) but never get CLL
What is expressed on CLL cells?
-CD19,20,24

**CD5 is the ONLY T cell marker expressed on the CLL B cells
-B cell is monoclonal, either kappa or lambda light chains
-low density Ig (IgD?)
How does CLL progress?
-asymptomatic at diagnosis
-indolent
-incidental diagnosis
-may have enlarged nodes or anemia
-progresses to involve BM, HSM, increased infections/anemia/thrombocytopenia, hypogammaglobulinemia, autoimmune complications (hemolytic anemia, TTP due to impairment in regulator T cell function influenced by CLL)

*in least aggressive form, patients die of other causes first, finish normal length of life

*in the long term, can evolve to a more aggressive disease, similar to diffuse large B-cell lymphoma
What are CLL patients most likely to die from (CLL-related)?
INFECTION

(CLL suppresses normal antibody production leading to infection especially with encapsulated organisms)
SLL
-small lymphocytic leukemia
-just like CLL but the disease is in the LN instead of the blood
-become the same disease as CLL in the long term, outcome is the same
What are the autoimmune complications of CLL?
-AIHA (coombs positive)
-pure red cell aplasia (Coombs negative, less common, deficiency of red blood cell precursors)
-immune thrombocytopenic purpura
-neutropenia
**Note: other autoimmune diseases are uncommon
Where would you find normal CD5+ B cells?
-edge of germinal centers in the mantle zone of lymphoid follicles

-produce polyreactive, low affinity autoantibodies
(this is involved in autoimmune Abs because they interact with malignant cells and T cells to produce polyclonal Abs not made by malignant clone)
Long term complications of CLL
-severe systemic infections
-bleeding
-Richter's transformation
-prolymphocytoid transformation
-second malignancies
-AML (usually due to treatment, not CLL itself)
What is Richter's transformation?
-most common complication of CLL
-CLL evolves into diffuse large cell immunoblastic lymphoma
-poor survival
What is prolymphocytoid transformation?
-CLL complication .. changes to larger cells with nucleoli, less dense chromatin

*refractory to typical chemo
How do you treat CLL?
-Watchful waiting
-palliation
-oral chemo
-cingle agent, combo
-high dose chemo and autoSCT
-allogeneic HSCT

*depends on aggressiveness and patient age b/c CLL is an indolent disease
What is the Rai and Binet classifications?
-staging systems for CLL
-correlate with patient survival
-Rai: stage 0 is at low risk (only in blood, bone marrow), I/II intermediate (lymphoid organ enlargement), III/IV high (hematopoietic compromise)

-since most patients are asymptomatic at diagnosis, this system is less useful b/c puts most pt at low risk
2 Versions that cause CLL (2)
-1) naive B cell (has not yet recognized antigen but has rearranged Ig) has oncogenic event and transforms to a CLL cell, has not undergone SHM so it is "unmutated" because it is the same as the original germ cell line

2) naive B-cell encounters antigen in the lymph node, activates, undergoes HSM and THEN gets an oncogeneic hit, becomes CLL cell; significant changes from germ cell line, therefore it is "mutated"
Which is better, mutated or unmutated CLL?
-mutated is better!!
-only 9 year survival if unmutated
Are CD38+ cells good or bad in CLL?
-patients with >30% CD38+ cells do poorly, similarly to the unmutated group

**note: even if poor risk groups, still live a long time, only limiting on life if the patient is young
What is the role of Zap 70?
-zeta assoc protein
-normally expressed in NK and T cells, associated with the CD3 zeta chain of TCR complex
-tyrosine kinase

**if expressed in CLL, worse prognosis!
Which abnormalities are associated with CLL? (seen by FISH)
-del 17p or 11q have an aggressive phenotype, poor prognosis

-del 13q has a better prognosis and longer survival

*no cytogenetic abnormality has intermediate prognosis
How should you treat CLL?
-so indolent that often treatment is not needed early on .. watch and wait until they are symptomatic

**treat with chlorambucil, an alkylator
*fludarabine is newer, more effective .. BOTH can be used as single agent treatment

*Rituximab works great but ONLY in combination with chemo!! doesn't significantly change toxicity .. MUST be given at same time as chemo for synergy <<THIS IS THE BEST TREATMENT
Alemtuzumab
-MoAb against CD52 (on all lymphoid cells)

*used for relapse in CLL

-has "compartmentalization" - doesn't work for all parts of body! great for blood and bone marrow, but not for lymph node disease

-not well tolerated, chills and sweats, fevers, myelosuppression
-affects both B and T cells makes pts especially immunosuppressed; can cause reactivation of CMV, other infections

-may be good for del17p patients
When to give high dose chemo, autologous HSCT?
-refractory or relapsed CLL patients
CLL vs. CML
CLL - proliferation of mature lymphocytes, multiple cytogenetic abnormalities involved, long survival with rare leukemic transformation, no available targeted therapy yet

CML - proliferation of maturing myeloid cells; only caused by the t(9,22) abnormality; has a 25% PER YEAR risk of AML transformation, but DOES have a targeted therapy (imatinib and TKIs against BCR-Abl fusion)
Review case
-p.350
What are plasma cell dyscrasias?
-clonal proliferation of abnormal mature plasma cells
-usually produce Ig
-blue cytoplasm, clock face eccentric nucleus

-Monoclonal Gammopathy of Undetermined Significance (MGUS)
-Multiple Myeloma
-Plasma Cell Leukemia
-Waldenstroms Macroglobulinemia
-Heavy Chain Disease
-Cryoglobulinemia
Immunoglobulin Structure
-2 light chains identical, 2 heavy chains identical, disulfide bonds
-each from recombination of v,d,j families; go through splicing to make variable regions of Ig for antigen specificity
What is the purpose of the Fc region? What are the different classes of Ig?
-encodes class of Ab (IgA,M,D, E, G)
-IgA most abundant, but usually secreted into GI
-IgM has high avidity, responsible for early immune response
-IgD binds B cell membrane and is an antigen receptor
-IgE activates mast cells and basophils
-IgG is most abundant in blood circulation, mediates mature immune response to infection (binds complement, recognized by effector cells for ADCC)
What is an M spike?
-M = monoclonal

-an abnormal peak on an electrophoresis created by a plasma cell malignancy (secretes the same Ig)

-different from the classic pattern of distrib into 1 of 5 regions (alpha1,2,beta,gamma,albumin)
Immunofixation
-staining proteins separated by electropheresis to MoAbs against different Ig classes, and against kappa/lambda light chains

-note: IgG,M,A most common

*can also see an M spike on this analysis and can identify the type of Ig and whether it's kappa or lambda light chain
What is MGUS?
-monoclonal gammopathy of unknown significance
*most common plasma cell dyscrasia (2% of population over 50!)
-associated with autoimmune disease (immune response not shut off after response to infection, antibody production continues)

*incidental diagnosis
-prognosis is good, 1%/yr progression
How do you diagnose MGUS?
-routine lab
-total protein exceeds albumin by a lot
-monoclonal Ig is present but not more than 3g/dl and <10% plasma cells in bone marrow
**If exceeds these criteria, = Multiple Myeloma!

Globulin protein = Total protein - albumin (so Total protein = globulin + albumin) therefore, if total protein highly exceeds albumin, the total protein is mostly globulin
Treatment for MGUS?
-just watch
-repeat the SPEP every 6-12 months
Most common emergent hematologic malignancy from MGUS?
Multiple myeloma
-can progress to the other PCDs
Multiple Myeloma: Epidemiology
-relatively uncommon (1% of all cancers, 10% of heme cancer)

-incidence rising slowly; pts living longer
*median age 65
-more common in African Americans and males; less in Asians
Causes of Multiple Myeloma
-progression from MGUS (not the majority)
-occupational exposure to radiation (uranium mines)
-chemicals used by sheet metal workers
-benzene

*genetics and environment also
What is multiple myeloma?
-clonal proliferation of terminally differentiated, Ig secreting plasma cells; it is a post-germinal center B-cell that have the ability to mature to plasma cells
-can switch Ig class, because it is not yet committed to a particular class
Where does MM live and why?
-rarely outsid ethe bone marrow
*out of the heme cancers, relies most on its environment due to its adhesion molecules
-make physical contact with bone marrow stroma; contact induces antiapoptotic signals
What is the role of cytokines in MM?
-IL-6, etc. are released and play a role in MM survival, provide survival signals
-also activate osteoclast which causes bone resorption after MM cells adhere to the bone marrow
What is the role of viruses in MM?
viral infections like HHV-8 that secrete an IL-6 like cytokine
-mutations in oncogenes also play a role (RB, p53, Ras, bcl-2, c-Myc)
How does the M protein affect the other immunoglobulins?
-monoclonal immunoglobulin is often in the serum and can spill out into the urine
-serves as negative feedback on normal plasma cells, causes overall Ig to fall, even the types of MM that don't secrete Abs and have no detectable protein (this type probably has mutation in Ig gene so they can't produce M protein, but still have same sx)
Multiple myeloma presentation
-bone marrow suppression sx (anemia from cytokine mediated suppression or from crowding out of BM, infection - may need Ig infusion)
-w/ or w/o lytic bone lesions (80%)***hallmark .. may also have bone pain or fractures
-hypercalcemia (causes constipation, lethargy, weakness, confusion .. due to increased osteoclasts)
-renal failure (from hypercalcemia+dehydration, or light chains getting trapped in the kidney causing abnormal accumulation of protein - amyloidosis, or light chains themselves are toxic to collecting tubules and cause irreversible damage)
-hyperviscosity (due to high levels of Ig in serum, slows blood flow and O2, increases risk of arterial thrombosis)
-amyloidosis (accumulation of protein due to tropism of M protein for nervous tissue)
-neurologic (autoimmune destruction of nervous system tissue)
What do the MM bone lesions look like?
"punched out"
-no reactive changes that indicate repair
-just lytic lesions from osteoclast activation
Which 2 tests are acceptable in MM? Which test would you not perform?
-plain xrays or MRI are good

**Bone scans show NO abnormality, not a good test!
How do you diagnose MM?
-imaging
-must meet two major criteria, or certain combinations of major and minor criteria (major have to do with too many plasma cells) .. see p.377

-can also do Serum Protein Electrophoresis (SPEP) or 24hr urine protein electrophoresis (UPEP)
-bone marrow aspirate and biopsy
-quantify Igs
-skeletal survey (plain xrays)
Plasmocytoma
-major criterion for MM
-can be seen in a biopsy from a pathologic fracture, or from an extramedullary site (most commonly in the nasopharynx)

-collection of malignant plasma cells
What is the staging system for MM?
-Durie/Salmon

-presence vs. absence of:
anemia
hypercalcemia
lytic bone lesions
amount of M protein
presence or absence of renail failure

*if all absent, stage I; if grossly abnormal, III, in middle is II

*if renal failure, add a B; if absent, add an A

**NO STAGE IV

Note: in D/S, IB is worse than IIIA (unlike with TNM system where IIIA is worse than IIB)

Now use a new ISS prognostic scoring system like the IPI using just two parameters
Treatment of MM
-always fatal if untreated (<1 yr survival)

-with chemo, survival is about 3 years
-traditional therapy is melphalan and prednisone
-autologous SC rescue with high dose mephalan wipes out patients BM permanently and increases survival to 5 years; only use if <70 yrs old
What is the role of a second transplant in MM?
-two transplants are better than 1 but works the best in patients who had an INCOMPLETE response to the first transplant
What is the role of allogeneic SCT in MM?
-very toxic with chemo and radiation .. not currently used
New potential drug for MM:
-MM has ability for angiogenesis because the tumor lives in the bone marrow, so can treat with thalidomide (anti-angiogenic); however, this is only a small portion of MM's abilities, and there are many thalidomide mechanisms, so doesn't work for all patients

-Best in combination with steroids for treatment naive patients (though it does work in some heavily pre-treated pts)

*significant side effects (sedation, periph neuropathy, DVTs)

Lenalidamide is a analog of thalidomide, but works more like chemo; has more of the immunomodulatory effects of thalidomide on cytokine signalling and immune stimulation
-fewer side effects
-20-30% efficacy with steroids
What about chemo+ thalidomide?
-very potent for MM, but toxicity is significant
-many subjects died of PE

"MPT" - mephalan, prednisone, thalidomide

Note: if administer anticoagulants, the toxicity is minimized
Do proteosome inhibitors work in MM?
-bortezemib has activity in MM
-causes apoptosis by preventing cell cycle progression
-MM cells are influenced by NFKb, a promoter of cell growth; it complexes with IKb which is regulated by the ubiquitin pathway

-inhibiting that pathway by stopping the proteosome makes Ikb accumulate which then inactivates NFkb, so MM cells can't grow, apoptose
Outpatient therapy for MM
-healthy patients under 70 (no comorbidity) ca nget thalidomide and dexamethasone orally for induction, then considered for one round of high dose chemo and autologous SCT (with a 2nd one if they don't respond)
What about MM patients over 70 or with comorbid conditions?
-not candidates for transplant, but can still give the MPT combo

-lenalidomide and bortezomib are also obtions (just more expensive, and bortezomib is IV whereas thal and lena are oral)
What is Plasma Cell leukemia?
-unusual subtype of MM
-high $ of malignant plasma cells circulating in blood (same as for MM)
-two forms: primary and secondary
-survival is short in both cases
Primary PCL
-no preceding diagnosis of MM (5%)
-emerge with ability to travel in bloodstream
-bias towards IgA
-patients present with extensive lytic bone lesions
-same tx as with MM (transplant, etc..leukemia tx don't work)
Secondary PCL
-end state of heavily treated MM
-can't travel in bloodstream until much later (acquire mutations over time)
Waldenstrom's Macroglobulinemia
-Plasma cell disorder
-Lymphoplasmacytic lymphoma
-malignancy of plasma cell like cells, can be confused with MM

*low grade lymphoma with a mix of mature B cells, plasma cells and in between cells
*IgM spike, leading to hyperviscosity!! BUT no bone lesions
-does have lymphadenopathy

*subtype of NHL
How do you treat Waldenstrom's Macroglobulinemia?
-like an NHL
-plasmapheresis for viscosity sx
Heavy Chain Disease
-VERY VERY RARE
-B cell lymphoproliferative disorder
-secretes a mutated monoclonal Ig that has a shortened piece of the Fc region; light chains and variable portion of heavy chain are missing

-1/3 cases come from CLL, 10% from MM
How does HCD present?
-constitutional symptoms
-HSM
-lymphadenopathy
-lytic bone lesions
-recurrent infection

-lymphocytes nad plasma cells on histology, vacuoles with abnormal mu protein
Treatment of HCD
-palliation, relieving tissue infiltration

-chemo is an option but the disease is so rare that experience is limited
Cryoglobulinemia
-presence of Igs in serum that PRECIPITATE at less than body temperature

-causes systemic inflammation from precipitated immune complexes

-Types I - III
Type I Cryoglobulinemia
-monoclonal Ig, usually IgM
-associated w/ underlying lymphoproliferative disorder (need abnormal B cell clone)

*does not activate complement, no systemic inflamm
-hyperviscosity, small vessel obstruction, digit gangrene
Type II Cryoglobulinemia
-mixed cryoglobulinemia
-monoclonal rheumatoid factor that binds Fc part of IgG
-also IgM from lymphoprolif disorders, autoimmune diseases, chronic infections (HepC, EBV, CMV, syphilis)

*immune complexes activate complex
-get arthralgias, myalgias, renal disease, cutaneous vasculitis (erythematous macules and purpuritic papules, ulcerations)
- if immune complexes in kidneys, causes membranoproliferative glomerulitis
-can have neuropathy or pulmonary infiltrates
Type III Cryoglobulinemia
-polyclonal, from rheumatoid factor IgM's against Fc part of IgG

*same sx as type II
Treating Cryoglobulinemia
-not needed until end organ damage
-avoid cold
-plasmapheresis

*Type I: treat underlying lymphoproliferative disorder

Types II and III: anti-inflammatories, steroids, immunosuppressants
-look for and treat HCV with IFN-alpha and ribaviron
Review cases
p.388
MPDs (6)
-"Pre-leukemic disorders"
CML**
-Chronic Neutrophilic leukemia
-Chronic Eosinophilic Leukemia/HES
-Polycythemia Vera
-Essential Thrombocytosis
-Primary myelofibrosis
Four common features of MPDs
-clonal disorder arising from a HEMATOPOIETIC stem cell
-overproduction of a specific lineage of myelo-erythroid cells
-tendency to progress to acute leukemia (100% in CML, less than 5% in ET ..10-20 in PV)
-abnormalities of coagulation (increase in thrombohemorrhagic events)
How does clonal hematopoiesis occur? (MPDs)
-acquired dominant bone marrow disorder
What is overexpressed in CML?
neutrophils and their precursors
What is overexpressed in Chronic eosinophilic leukemia?
eosinophils
What is overexpressed in PV?
mature erythrocytes, increased red cell mass
What is overexpressed in ET?
platelets and bone marrow megakaryocytes
What is overexpressed in primary myelofibrosis?
marrow fibroblasts with extramedullary hematopoiesis (must rule out CML and MDS first)

(React to megakaryocyte signals)
Thrombohemorrhagic complications of MPD
-arterial and venous thromboses
-microcirculatory disorders like erythromelalgia (painful red fingers and toes)
-mild, mucosal bleeding episodes
What is often the underlying pathogenesis for MPDs?
-dysregulated tyrosine kinases
CML epidemiology
-low incidence; 20% of adult leukemia
*people live a long time so prevalence is high
What is the only documented risk factor for CML?
-ionizing radiation (A-bomb survivors, etc)

-possibly post therapy, not proven .. and genetics, etc. are not associated
What are the three phases of CML?
-Chronic phase
-Accelerated phase
-Blast crisis (progression to AML or ALL)
What are lab findings associated with CML?
-low leukocyte alkaline phosphatase (LAP) = how you tell infection from CML
-splenomegally
-mild anemia
-WBC > 100,000
-high platelets (>600,000)
What are the clinical manifestations of CML?
-asymptomatic at dx often
-if sx, fatigue, malaise, weight loss, SWEATING
-abdominal fullness and early satiety (splenomegaly)
-acute gouty arthritis from hyperuricemia
-bleeding episodes from platelet dysfunction
Chronic phase
- large increase in committed myeloid progenitors with leukocytosis and thrombocytosis, left shift, can see basophils
-neutrophils can cause organ infiltration, HSM ..
*relatively stable phase
Accelerated phase
-increasing WBC
-increasing splenomegaly, tissue infiltration
-need more intensive myelosuppression therapy
-impairment of neutrophil differentiation
-increasing left shift
-increasing circulating and BM blasts

-still good myeloid cell maturation (unlike AML/ALL)

**percentage of blasts is <20%
Blast crisis
-resembles acute leukemia
-profound block in differentiation of cells

2/3 of cases are AML, 1/3 ALL
-inevitable progression within 3-5 yrs after dx and 3-18mo after accelerated phase
Diagnosis of CML
*requires detection of Ph chromosome or if cytogenetics negative, detecting BCR-ABL gene by FISH
BCR-ABL mechanism
-ABL on 9q moves to BCR on 22q
-creates novel fusion protein that increases levels of tyrosine phosphorylated proteins, constitutive activation of pathways leading to increased proliferation, survival, and resistance to DNA damage
What is the purpose of reverse-transcriptase PCR?
-detects minute amounts of BCR-ABL fusion mRNA

*used to monitor response of disease to therapy (more accurate because just restoring hematopoiesis and having negative Ph by cytogenetics/FISH isn't definitive; low sensitivity)

Remember: on FISH, yellow = BCR ABL fusion (normally 2 red and 2 green dots, if Ph chrom has 1 red, 1 green, 2 yellow)
How does atypical CML present?
-analyze by FISH or RT-PCR detects BCR-ABl fusion

-behave just like classic Ph+ CML
How are patients who lack BCR-ABL classified?
MDS or other atypical chronic myeloid disorder
-associated with other dysregulated tyrosine kinases, may be treatable with a different TKI
How do you treat CML?
**gold standard = kinase inhibitor therapy
-hydroxyurea
-busulfan
Effects of hydroxyurea and busulfan
-hydroxyurea inhibits ribonucleoside reductase which inhibits DNA synth

-both drugs lower leukocyte count and decrease spleen size, but do not prevent progression to blast crisis!!
-also affect normal BM cells equally
**NO SURVIVAL BENEFIT
Effect of IFN alpha
-survival benefit
-drug is toxic, inconvenient, and does not give a complete response
-get flu like symptoms
What chemo drug can you combine with IFNalpha?
-cytarabine
-better than IFN alpha alone
Imatinib
-BCR-ABL inhibitor
-phenylamino pyrimidine core

*suppresses growth of Ph+ myeloid progenitors without affecting normal progenitors!

MUCH better than IFNa because:
-complete hematologic remission (normalizes blood counts)
-major ctogenetic remission (<35% Ph cells)
-major molecular response (>1000 fold drop in BCR-ABL transcript levels)
**
TK inhibitor mechansim
-competes with ATP for binding to kinase catalytic domain
Oncogene addiction
-specific killing of Ph+ chrom cells demonstrated by imitinab
-suggests that Ph+ cells have become dependent on the activity of BCR-ABL
Problems with Imitinab?
-most patients dont go into molecular remission (don't become PCR negative)
*suppresses disease but not curing it
-also, quiescent Ph+ progenitor/stem cells are not killed by imitinab
-many still have progression of their disease on the drug (don't stay in chronic phase)

-toxicity: edema, N/V/D, cramps, myelosuppression
How can CML become resistant to Imitinab? (2)
-mutations in ABL
-T3151 (point mutation)
-doesn't allow drug to bind, so it becomes resistant

-2nd mechanism: amplification of BCR-ABL causing overproduction, so it is harder to totally inhibit it with the drug
What are the two major complications of ET?
-hemorrhage

-thrombosis
What is the goal of ET treatment and what is used to treat it?
-reduce platelet count below 400,000 to reduce risk of thrombosis
-hydroxyurea
-also use low dose aspirin to reduce thrombosis

*if low risk, can be managed to observation alone
Anegrelide
-lowers platelet counts in ET but doesn't reduce thrombosis risk
What is primary myelofibrosis?
-MPD with clonal HSC disorder, but has increased bone marrow fibrosis
-BM myeloproliferation, dysplastic Mks, nonclonal BM fibroblast proliferation due to TGFB and PDGF (causes overgrowth of BM space)
-overgrowth causes HSC to circulate in the blood and add areas of new hematopoiesis in spleen, etc...

**some evolve from PV or ET
-extramedullary hematopoiesis
What mutation is the cause of PMF?
-JAK 2 mutation (30% of cases)
**Note: this also occurs in ET
Risk factors for PMF (3)
-ionizing radiation
-benzene
-thorium dioxide
Presentation of PMF
-nonspecific systemic symptoms, fatigue
-marked HSM
-anemia
-high or low platelets and WBC counts
Labs for PMF?
-normocytic with TEARDROPS (dacrocytes) on smear
-leukoerythroblastic changes
-BM shows fibrosis, fatty change
-massive splenomegaly

*cytopenias
Dacrocytes
-in peripheral blood
-permanent RBC deformation, caused when immature erythrocyte moves from fibrotic bone marrow
What is the critical finding on the BM biopsy for PMF?
-increased collagen fibrosis (reticulin stain)
Second generation ABL kinase inhibitors for CML
-nilotinib (more potent)
-dasatinib (dual ABL/SRC inhibitors)

-work for some BCR-ABL mutants
What is the only known curative therapy for CML?
-allogeneic HSCT due to strong GvL effect
-best with HLA matched sibling
**give as a last option, due to major difficulties that many patients go through after (GvHD, etc)..try TKIs first
Prognostic factors for CML
-depends on age (increase in GvHD)
-CP responds better than AP >> BC
-time from Dx
-prior Rx (IFNa, busulfan, Gleevec)
What are the disadvantages of matched unrelated donors (MUD)?
-increased graft failure and GvHD
-decreased overall and DF survival
-better outcomes in young (<35) patients
Chronic Eosinophilic Leukemia
-rare clonal chronic MPD like disorder
-dysplastic eosinophils in BM, peripheral blood overproduced
-secondary cause of eosinohpilia must be excluded (infection, allergy, etc)
-exclude eosinophilic variant of CML (PH+) or AML-M4 (inv16)
What is the mutation in CEL?
-*deletion on chrom 4 that causes fusion of PDGFRalpha (receptor TK) with another protein
**also respond to imatinib!!
Polycythemia vera
-autonomous overproduction of mature RBCs (Epo-independent)
-increased hematocrit and Hgb, increased total body red cell mass
How does PV present
-often asymptomatic
-hemolytic anemia
-fatigue, dizziness
*post shower pruritis and erythromelalgia
-A/V thrombosis and gout
-plethora
-HSM
What do the labs show in PV?
-increased leukocytes
-increased platelets
-erythrocytosis

*cytogenetics normal!!
Diagnosing PV
-Hct>48% if female, 52% if male
-Hgb>16.5 or 18.5
-increased red cell mass
-normal arterial O2 saturation with no secondary polycythemia
-splenomegaly, cytogenetic abnormalities, platelets >400,000, absolute neutrophils >10,000, low Epo

-1 in 10 patients with polycythemia actually have P.vera, and only 40% of increased Hct have polycythemia
(ex: polycythemia can be caused by COPD, etc due to increased Epo)
Secondary Polycythemia
-chronic hypoxia-smoking, COPD, CO exposure
-Epo secreting tumors or RCC/hemangioblastoma
-hemoglobinopathy with increased O2 activity
-congenital polycythemia-EpoR mutation (Chauvash polycythemia)

*erythrocytosis caused by increased Epo levels .. NOT PV! may just be at high altitude, etc..
What is the mutation in PV?
-JAK 2 (nonreceptor tyrosine kinase)
-increase in JAK2 in absence of Epo mimics effects of Epo on erythroid cells
-analogous to BCR-ABL fusion only with V617F mutation
-mutation is in the autoinhibitory domain of JH2, activates the kinase in absence of Epo stimulation

**can be detected molecularly (JAK2 V617F)
Treatment for PV
-no targeted JAK 2 inhibitors currently available
*goal in treatment is to reduce hematocrit to normal, which reduces risk of fatal MI or stroke

*phlebotomy (reduces hematocrit, but red cells are quickly replaced, inconvenient, exacerbate PV)
-myelosuppressive therapy with hydroxyurea for high risk patients
-can also give low dose aspirin
Treatment for refractory patients?
-IFNa
-anagrelide if really refractory
-allopurinol to treat hyperuricemia
Essential thrombocytopenia
-too many platelets
-median age 60 yrs
-clonal disorder with a FEMALE disposition
-same JAK2 V617F mutation
-1/3 asymptomatic, can have vasomotor sx, thrombosis, hemorrhage
-low incidence of AML transformation
-spontaneous abortion in 1st trimester
How do you diagnose ET?
-its a diagnosis of exclusion
-persistently increased platelets > 600,000
-abundance of platelets with Mk fragments
-bone marrow biopsy shows megakaryocytic hyperplasia
*must be no evidence of BCR-ABL fusion by FISH or PCR, and no evidence of MDS - cytogenetics usually normal
How are PV and ET different? (Besides that one is RBCs and one is platelets)
-PV has low Epo levels
-ET has variable thrombopoietin levels
What are some cytogenetic abnormalities in PMF?
-13qdel, 20qdel
**must not have Ph chromosome+

(remember: must exclude causes of secondary fibrosis like infection, malignancy, or other Ph-MPD)
PMF prognosis?
-very poor, average 2-3 years following diagnosis
-only curative treatment is alloHSCT (works even though BM is fibrosed)
-mostly supportive tx (transfusions and growth factors)
-treat anemia (androgens, danazol, corticosteroids, thalidomide)
-chemo (for organomegaly or constitutional sx with melphalan, busulfan, HU
-splenectomy for pain, transfusion dependent anemia, or thrombocytopenia
Significance of the JAK2 mutation?
-if positive, means its a myeloproliferative neoplasm rather than a benign condition
-if it is negative in a ptient with polycythemia, probably a benign cause
MDS syndromes (6)
-Refractory anemia
-Refractory anemia+ringed sideroblasts
-Refractory cytopenia w/ multilineage dysplasia (RCMD
-RCMD + ringed sideroblasts
-Refractory anemia with excess blasts types I (5% blasts) and II (5-19% blasts)
-Deletion(5q)
What are myelodysplastic syndromes?
-clonal hematopoietic stem cell diseases that can't properly differentiate
-cytopenias with cellular BM (NOT aplastic anemia, just have dysplasia), dysplasia, and ineffective hematopoiesis in 1+ cell lines
-variable progression to acute myeloid leukemia
-may be de novo or after therapy
What do the stem cells in MDS look like?
-defective self renewal and differentiation
-abnormally increased cell death, apoptosis, in MDS marrow (ineffective hematopoiesis)
What does the bone marrow look like in MDS?
-increase of myeloblasts, but <20%
-but lack of mature blood cells
Who gets MDS?
-elderly, median age is 70 yrs
-very increased risk if they've received chemo for another malignancy (alkylating agents, topo inhibitors) ..10-15% of cases
What are the causes of de novo MDS?
-unknown
-viruses
-benzene and chemicals
-cigarette smoking
-fanconi anemia (congenital BM failure)
What causes Therapy related MDS (t-MDS)?
-exposure to chemo, esp alkylating agents (and topo II inhibitors)
-radiation therapy
**has a latency period before it develops after treatment
*see complex karyotypes
MDS prognosis?
-poor, very resistant to most therapies
How do patients present with MDS?
-many asymptomatic
-previously unrecognized anemia (fatigue, exercise intolerance, etc..)
-neutropenia, thrombocytopenia (infection, bruising)
-fever, weight loss later
Does MDS progress to AML?
Yes
Why do people with MDS get infections?
-neutropenia
*major clinical problem in MDS, main cause of death
-esp. if ANC is below 1000, esp below 500
-bacterial infections especially, some fungal
-skin is most common site
(also due to impaired granulocyte function)
What are the three ways to diagnose/classify MDS?
-peripheral blood cytopenias, which ones?
-BM morphology
-BM cytogenetics
Name some possible peripheral blood findings in MDS (8)
-anemia w/ low reticulocyte
-normo or macrocytic RBCs (must rule out B12 deficiency)
-ovalomacrocytosis, elliptocytes, teardrops, stomatocytes, acanthocytes
-basophilic stippling, Howell-Jolly bodies, megaloblastoid nucleated RBCs
-leuko or thrombocytopenia
**Pelger-Huet anomaly (Pince nez)
-sometimes monocytosis or large platelets
What does the bone marrow look like in MDS?
-HYPERcellular (90%..normal is 40-50%..opposite of aplastic anemia )
-erythroid dysplasia
-sometimes "ringed sideroblasts"
-myeloid dysplasia + karyorrhexis (fragmented nuclei)
-micromegakaryocytes (hypolobarity)
Types of dysplasia in the erythroid lineage
-megaloblastoid change (nuclear:cytoplasm ratio)
-cytoplasmic vacuolization
-binucleation, nuclear budding or irregularities
-ringed sideroblasts
What are ringed sideroblasts?
-dysplastic erythroid precursors w/ iron laden mitochondria surrounding nucleus
Types of dysplasia in the myeloid lineage
-pseudo Pelger-Huet nucleus (bilobed)
-nuclear hypersegmentation or abnormal shape
-cytoplasmic hypogranulation or abnormal granulation
Types of dysplasia in the Megakaryocyte lineage
-small size
-nuclear simplification
-separated nuclear lobes
What are the most common cytogenetic abnormalities in MDS? (3)
-del5q
-del7q
-11q23

in 40-70% of patients with primary MDS
What is the significance of an 11q23 cytogenetic abnormality?
-common in t-MDS secondary to topo inhibitor therapy (but can also have primary 11q23 MDS)
What is the significance of a del5q or 7q cytogenetic abnormality?
-can be primary or secondary MDS
-but if t-MDS, caused by the ALKYLATORS (topo inhibitors cause the 11q23 translocation)
Which abnormalities are favorable?
-del5q
-normal karyotype
-del20q
-Y
Which abnormalities are not favorable?
-del7q
-complex karyotype (>3 abnormalities)
-11q23
Refractory anemia
-anemia with few or no blasts
(erythroid dysplasia)
Refractory anemia with ringed sideroblasts
-anemia with no blasts, ringed sideroblasts
(erythroid dysplasia)
RCMD
-cytopenias involving 2+
-no or few blasts
-ringed sideroblasts if they have RCMD RS

(dysplasia of 2+ lineages)
Refractory anemia with excess blasts
-cytopenias
-excess blasts (5-9% in type I, 10-19% in type II)
5q syndrome
-40% of t-MDS
-overrepresented in femalse
-megakaryocytes with hypolobated nuclei
-refractory macrocytic anemia
-normal or increased platelets

**good clinical course

Possible cause: tumor suppressor gene (RPS14) on 5q
IPSS in MDS
-looks at marrow blast %, marrow karyotype, # and degree of cytopenia, and age

-predicts clinical survival
Treatment of MDS: Goals
-control symptoms from cytopenias
-improving quality of life, minimizing toxicity of therapy
-decreasing progression to AML
How do you treat MDS?
-transfusion with iron chelation therapy
-growth factors (G-CSF, Epo)
-low dose chemo (cytarabine) has a little role because dysplastic cells can't be eliminated
-allogeneicHSCT(ONLY CURATIVE TX, but limited to young patients with HLA matches)
Revlimid
-new treatment
-derivative of thalidomide
-works for 5q syndrome
Decitabine, azacytidine
-demethylating agents
-suppress MDS by altering gene expression patterns
(immunomodulatory)
When do you consider bone marrow transplant?
-patients who relapse after chemo and receive salvage chemo, because their long-term survival is basically zero (except for lymphoma)
-consider for transplant as opposed to undergoing more chemo
**esp because risk of 2ndary leukemia increases with more chemo
Why get an SCT from sibling or unrelated donor?
-immunological graft versus tumor effect (GvT), even cells that high doses of chemo can't kill
-with GvT, controls re-emerging clonal cells via the donor derived immune system (the donor's T cells, NK cells and cytokines)
What is the pre-transplant workup?
-cardiac function test (MUGA) & pulmonary function test .. these can be affected by high dose chemo/radiation
-check for hepatitis, HIV, CMV, EBV (can reactivate during neutropenia period after transplant)
-dental eval if poor dental health
-radiation-oncology eval
-check vascular access
What cells are harvested in SCT?
-early hematopoietic stem cell
-quiescent, non-dividing, but can give rise to committed progenitor cells
*CD34+
*need 2x10^6/kg stem cells to repopulate a myeloablated patient
What are the three sources of stem cells in body?
-marrow from upper iliac crest by aspiration under general anesthesia
-peripheral blood stem cells are mobilized to enter blood and collected by apheresis
-umbilical cord blood donated after the delivery of a baby
How do you stimulate stem cells to enter the periphery for a PBSC collection?
-five days of G-CSF - Neupogen
How do you prepare (condition) a patient for HSCT?
-5-7 days of either myeloablative therapy or reduced intensity (if older or comorbidities) myeloablation

Purpose: kill any residual cancer cells in patient's body
-if it's an allogeneic HSCT, induce an immuno-suppressed state that allows it to accept the transplanted graft and not reject it
Myeloablative prep
-2 days high dose cyclophosphamide
-3 days TBI
total 5 days

Reduced intensity is 6 days, first 2 are photopheresis, then 2 days pentostatin and 2 days TBI
How do you prepare for an autologous SCT?
-1 day of cyclophosphamide to kill cancer cells and committed progenitor cells
-gets neutropenia for 2 weeks (G-CSF administered to help them recover)
-then stem cell collection from their peripheral blood and freeze it
-patient is admitted and given high doses myeloablative chemo (melphalan) just before the stem cell influsion
How do you harvest allogeneic stem cells?
-either do leukapheresis (give donor Neupogen for 5 days and then collecting)
-or through a bone marrow harvest (donor placed under anasthesia, take from iliac crest) performed on the same day as the transplant
How do you prepare patient for an allogeneic SCT?
-for patient: 2 days photopheresis, 2 days pentostatin, 2 days

Note: stem cells for allogeneic are not cryopreserved, more like a blood transfusion
Complications of an autologous SCT
-rare
-related to cryoprotectant used to freeze the cells
-fluid overload, fever, hypertension, bradycardia, SOB, hypotension, shock, death
How do you handle patients after an autologous transplant/post-transplant complications?
-very neutropenic for 8-12 days, give prophylactic antibiotics
-may need parental nutrition from mucositis or lack of appetite
-watch out for SOS
-pulmonary hemorrhage is rare, usually in pts with prior lung radiation
Sinusoidal Occlusion Syndrome
-patients who have been heavily pre-treated can have organ compromise of the liver (can also hvae renal failure)
Mortality of an autologous SCT? allogeneic?
1-2% for autologous

10-30% for allogeneic
What about patient tx after an allogeneic SCT?
-same care as autologous + immunosupprsesive meds to prevent rejection of the graft (this is rare in comparison to solid organ transplants) and GvHD
What is GvHD?
initiated by the donor's T-lymphocytes
-recognize minor antigens on patient tissue
-produce cytokines like TNF that cause tissue damage in 3 organs
Which 3 organs are affected by GvHD?
-skin (rash is mild or can burn, exfoliate)
-GI system (N/V/D, severe abdominal pain, bleeding, ulceration)
-liver (transaminase, bilirubin elevation)
When does GvHD develop?
-2-4 weeks after transplant (when donor cells engraft in recipient)
What is the frequency of GvHD?
-50% of patients even with prophylaxis
How do you treat GvHD?
-steroids
What is chronic GVHD?
-after several months of insufficiently treated/non-responsive acute GvHD

-can also occur denovo

-autoimmune diseases:
vitiligo
dyspigmentation
scleroderma-like
contractures
hair and nail abnormalities
dry eyes (keratitis)
immune deficiency
lung
vagina
Define "cure"
-long term survival without disease recurrence
-more than 10 years

*can be achieved with HSCT (autologous) in MM after induction, NHL and HD after remission, AML

*for allogeneic HSCT, can cure aplastic anemia, AML, ALL, MDS, CML, MM, NHL (peripheral T cell/MCL), CLL, hemoglobinopathies
Why give autologous HSCT instead of allogeneic?
-less mortality (death with these is more likely due to recurrence of primary disease; with allogeneic, more likely due to a complication of the transplant like infection)
-if patient is elderly or comorbidities

*these only allow you to increase patient's ability to tolerate higher doses of chemo; lacks the GvT effect of an allogeneic transplant
How likely is an HLA match between siblings?
-25%
-inherit one gene haplotype from each parents
-4 possible different haplotype combinations

-if no sibling donor found, search National Marrow Donor Registry
Which ethnic group is most likely to have a match? For which is a match difficult to find?
...see slide on TUSK
How do you obtain cord blood?
-let gravity empty placenta
-80-100 cc of blood
-shipped to blood bank for storage
What are the limitations of using cord blood?
-only a limited number of stem cells in the umbilical cord blood
-ideal source of transplantation for kids, but not enough for adults
Why use marrow/blood as opposed to cord blood and vice versa?
-marrow/blood takes 3-4 months with potential for diseaes transmission and GvHD, but has enough stem cells for an adult

-cord blood only takes 13.5 days to find a match, and no donor issues or viral infection risks; less GvHD
-less useful in adults unless there is a disease progressing so fast there is not enough time to find an unrelated donor
2 most common uses of autologous HSCT
-MM
-lymphoma
2 most common uses of allogeneic HSCT
-leukemia
-MPD
Mini transplants
-safer, lower doses of chemo
-only done in patients who are in remission at time of transplant
-still get GvT (more important than in regular allogeneic SCT because you are giving less chemo)
High vs. low penetrance genes
*intrinsic susceptibility variables HIGH
-low frequency (affect small proportion of patients)
-high risk
-environment plays a minor role in causation

LOW
-affect most people with cancer
-environment has a much larger role in cancer causation
How much of breast cancer cases have a family history?
-20%

**80% don't have a family history!!
Is this type of breast cancer caused by high or low penetrance genes?
-undefined low penetrance genetic predispositions (not known germline mutations)
-risk increases with increasing number of affected relatives
How much of breast cancer is related to familial cancer syndromes, and what type of genes cause this form of breast cancer?
-10%
-2/3 are BRCA1 or BRCA2 genes, which are predisposing, high penetrance genes
-autosomal dominant inheritance
-associated with other cancers like ovarian, prostate, colon, etc.
-secondary breast cancer (BRCA1) or male breast cancer (BRCA2)
-extremely high lifetime risk of breast cancer
**increased in Ashkenazi Jews, populations with restricted gene pool mixing
Is BRCA1 hormone receptor positive?
usually not
Is BRCA2 hormone receptor positive?
usually positive!
What other types of familial diseases can increase risk of breast cancer?
-Li Fraumeni (p53), Cowden syndrome (PTEN), CDH1, ataxia-telangectasia(ATM)
-Klinefelter's syndrome increases risk of male breast cancer 50!! equivalent to female risk!

-if a female patient known to have one of these diseases, can do further testing like MRI screening in addition to mammograms
What has the highest incidence of cancer in women?
breast cancer
-also a very high prevalence (22% of cancer)
Why has the incidence of breast cancer been increasing?
-likely not true incidence increase but due to better detection methods; coincides with mammography
-this is further evidenced by the corresponding increase in early stage detection/in situ carcinomas (microcalcifications that are not clinically palpable, still localized to breast)
-similar thing has occurred with prostate cancer and PSA screening
Has this corresponded with a decrease in mortality for breast cancer?
-yes
-more women are treated at a stage that is more curable
What is the defect in Hereditary Non-Polyposis Colon cancer?
-microsatellite instability and repetition
Hereditary Melanoma/Xeroderma pigmentosum
-defects in CDKN2A locus (cell cycle inhibitor); XP genes have defective nucleotide excision repair, UV induced damage causes melanoma
Retinoblastoma
-most common childhood intraocular tumor is 40% hereditary with a high risk of developing other sarcomas and melanoma
Pheochromocytomas
-rare catecholamine secreting tumors of adrenal medulla

*15% familial (multiple endocrine neoplasia syndrome - RET tyrosine kinase defect, von Hippel Lindau - VHL tumor suppressor, Neurofibromatosis)
Neurofibromatosis
-cutaneous disorder, cafe au lait spots, neurofibromas

Type 1 has a defect in NF1 gene, half of cases are familial

-neurofibromin normally inactivates ras
Extrinsic vs. Intrinsic exposure (ex: estrogens)
-extrinsic factor = exposure to estrogen

-oral contraceptives, HRT
-in Klinefelter's, more endogenous estrogen

-intrinsic factor = # periods (early menarche, late menopause), genes
Favorable risk factors in breast cancer
-higher # pregnancy = intrinsic
-longer periods breast feeding = intrinsic
Other factors that increase breast cancer risk
-higher bone density (higher circulating estradiol) =intrinsic
-increased age at first life birth (after age 30) = intrinsic
-prenatal use of diethylstilbestrol = extrinsic (nonsteroidal synthetic estrogen used for post-menopause and prematurity, no longer used) causes bc in the mother; baby girl at risk for vag/cervical cancer but not bc
What other cancer is most likely to lead to breast cancer in young women?
-Hodgkins lymphoma in early 20s (or NHL) if patients receive ionizing radiation to chest
-10-30% increase in risk
What is the "ultimate" extrinsic risk factor?
-smoking!!
-increase in incidence in lung cancer and other related cancers
Diet
-extrinsic factor extremely important in development of colorectal cancer
"Western" diet is bad; African diet is good b/c high in fiber
Alcohol
-contributes to development of many cancers
-co-carcinogen with smoking! enhances carcinogenicity of tobacco (mouth, larynx, pharynx, esophagus) AND breast cancer
Viruses
-HBV, HCV = hepatocellular carcinoma
-HPV = cervical cancer
-HHV8 = vascular tumor in AIDS pts
-EBV = NHL, HL, Burkitt's, etc)
-H. pylori
Other lifestyle factors in breast cancer
-alcohol
-high BMI
-sedentary lifestyle
Origins of breast cancer
-95% epithelial (most from ductal cells, 10% lobular cells)
How do you describe the location of a breast tumor?
-upper outer, upper inner, lower outer, lower inner quadrant
OR
-clock face (ex: 1cm mass in UOQ 3 cm from the nipple towards 11:00)
Lymphatic drainage of the breast
-axillary, then supraclavicular chains (may be palpable if abnormal)
*Note: lower inner quaddrant drains a bit to internal mammary chain
How do you stage breast cancer?
-TNM system
-T0 = in situ
-T1 = <2cm, T2 = 2-5, T3 = >5, T4 = any size but involves skin or chest wall
-N0 = no nodes, N1 = 1-3, N2 = 4-9, N3 = >10
-M0 = no metastasis, M1 = metastasis
Stage 0 breast cancer
T0N0M0 ... in situ
Stage I breast cancer
-T1 N0 M0
Stage IIA breast cancer
-T0 N1 M0 up to T2 N0 M0
Stage IIB breast cancer
-T2 N1 M0 up to T3 N0 M0
Stage IIIA breast cancer
-T0 N2 M0 up to T3 N2 M0
Stage IIIB breast cancer
-T4 N0 M0 up to T4 N2 Mo
Stage IIIc breast cancer
any T, but 3 nodes involved, M0
Stage IV breast cancer
-any T, any N, M1 (metastasis)
*18% 10 yr survival (in contrast to in situ which is stage 0 and 92% survival)