Week 3 Flashcards

1
Q

o Activated lymphocytes:

A

differentiate into effector cells once bound to antigen, which eliminate the offending organism, and memory cells, which can be reactivated upon second exposure.

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

o Effector Lymphocytes:

A

produced by the differentiation of T cells and produce distinct sets of cytokines and perform different functions (i.e. Th1, Th2, Th17)

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

o Memory Lymphocytes:

A

are expanded pools of antigen-specific lymphocytes which respond faster and more effectively when re-exposed to antigen than do naïve cells. One goal of vaccination is to generate memory cells

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

• Describe how light chain expression can be used to determine if a B-lymphocyte proliferation is clonal. Describe the use of T-cell receptor gene rearrangement studies and B-cell immunoglobulin gene rearrangement studies.

A
o	B lymphocytes clonal proliferations typically express only one type of immunoglobulin and thus only one light chain (kappa or lambda) since light chains do not change upon class switching. 
o	T lymphocytes recognize specific bound antigen through binding to their TCRs
•	For both B and T lymphocytes, due to clonal proliferation actions performed by both cells it can be difficult to distinguish between normal clonal proliferation and clonal neoplastic proliferation; however, Ig and TCR gene rearrangement analysis can help, respectively
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

o Generative lymphoid organs

A

sites where T and B lymphocytes mature and become competent to respond to antigens (bone marrow and thymus)

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

o Peripheral lymphoid organs

A

sites where adaptive immunity is initiated (lymph nodes, spleen, mucosal and cutaneous lymphoid tissues)
• B cells found in follicles of lymph node; T cells in paracortical region
• Lymphocytes constantly recirculate between tissues and home to particular sites

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

o Major Histocompatibility Complex (MHC)

A

physiologic function is to display peptide fragments of proteins for recognition by antigen specific T cells
• MHC genes found on chromosome 6
• MHC aka human leukocyte antigen (HLA) complex and produce membrane bound glycoproteins which are found on all nucleated cells (except mature RBCs)
• MHC system is highly polymorphic (many different alleles of each MHC gene)
• MHC I: display proteins derived from the cytoplasm (viral antigen); recognized by CD8+ T cells and NK cells; coded by HLA-A, HLA-B, and HLA-C genes
• MHC II: display antigens that have been internalized into vesicles; recognized by CD4+ T cells; coded by HLA-DP, HLA-DQ, DLA-DR genes

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

o HLA Testing Uses

A

• A variety of diseases are associated with the inheritance of certain HLA alleles; HLA testing can be used to determine disease risk
• Used in the transplantation workup as close matches of HLA-A, HLA-B, HLA-C and HLA-D in both the donor and graft recipient increase the chance of graft survival
o Induction and regulation of immune responses involve multiple interactions that are mediated by cytokines

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

• Describe the key steps and functions of cell-mediated immunity

A
  • Dendritic cells (DCs) capture microbial antigens from epithelia and tissues and transport the antigens to lymph nodes. During this process, the DCs mature, and express high levels of MHC molecules and costimulators.
  • Naïve T cells recognize MHC-associated peptide antigens displayed on DCs. The T cells are activated to proliferate and to differentiate into effector and memory cells, which migrate to sites of infection and serve various functions in cell-mediated immunity.
  • CD4+ effector T cells of the TH1 subset recognize the antigens of microbes ingested by phagocytes, and activate the phagocytes to kill the microbes.
  • CD4+ T cells also induce inflammation.
  • CD8+ cytotoxic T lymphocytes (CTLs) kill infected cells harboring microbes in the cytoplasm of the infected cell.
  • Not shown are TH2 cells, which are especially important in defense against helminthic infections. Some activated T cells differentiate into long-lived memory cells.
  • Following T cell migration, the activated T cell causes: inflammation, macrophage activation (killing ingested microbes), and T cell-mediated killing of infected cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

• Describe the key steps and functions of humoral immunity.

A
  • Naïve B lymphocytes recognize antigens, and under the influence of TH cells and other stimuli (not shown), the B cells are activated to proliferate and to differentiate into antibody-secreting plasma cells.
  • Some of the activated B cells undergo heavy-chain class switching and affinity maturation, and some become long-lived memory cells.
  • Antibodies of different heavy-chain classes (isotypes) perform different effector functions, shown on the right
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

List the four types of hypersensitivity reactions

A

individuals previously exposed to an antigen become sensitized and upon repeat exposure(s) some individuals develop a pathologic immune reaction to the antigen
• Exogenous and endogenous antigens elicit hypersensitivity reactions
• Development of Hypersensitivity Diseases is often associated with inheritance of particular susceptibility genes (HLA and non-HLA genes)
• Hypersensitivity reflects an imbalance between the effector mechanisms of immune responses and the control mechanisms that serve to limit such responses.
• Hypersensitivity diseases can be classified on the basis of the immunologic mechanism that mediates the disease
Type of Reaction Prototypic Disorder Immune MOA Pathologic Lesions
Immediate
(Type I) Anaphylaxis; allergies Production of IgE Ab → release of vasoactive amines from mast cells; recruit inflammatory cells later Vascular dilation, edema, smooth muscle contraction, mucus production, tissue injury, inflammation
Antibody-mediated
(Type II) Autoimmune hemolytic Anemia; Goodpasture syndrome Production of IgG, IgM → binds to Ag on target cell/tissue → phagocytosis/lysis of target cell Phagocytosis and lysis of cells; inflammation
Immune Complex-mediated
(Type III) SLE, Poststreptococcal Glomerulonephritis, Serum sickness Deposition of Ag-Ab complexes → complement activation → recruit leukocytes to release enzymes and toxic molecules Inflammation, necrotizing vasculitis (fibrinoid necrosis)
Cell-mediated (Type IV) Contact dermatitis; MS, T1D, RA, IBS, TB Activated T cells 1) release cytokines, 2) initiate T cell-mediated cytotoxicity Perivascular cellular infiltrates, edema, granuloma formation; cell destruction

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

o Atopy

A

predisposition to develop localized immediate hypersensitivity reactions
• Atopic individuals tend to have higher serum IgE levels and produce more IL-4 producing Th2 cells

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

• Describe the mechanisms involved in the immediate and late phase reactions of type I hypersensitivity reactions.

A

o Immediate hypersensitivity reactions are initiated by the introduction of an allergen, which stimulates TH2 responses and IgE production in genetically susceptible individuals. IgE binds to Fc receptors (FcεRI) on mast cells, and subsequent exposure to the allergen activates the mast cells to secrete the mediators that are responsible for the pathologic manifestations of immediate hypersensitivity.
• Characteristic features of immediate hypersensitivity reactions: vasodilation, vascular leakage, smooth muscle spasm
o Late phase hypersensitivity reactions are characterized by leukocyte infiltration, epithelial damage, and bronchospasm
o In some individuals, immediate hypersensitivity reactions are triggered by temperature extremes and exercise (non-atopic allergy) and do not involve T helper cells or IgE. In these cases it is believed that the mast cells are abnormally sensitive to activation by various non-immune stimuli

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

• Define localized allergic reaction, and list some common examples. Describe the treatment and prevention of localized allergic reactions.

A

o Localized reactions occur within a set, small space and include: allergic rhinitis (hay fever), bronchial asthma (atopic forms), urticarial (hives), and allergic gastroenteritis (food allergy)
o Treatment typically involves:
• Avoiding the offending allergen, if possible.
• Use of various medications, such as antihistamines, corticosteriods, agents that inhibit release of histamine from mast cells, leukotriene modifiers (to name just a few).
• In some cases, immunotherapy (desensitization therapy) is also used

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

• Define systemic anaphylaxis, and describe the mechanisms and clinical findings in systemic anaphylaxis. List some of the common inciting agents in fatal systemic anaphylaxis.

A

o Systemic anaphylaxis refers to a life threatening systemic allergic reaction typically characterized by vascular shock, widespread edema, and difficulty breathing (massive mast cell activation).
o Common inciting agents:
• Therapeutic agents (e.g. antibiotics - penicillin and cephalosporins, radiocontrast agents)
• Exposure to food products (e.g. peanuts, seafood)
• Insect toxin (e.g. bee or wasp sting)
• Latex allergy
o Clinical Findings: asphyxiation or respiratory failure due to bronchial constriction/obstruction and/or shock with CV collapse → death within minutes if untreated
• Median time for onset of symptoms and respiratory/CV collapse:
• Iatrogenic anaphylaxis (medical or surgical treatment) – 5 minutes
• Insect venom – 15 minutes
• Food-induced anaphylaxis – 30 minutes
o Treatment: IM epinephrine
• Venom immunotherapy as prevention; form of desensitization therapy: repeat injections of allergen in increasingly greater amounts results in production of IgG antibodies that can attach to allergens and prevent their binding to mast cells

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

• Define type II hypersensitivity reaction, and list the mechanisms of injury.

A

o Type II hypersensitivity reactions are caused by antibodies that react with normal or altered cell surface antigens, or with antigens in the extracellular matrix; autoimmunity
o Mechanisms of Injury:
• Opsonization of cells by antibodies and complement components cause ingestion by phagocytes
• Inflammation induced by antibody binding to Fc receptors of leukocytes and by complement breakdown products
• Anti-receptor antibodies disturb normal function of receptors (Myasthenia gravis)

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

Goodpasture Syndrome

A

target: Noncollagenous protein in basement membrane of kidney glomeruli and lung alveoli

mech of disease: Complement and Fc receptor mediated inflammation

clinical presentation:
Nephrites, lung hemorrhage

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

• Define type III hypersensitivity reaction, and list the mechanisms of injury. Describe some examples of this type of reaction, and explain why the clinical manifestations can be quite diverse.

A

o Type III hypersensitivity reactions are caused by antigen-antibody complexes which elicit inflammation at the sites of deposition.
o Mechanisms of Injury:
• The pathologic reaction begins when antigen combines with antibody in the circulation, and the circulating immune complexes are deposited in vessel walls.
• The involved antigens can be exogenous or endogenous, and the sites of involvement can be systemic or localized.
o Pathogenesis: Complement is consumed as part of the pathogenic process, and the levels of C3 can be used to monitor disease activity (low levels indicate active disease)
o Diseases associated with type III:
• SLE → antibodies to DNA
• Poststreptococcal glomerulonephritis → antibodies to glomerular basement membrane deposited by streptococcal cell wall antigens
• Polyarteritis nodosa → antibodies to antigen from hepatitis B virus
• Reactive arthritis → Ab to bacterial antigens
• Serum sickness → venom antigen is circulating and antibodies bind the venom

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

• SLE

A

antibodies to DNA

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

• Poststreptococcal glomerulonephritis

A

antibodies to glomerular basement membrane deposited by streptococcal cell wall antigens

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

• Polyarteritis nodosa

A

antibodies to antigen from hepatitis B virus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

• Reactive arthritis

A

Ab to bacterial antigens

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

• Serum sickness

A

→ venom antigen is circulating and antibodies bind the venom

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

• Define type IV hypersensitivity reaction, and list the mechanisms of injury. Describe the mechanism of granuloma formation, and list some examples of this type of reaction.

A

o Caused by T cell mediated immunity (no antibodies involved); CD4+ or CD8+ T lymphocytes are sensitized to exogenous or endogenous antigens. The resultant T cell immune response results in cell or tissue injury
o There are two mechanisms of T cell mediated (type IV) hypersensitivity:
• Delayed-type hypersensitivity reaction
• T cell mediated cytotoxicity
o Type IV Hypersensitivity used in Mantoux Test: inject antigen from TB (dead) and look for memory T cell activation to the antigen to form a nodule; measure area of induration
• In some places the Mantoux test is being replaced by quantiFERON-TB test which involves incubated the patient’s lymphocytes for 16-24 hours with a cocktail of TB antigens
• If patient has been previously exposed to TB the T cells will release cytokines (IFNγ) which can be measured using an immunoassay
o Granulomatous Formation: APC-bound antigen presents to Th1 cell which secretes IFNγ which acts on monocytes to create giant cells which are indicative of granulomatous formation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Q

• List the four types of genes typically mutated in cancer.

A

o Growth promoting proto-oncogenes
• Ex. BCR-ABL mutation in CML, K-ras in Colon Cancer, myc in Burkitt’s Lymphoma
o Growth initiating tumor suppressor genes
• Ex. BRCA1/BRCA2 in Breast Cancer, Rb in Retinoblastoma, TGF-β in pancreatic and colorectal carcinomas
o Genes that regulate apoptosis
• Ex. Bcl-2 overexpression → blocks apoptosis
o Genes involved in DNA repair (ex. PARP)
• Ex. telomerase expression to increase telomere length

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
26
Q

• Using only a few sentences for each, list the 8 essential alterations involved in malignant transformation of cells.

A

o 1. Self-sufficiency in growth signals
• Cell proliferation occurs due to proto-oncogenes becoming oncogenes. These oncogenes make oncoproteins which are devoid of regulatory elements and cause transformation in cells, but do not rely on growth factor or external stimuli.
o 2. Insensitivity to growth-inhibitory signals
• When tumor suppressor genes are silenced, it is considered a LOH, which typically causes expression of an oncogene to occur.
o 3. Evasion of apoptosis
• Over expression of Bcl-2 causes the blocking of apoptosis. This is a common characteristic of 85% of B-cell lymphomas which protects the B-cells from apoptosis, allowing the B-cells to survive for long periods of time.
o 4. Limitless replicative potential
• Telomerase expression results in maintenance of telomere length which avoids senescence. Maintenance of telomeres is seen in virtually all types of malignant neoplasms.
o 5. Sustained angiogenesis
• Tumors need vasculature to grow and proliferate. Angiogenesis is required for not only continued tumor growth, but also for access to the vasculature and hence metastases. VEGF is a common angiogenesis factor.
o 6. Ability to invade and metastasize
• This process involves invasion of the ECM and vascular dissemination, homing of tumor cells, and colonization. Invasion of ECM involves breaking through the basement membrane and intravasating into the bloodstream. Once in the bloodstream, the metastasizing cells need to get out (vascular dissemination). Homing of tumor cells results in association with other tumor cells. Colonization involves tumor cells secreting cytokines, growth factors, and ECM molecules to make the metastatic site habitable for the cancer cell.
o 7. Defects in DNA repair
• Effective DNA repair mechanisms are essential to maintain the integrity of the genome. Many inherited and sporadic cancers have defects in DNA repair mechanisms that allow for mutations in other genes during cell division.
o 8. Escape from immune attack
• The immune system is involved in surveying the body for malignant cells and destroying them. For tumor cells to survive, they must evade the immune system by various mechanisms: loss/reduction of MHC molecules, immunosuppression, antigen masking, and apoptosis of cytotoxic T-cells to name a few.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
27
Q

• ABL-BCR fusion gene:

A

results in increased tyrosine kinase activity (treat with tyrosine kinase inhibitor imatinib mesylate, Gleevac). [Slide 21 pathogenesis]

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

• HER 2/neu

A

results in over expression of cell membrane epidermal growth factor receptor - EGFR (in breast cancer), treat with monoclonal antibody to Her2/neu receptor, trastuzumab (Herceptin). Need to be Her2/neu + for tx to work.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
29
Q

• RAS:

A

results in persistent activation of the RAS signal (KRAS for colon cancer; presence of KRAS in colon cancer can be predictive of lack of response to certain forms of chemotherapy). Anti-EGFR antibody only effective if have KRAS mutation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

o Familial adenomatosis polyposis (FAP

A

Patients inherit a single APC (adenomatosis polyposis coli) mutation, and then acquire a second APC mutation. The loss of tumor suppressor function results in large numbers of adenomatous polyps in the colon and rectum during late childhood, adolescence, and early adulthood. There is a 100% risk of colorectal adenocarcinoma (mean age of carcinoma 35-40 yrs, can occur in the teenage yrs)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

• Describe how overexpression of BCL-2 can lead to follicular lymphoma

A

o Bcl-2 protein is an anti-apoptotic protein which limits cytochrome c release. 85% of B-cell lymphomas of the follicular type carry a characteristic t(14;18) translocation which results in overexpression of the Bcl-2 protein. This mutation allows lymphocytes to evade apoptosis and survive for long periods of time. They tend to be indolent (slow growing).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
32
Q

• Describe how overexpression of telomerase can lead to limitless replication.

A

o Telomerase is a protein that allows for elongation of telomeres on the ends of chromosomes. Typically telomeres shorten with time and result in senescence once they become too short → apoptosis. Maintaining telomere length is a protection mechanism for cancer cells that is attained through the action of telomerase.

33
Q

• Describe how cancer cells can initiate neoangiogenesis. What cytokine is typically involved?

A

o It is thought that angiogenesis is controlled by an increase in the production of angiogenic factors (such as VEGF) and/or a loss of angiogenic inhibitors. Neoangiogenesis is required not only for continued tumor growth but also for access to vasculature & hence metastases

34
Q

• Using just a few sentences, describe the steps involved in invasion and metastases. List some of the ways that cancer cells can evade immunologic detection.

A

o Invasion and metastasis are the biologic hallmarks of malignant tumors and are the major cause of cancer-related morbidity and mortality.
o Two major steps:
• Invasion of ECM – involves dissociation of tumor cells from one another, degradation of basement membrane and interstitial CT, attachment of tumor cells to ECM proteins, migration of tumor cells within ECM, and intravasation into blood vessels
• Vascular dissemination – once in bloodstream tumor cells may attach to endothelial cells; however, certain tumors have preferential spread to certain organs
• Organ tropism related to different expression of endothelial adhesion ligands and different chemokine receptor expression
• Colonization occurs from tumor cells secreting cytokines, growth factors, and ECM molecules that make metastatic site habitable for cancer cell
o Mechanisms that tumor cells use to evade the immune system:
• Selective outgrowth of antigen-negative variants.
• Loss or reduced expression of MHC molecules.
• Lack of costimulatory molecules (T-cell sensitization requires two signals, one by a foreign peptide presented by MHC molecules and another costimulatory molecule).
• Immunosuppression (either as a result of therapy or by tumor products).
• Antigen masking (surface antigens masked by glycocalyx molecules).
• Apoptosis of cytotoxic T cells (tumor cells express proteins which induce T-lymphocyte apoptosis when they come into contact with the lymphocyte)

35
Q

o Hereditary Nonpolyposis Colon Cancer Syndrome

A

autosomal dominant):
• Increased risk for carcinomas of the colon without previous polyps.
• Results from inactivation of genes involved in DNA mismatch repair (affected individuals inherit one defective allele, and a second hit occurs in the colonic epithelial cell randomly, producing the defect) – just like tumor suppressor genes
• Clinical finding: microsatellite instability (tandem repeats of 1-6 nucleotides; abnormal number/length when mutated)

36
Q

• Describe some of the structural chromosome changes that can be seen in malignancies.

A

o Genetic changes that occur in oncogenes or tumor suppressor genes can be subtle (point mutations) or large, involving big segments that can be detected using karyotyping. Chromosomes can undergo translocations, deletions, reduplication, and amplification.

37
Q

• Explain how gene expression profiles are used in the assessment of breast cancer

A

o Each cancer results from the accumulation of multiple mutations. Studies have shown that individual breast and colon cancers typically accumulate an average of 90 mutant genes. The emergence of malignant tumors requires the mutational alteration and loss of many genes, including oncogenes, tumor suppressor genes, and genes that regulate apoptosis and senescence.
o BRCA1/2 are tumor suppressor genes in breast cancer that can be altered through epigenetics – active area of research

38
Q

o Th1 Helper Cells

A
  • Induced by: IL-12 and IFNα
  • Proliferate in lymph nodes, some daughter cells leave and enter circulation; when encounter antigen they can secrete lymphokines
  • Most important lymphokine is IFNγ which is a pro-inflammatory and chemotactic agent for macrophages. These macrophages are activated by IFNγ to become classically activated M1 macrophages which avidly ingest and kill bacteria or other foreign invaders
  • Macrophages secrete pro-inflammatory cytokines such as TNFα and IL-1
  • Note, when you have inflammation you will have fever due to IL-1 production which works in the preoptic anterior hypothalamus which stimulates production of PGE2. PGE2 slows the firing rate of certain temperature-control neurons which activates the heat generation response (shivering) contributing to a fever.
39
Q

o Th17 Cells

A
  • Induced by: TGF-β and IL-21
  • Function:
  • Produces inflammatory lymphokines (IL-17, IL-22)
  • Resembles the job that Th1 cells have which is aggressive pro-inflammatory production leading to classic-activation of macrophages
  • Role in maintaining mucosal surface integrity
40
Q

o Th2 Cells

A
  • Th2 cells leave the lymph node and enter circulation and when encounter antigen they will become activated
  • Once activated, the Th2 cells will produce IL-4 and IL-13 which attracts macrophages (however this is different than the way IFNγ does) and are alternatively activated M2 which are more involved in healing (removes debris, scar formation, walling off pathogens)
  • Th2 helper cells seem to give rise to Tfh cells which migrate to lymphoid follicles; there high amounts of IL-4 push the B cell to switch from its naïve IgM/IgD state to making IgE (Ab for parasite resistance and allergy)
  • Thus, Th2 has two roles through parasite immunity: 1) M2 macrophage activation, 2) stimulation of IgE production
41
Q

o Follicular Helper T Cells, Tfh

A
  • Soon following arrival of antigen-presenting DC in the lymph node, some T cells migrate into the follicles of the cortex where B cells are abundant
  • Role: help B cells that have recognized antigen to become activated and differentiate into plasma cells that secrete antibodies and a variety of cytokines which direct the B cells to switch classes
  • Tfh are heterogeneous meaning that they cause B cells to preferentially switch classes to a certain Ig class; i.e. Tfh in gut → IgA, Tfh in spleen → IgG
  • Existence (or lack thereof) of antibody may be a read-out of T cell function just as much as B cell function. If Tfh cells can’t communicate correctly with B cells there may be difficulty making a certain class of antibody, especially those downstream of IgM
42
Q

o T regulatory Cells, Treg

A
  • Tregs are a small population of cells (about 5% of all Th cells) who have been identified to suppress the activation and function of all other Th cells.
  • Most regulatory T cells have the phenotype CD4+/CD25+.
  • Surface CD4 puts them in the helper family but they are probably best defined as antihelpers.
  • Produce TGFβ and IL-10 and are very potent to the other T helper cells and actually cause them to decrease their activity. One Treg can suppress 1,000 other Th cells.
  • If lack Treg, the person will have autoimmunity, overactive immune responses, and self-reactivity
43
Q

o Cytotoxic CD8+ (Killer) T cells, CTL

A
  • Function: give the “kiss of death” to cells that are infected → induces apoptosis causing the cell to destroy its DNA and for the cell to disintegrate (lyse); the cell can then b phagocytosed
  • Two ways a CTL can signal a cell to undergo apoptosis:
  • 1) Engage the “death receptor” Fas (CD95) on the target cell by binding with the CTLs FAS ligand (CD95L). Cross-linked Fas activates a latent apoptosis pathway.
  • 2) Secrete “lytic granule” contents which contain proteases called granzymes (degrade infected cell contents) and perforins (pore-forming) which allow the CTL to kill the target cell via apoptosis
  • CTLs are activated in lymph nodes after contact with APC (usually DC) and also require help from Th1 in the form of IL-2 for activation and conversion into memory cells via IL-12.
  • Note, DCs have both MHC I and II molecules → activate both helper and killer T cells
44
Q

• Describe the markers that Th1, Th2, and killer T cell subpopulations have on their surfaces.

A

o Th1, Th2, Th17: CD3+, CD4+
• Need to look at lymphokines each T cell secretes to differentiate between the Th cells
• Treg cells have CD25 in addition to CD3+ and CD4+ on their surface
o CTL: CD3+, CD8+

45
Q

• Describe how T cell receptors are generated.

A

o TCRs are structurally reminiscent of antibody and are composed of two chains: alpha and beta or gamma and delta – each has a constant and a variable portion
o T cell makes its receptor out of V, (D), and J regions from T cell genes (not B cell ones!!) and this process occurs in the thymus; once the T cell leaves the thymus the TCR does not change
• Both alpha and beta chains have transmembrane domains → TCRs are membrane bound!!
• The α chain, like the Ig light chain, is encoded by V, J, and C gene segments.
• Rearrangement leads to a selection of a VJ combination attached to a single C segment.
• The α chain has a transmembrane cytoplasmic tail making the TCR α receptor a membrane-bound chain.
• Two α chains can be produced by a single cell
• The β chain, like the Ig heavy chain, is encoded by V, D, J, and C gene segments.
• Rearrangement leads to a selection of a VDJ combination attached to one of two C gene segments.
• The β chain also has a transmembrane cytoplasmic tail making the TCR β receptor a membrane-bound chain.
• Each T cell will produce a single β chain from just one of the chromosomal loci (allelic exclusion). There is no way to switch to the other allelic version
• Pre-T cell expresses RAG 1 & 2 and are used in recombining the V(D)J segments which then the intervening DNA is “thrown away”
• Recombining VDJ segments generate TCRs with a variety of antigen-binding specificities and increased genetic diversity is gained by alternative joining of the D sequences: VJ, VDJ, or VDDJ chains
o TCRs undergo somatic mutation (up to 6 nucleotides can be added to the junction between V and D or D and J) but do not undergo somatic hypermutation
• If T cells had hypermutation this would lead to autoimmunity ☹
o Since there are two possible α chains and one β chain, there would be two αβ TCR sets that may be expressed on any given T cell
• How is only one αβ TCR set chosen? Thru positive and negative selection in the thymus
• Only ~2-5% of all T cells will survive this process
o Intimately associated with the TCR is CD3 and CD3 serves as a transduction signal for the T cell
• CD3 molecules have cytoplasmic signal transduction domain called ITAM (immunoreceptor tyrosine-based activation motif)
• Significance: when a T cell binds correct antigen and MHC with its TCR, the actual signal that turns the T cell on is transmitted through CD3 → this process needs to be controlled!
• Process controlled through two ‘hits’: 1) TCR/CD3 complex, 2) DC cytokines

46
Q

• Compare and contrast the antigen receptors of T and B cells.

A

o The antigen binding sites for T and B cells are found on TCRs and antibodies, respectively
o T cells undergo somatic mutation whereas B cells undergo somatic hypermutation as well
• Thus, antigen binding for B cells can change throughout the course of the B cell’s existence but for T cells this does not change once the T cell leaves the thymus
o B cells can change their receptor types via class switching; however, T cells will express their one TCR once they leave the thymus
o B cells can recognize antigen without the need for other factors. T cells recognize antigen only when the antigen is processed and presented to the T cell through the use of the MHC
o Because T cells see antigen only when it is complexed with cell-surface MHC molecules, T cells focus their attention on cell surfaces, and do not interact with free antigen; that is a job for the B cell and its antibodies

47
Q

• Discuss the structures recognized by T cell receptors. Distinguish between what is recognized by helper and cytotoxic T cells. Explain the special role of dendritic cells in this process.

A

o All T cells bind antigen when it is presented by an APC
o The APC presents the antigen to the T cell by having the antigen attached to an MHC molecule
o MHC (major histocompatibility complex) molecules, aka human leukocyte antigen (HLA), are subdivided into three different classes: I, II, and III
• Class I MHC molecules present antigen to CD8+ T cytotoxic T cells.
• Class II MHC molecules present antigen to CD4+ helper T cells.
• Other MHC proteins have been grouped together as the Class III MHC molecules and include a diverse group of proteins: complement proteins, TNFα/TNFβ, various enzymes, HSPs, etc.

48
Q

• Describe how Class I and Class II MHC molecules bind antigenic peptides.

A

o Class I MHC presents antigen to CD8+ CTLs.
• If there are appropriate co-stimulatory signals & cytokines produced, CTLs can be activated.
• Activation of CTLs results in the death of cells expressing the recognized antigen.
• Structure: β2 microglobulin; transmembrane protein
• Binding cleft: closed at both ends; 8-10 amino acid peptide
o Class II MHC presents antigen to CD4+ helper T cells.
• If there are appropriate co-stimulatory signals and cytokines produced, activation of the helper T cells may occur.
• Activation of helper T cells may result in the activation of B cells to proliferate and differentiate into plasma cells.
• Activation of helper T cells may result in the activation of CD8+ CTLs to proliferate & attack antigen bearing cells or in the ability of B cells to become antibody-producing plasma cells
• Each Class II MHC gene locus encodes two peptides, an α subunit and a β subunit that are expressed together on the cell surface
• Binding cleft: open at both ends; 13-18 amino acid peptide

49
Q

• Describe the CD4 and CD8 co-receptors and their functions.

A

o Both CD4 and CD8 T cell co-receptors contain Ig-like domains
o CD4 molecule: four Ig-domains
• Binds to:
• Conserved region on Class II antigen (internalized)
• Signal transduction molecule p56lck and forms a bridge that also binds to the ζζ chains of CD3
o CD8 molecule: one Ig-domain
• αβ heterodimer or αα homodimer with one Ig-like domain held together by a disulfide bond
• Binds to conserved region of Class I antigen (intracellularly made)
o Affinity of TCR for MHC/peptide is relatively modest; however, the presence of CD4 or CD8 adds to binding affinity
• Other co-receptors that add to binding affinity:
• CD2 binds to LFA-3 (co-stimulatory molecule)
• LFA-1 (leukocyte functional antigen (integrin)) binds to ICAM-1 (intercellular adhesion molecule).
• CD28 binds to B7 (aka CD80/CD86)
• CD45R binds to CD22
o CD4 Signaling Pathway: ultimately, leads to activation of NFkB and AP-1 which produces IL-2

50
Q

• Discuss what is meant by “MHC-restriction”. Name the classes of MHC molecules by which CTL and helper T cells are restricted.

A

o MHC restriction = the fact that a given T cell will recognize a peptide antigen only when it is bound to a host body’s own MHC molecule
• This is because the T cells are trained to recognize their own body’s MHC molecules and if the T cells were transplanted to another person, they would not be able to recognize that person’s MHC molecules on their APCs thus adaptive immunity would not be triggered
• MHC restriction is caused by gene loci polymorphism (see below)
o Class I → CD8+
• Class I products are on all nucleated cells (except mature RBCs)
• Antigen is presented from proteins synthesized within the cell itself; can derive from abnormal molecules, especially from internal pathogens such as virus-encoded molecules
o Class II → CD4+
• Class II products are expressed on the surfaces of dendritic and macrophage-type cells, B cells, and others all of which present antigen to Th cells: Th1, Th2, Th17, Treg, Tfh
• Antigen is endocytosed by DC and presented to a Th cell
o Note, DCs are special in that they can present both MHC I and II at the same time! This is called cross presentation
o Other APCs:
• DCs are most effective at presenting antigen due to:
• Constitutively expressing high levels of Class II MHC molecules
• Constitutively expressing B7 and other costimulatory molecules
• Macrophages must be activated by phagocytosis to express MHC II and costimulatory molecules
• B cells constitutively express MHC II but must be activated by antigen binding to antibody before expressing costimulatory molecules

51
Q

• Describe genetic polymorphisms related to MHC molecules.

A

o Each MHC Class has Subclasses:
• MHC I: three “classical” class I gene loci (with two different possible alleles, Mom/Dad)
• MHC Class I gene A encoding HLA-A
• MHC Class I gene B encoding HLA-B
• MHC Class I gene C encoding HLA-C
o Each of the Class I gene products associates with another peptide known as the β2 microglobulin, when expressed on the cell surface
o Therefore there is potential for 6 different MHC I combinations
• MHC I polymorphisms: most found in the cleft region (antigen-binding site)
o 1448 A alleles
o 1988 B alleles
o 1119 C alleles
• MHC II: gene loci encode two peptides - α subunit and a β subunit
• MHC Class II gene DP encoding DP αβ
• MHC Class II gene DQ encoding DQ αβ
• MHC Class II gene DR encoding DR αβ
o Therefore there is potential for 12 different MHC II combinations
• MHC II polymorphisms:
o DR locus: 2 DR α alleles, 860 DR β1 alleles, ~71 DR β2-9 alleles
o DQ locus: 47 DQ α1 alleles, 126 DQ β1 alleles
o DP locus: 17 DP α1 alleles, 134 DP β1 alleles
o Each gene locus is polymorphic which gives each person his/her unique identity and permits the recognition of self vs. non-self (also is an impediment to organ transplantation) and also affects:
• the ability to make an immune response
• the resistance or susceptibility to infectious diseases
• the susceptibility to autoimmune diseases and allergies
o Medical Significance of MHC polymorphisms:
• HLA-B27 individuals are 90x more likely to develop ankylosing spondylitis (destruction of the vertebral cartilage).
• Also linked to psoriasis, inflammatory bowel disease, and Reiter’s syndrome.
• HLA-DR2 individuals are 130x more likely to develop narcolepsy.
• Also linked to multiple sclerosis, hay fever, and SLE.
• HLA-A3/B14 individuals are 90x more likely to develop hemochromatosis (too much iron adsorption which can lead to internal organ damage).
• HLA-DQ2/GQ8 is linked to Celiac disease.
• HLA-DR3 is linked to diabetes mellitus type I, Grave’s disease
• HLA-DR4 is linked to rheumatoid arthritis and diabetes mellitus type 1
• And others…

52
Q

• Be able to describe the specific functions of folate and vitamin B12, and relate these functions to the therapeutic effects of METHOTREXATE and TRIMETHOPRIM.

A

o Functions: Folate and Vitamin B12 are required for DNA synthesis and for maintenance of neurons and RBCs; targets for antineoplastic drugs
• Folate is the critical precursor in neosynthesis of TH4 (Tetrahydrofolate)
• TH4 is a 1 carbon donor in several reactions including:
o Thymidylate and purine synthesis
o Methionine synthesis
o Amino acid metabolism
• Vitamin B12 an intermediate in:
• conversion of methylTH4 to TH4 – homocysteine is converted to methionine
• conversion of methylmalonyl CoA to succinyl CoA
o Therapeutic effects of MTX and Trimethoprim:
• MTX – blocks DHFR from converting folate into TH4, the active form of folate which is essential for both purine and pyrimidine synthesis
• Therapeutic uses: cancer, immunosuppressant (RA, psoriasis, IBD/Crohn’s disease), antibiotic, abortifacient (given in 1st trimester in combo with misoprostol – PGE1)
• Related drugs: Pemetrexed, Pralatrexate
• Trimethoprim – is a folic acid inhibitor and is a selective, competitive inhibitor of bacterial DHFR; commonly used in conjunction with sulfamethoxazole (blocks dihydropteroate synthetase) and this combo synergistically blocks successive steps in folate synthesis pathway in bacteria
• Related drugs: Pyrimethamine (antimalarial drug)

53
Q

• Explain the mechanism by which the folate transporter & reduced folate carrier are involved in the differential sensitivity of cancer cells & bacteria to folic acid inhibitors.

A

o Folate Transporter
• High affinity for folate, low capacity system due to potocytosis (essentially endocytosis)
• Occurs at physiological [ ]s
• Features: folate taken up&raquo_space;» MTX
• Note, some cancers overexpress Folate receptor → cancer cells are more sensitive to MTX than normal cells (differential sensitivity)

Reduced Folate Carrier
• Low affinity for folate, high capacity transport
• Important for pharmacological [ ]s
• Features: Leucovorin transported better&raquo_space; Folate&raquo_space; MTX
• Note, some cancers have decreased expression of RFC and thus have primary resistance to MTX; however, high doses of MTX can override this effect
• Subsequent administration of leucovorin allows normal cells with functional RFC to overcome the drug-induced inhibition of DHFR (by MTX) and is the basis for rescue therapy

o Folate storage is mediated by polyglutamination
• Polyglutamination increases the affinity for folate for target enzymes as well as keeping folate in the cell
• Key is thymidylate synthase
• Cancer cells are better polyglutinators than normal cells (differential sensitivity)

54
Q

o Consequences of Folate and Vitamin B12 deficiencies:

A
  • deficiency of either folate or vitamin B12 prevents DNA synthesis, with the greatest effect being on rapidly growing cells (esp. bone marrow and GI mucosa), and neurons
  • The earliest and most prominent clinical sign of deficiency is megaloblastic anemia
55
Q

• Folate Deficiency:

A
  • Etiology: insufficient dietary intake, high demand (pregnancy), alcoholism, intestinal disease (sprue), inhibitors
  • Results in: megaloblastic anemia, neural tube defects, other congenital abnormalities
  • Folate supplements will reverse anemia but not nervous system damage → look @ B12
56
Q

• Vitamin B12 Deficiency:

A

• Etiology: malabsorption of B12 due to deficiency of intrinsic factor (cannot treat orally)
• Results in: folate deficiency (no access to stored TH4), megaloblastic anemia (pernicious anemia if caused by impaired IF), and neurological damage (paresthesias, weakness → spasticity, ataxia, other CNS disorders)
• Correction of B12 def. can stop the progression of neurological effects, but not reverse them entirely
Folate Deficiency B12 Deficiency
RBCs Megaloblastic Anemia Pernicious Anemia
Neurons Neural Tube Defects Neurological Problems

57
Q

o Replacement Strategies for folate and b12

A

• Folate:
• Folic acid = synthetic (more stable) form of folate used in supplements
o Administration: oral available; IM, IV, or SC if GI absorption impaired
• Leucovorin = naturally occurring compound that replaces folate in rescue therapy
o Leucovorin does not require DHFR for conversion to TH4
• Vitamin B12:
• Naturally occurring forms: adenosylcobalamin, methylcobalamin
• Medicinal forms (give IM, IV, transdermal, spray, rarely oral due to lack of IF):
o Hydroxocobalamin: tx for cyanide poisoning; binds plasma proteins well → remains in circulation longer
o Cyanocobalamin
o Controversy over folate fortified foods:
• Folate fortification decreases the incidence of neural tube defects, but makes the detection of vitamin B12 deficiency more difficult in millions of elderly people by eliminating anemia which pre-stages the neurological damage

58
Q

• Describe the epidemiology of Malaria.

A

o Most prevalent in Africa, Asia, Latin America
o 216 million affected, 655,000 die each year
o 90% of deaths occur in Sub-Saharan Africa: 1 child dies from malaria each min in Africa
o 1,500 cases in US each year
o Transmission: mosquito bite
o Vector: anopheles mosquito (females)
o Tropism: hepatocytes and RBCs

59
Q

• Know the major relative differences between the four species of plasmodia in terms of parasite burden, degree of anemia, and relapse and fatality rate.

A

• Most common = P. falciparum (Africa), P. vivax (elsewhere)
• Most deadly = P. falciparum
• Relapses = P. vivax and P. ovale
page 19

60
Q

• Describe the life cycle of plasmodia, including the names of the different parasite forms in the hepatic, erythrocytic, and mosquito stages. Which species form hypnozoites, and why is this important?

A

o Life cycle takes place in both humans (asexual) and mosquitoes (sexual):

• Human (asexual):
Roughly 50 sporozoites are needed to initiate a malarial infection
The sporozoites enter the liver and infect hepatocytes and form hepatic schizonts (dividing sporozoites → multiple merozoites upon rupture)
Merozoites enter the blood and infect RBCs:
• Most species have a ring form as the beginning of RBC infection
• Then form trophozoites
• Then a schizont (forming multiple merozoites)
• Then merozoites are released from RBCs into the blood stream
• The merozoites can then become either male or female gametocytes
• Mosquito (sexual):
Male merozoites undergo exflagellation and become micro-gametes
Female merozoites form macro-gametes
Then male and female merozoites sexually combine to form sporozoite → start of cycle again
o Blood smear will show the different species of plasmodium in the forms of trophozoites, schizonts, and gametocytes:
• P. falciparum: gametocytes are banana/sickle-shaped
• P. ovale and P. vivax: Schüffner’s dots in cytoplasm throughout different life cycle stages

61
Q

• Be able to recognize, in general, the trophozoite, schizont, and gametocyte stages of plasmodia. Know the specific appearance of the gametocyte stage in Plasmodium falciparum.

A

o Obtaining a blood smear of the different plasmodium species results in visualization of the different life stages each of these species undergo:
• These are the main differences between the species:
P. falciparum: gametocytes are banana/sickle-shaped
P. ovale and P. vivax: Schüffner’s dots in cytoplasm throughout different life cycle stages
• Trophozoite stage: ring forms, RBCs express PFEMP-1
• Schizont stage: multiple dark-staining granules
• Gametocyte stage: single, small ‘nucleus’

62
Q

Know the mechanisms that make Plasmodium falciparum a more dangerous species

A

o P. falciparum is able to infect red cells of any age!!!!
o It causes red cell pathology:
• “Rosettes” = infected cell surrounded by normal RBCs
• Abnormal binding to endothelium = plaque formation (blood flow is impeded)
• Main cause of death in children = cerebral ischemia (due to blood flow occlusion)
o Stimulates high production of cytokines such as: TNF, IFNγ, IL-1
• These cytokines suppress red cell production, cause fever, tissue damage, and red cell binding to endothelium
o Plasmodium species enter RBCs through binding to glycophorin and once inside can make trophozoites (these produce PFEMP-1 (‘knobs’) that bind to endothelial cells)→ schizonts → merozoites → further systemic infection

63
Q

• Know the typical clinical symptoms of malaria, and know which species are characterized by quotidian, tertian, and quartan fevers.

A

o Incubation period = 1-2 weeks
o Prodrome: Flu-like illness
o Paroxysms: fever/chills, sweating, myalgia
• Quotidian (daily) – P. falciparum
• Tertian (every 48 hours) – P. vivax or ovale
• Quartan (every 72 hours) – P. malariae
Caused by RBCs breaking open and releasing merozoites
o Signs:
• Splenomegaly due to parasitic RBCs, super-active macrophages
If chronic this will result in fibrosis, grayish color (due to hemozoin pigment deposition)
• Hepatomegaly with grayish pigmentation
• Cerebral occlusion and ischemia → red cell rosettes, hypoxia around blood vessels, ischemia
• Heart and lungs may also be involved/affected

64
Q

• Understand the rationale for prophylactic treatment and the ‘radical cure’

A

o Prevention is key: use netting and insect repellant; avoid mosquito-malaria endemic areas, especially during periods of high insect activity
o Prophylactic treatment for malaria is advised for individuals traveling to a malaria endemic area because preventing the disease is much easier than treating the aftermath
o If contract disease, treat it with the appropriate anti-malarial drugs (resistance dependent – see selectivity basis above); use ‘radical cure’ if indicated
• ‘Radical Cure’ involves eradicating the persistent (latent) forms of plasmodium if the patient is infected w/ P. vivax or P. ovale since these forms persist in hepatocytes
These patients are treated with primaquine

65
Q

• List the major types of leukemia and lymphoma. Know the main difference between leukemia and lymphoma.

A

o Leukemias:
• Start in bone marrow and spreads to blood and nodes
• Myeloid or lymphoid
• Acute or chronic
o Lymphoma:
• Starts in lymph nodes and spreads to blood and marrow
• Lymphoid only
• Hodgkin or Non-Hodgkin
o Myeloid vs. Lymphoid
• Myeloid: AML, CML
• Lymphoid: ALL, CLL; Hodgkin’s, Non-Hodgkin’s Lymphoma; Multiple Myeloma
• Plasma cell disorders: Multiple myeloma

66
Q

• Know how a bone marrow biopsy is performed, and what samples are obtained.

A

o Bone marrow biopsies are performed by using a Jamshidi needle that is injected into the iliac crest
• Obtain bone marrow aspirate through the Jamshidi needle with an aspirate syringe
• Look for cellularity vs fat percentage (more fat seen in elderly patients)
• Stain with 1) Wright-Giemsa stain, 2) cytochemical stain (non-specific esterase, NSE) = monocyte stain

67
Q

o Acute Leukemia

A

• Sudden onset
• Can occur in either adults or children
• Rapidly fatal without treatment
• Composed of immature cells (blast cells)
Look for nucleoli with light-staining cytoplasm
• Types: ALL, AML

68
Q

o Chronic Leukemia:

A
  • Slow onset
  • Occurs only in adults
  • Longer course
  • Composed of mature cells
69
Q

• Overall Review of Acute Leukemias: ALL, AML

A

o Definition: malignant proliferation of immature myeloid or lymphoid cells in the bone marrow
o Etiology:
• Clonal expansion
• Maturation failure – stop progressing at a certain stage of maturation → classification system
o Pathophysiology: crowd out normal cells, inhibit normal cell function, infiltrate other organs
o Clinical Findings:
• Sudden onset (days) → bone marrow full of cells (lack of fat)
• Sx: bone marrow failure to make normal cells – fatigue, infections, bleeding (epistaxis, etc.)
• Bone pain (expanding marrow)
• Organ infiltration (liver, spleen, brain)
o Lab findings:
• Blasts/immature cells in blood
• Leukocytosis (↑WBC)
• Anemia
• Thrombocytopenia

70
Q

o Essentials for AML

A

• Malignant proliferation of myeloid blasts in blood, bone marrow
• 20% cutoff for diagnosis – at least 20% of nucleated cells are blasts
Burden of disease is followed by ‘blast’ count - % of blasts
• Many subtypes
• Bad prognosis

71
Q

o How to determine if a leukemia is myeloid:

A
  • Dysgranulopoiesis – granulocytes have funny morphology
  • Auer rods – primary granules → AML!!!!!
  • Cytochemistry
  • Immunophenotyping
  • Cytogenetics: AML translocation (8:21), etc.
72
Q

• t(8;21)

A

(AML-M2); cells become huge

good prognosis

73
Q

• t(15;17)

A

AML-M3); ALL cases have this translocation

good prognosis

74
Q

• inv(16)

A

(AML-M4, Eo); gigantic eosinophils

good prognosis

75
Q

• 11q23

A

doesn’t indicate what problem is occurring; confers a bad prognosis
11 q two three → two = band, three sub-band
Usually seen with AML with a monocytic series

76
Q

• FLT-3 Mutation

A

Present in 1/3 of cases of AML!
Monocytic cells
Poor prognosis

77
Q

• Know the morphologic characteristics of myelodysplasia (in general - not all the subtypes) and describe the treatment options.

A
o	Myelodysplastic Syndrome (MDS)
•	Problem: abnormal stem cells
•	Dysmyelopoiesis
•	Maybe ↑ blasts (not up to 20%...)
•	May evolve into acute leukemia
•	Clinical and Lab Findings:
 	Older patients
 	Asymptomatic, or BM failure → (leukopenia, thrombocytopenia, anemia)
 	Macrocytic anemia
 	Dysplasia: 
•	Red cells: megaloblastic nuclei, fragmentation
•	Neutrophils: hypogranulation, hyposegmentation
•	Megakaryocytes: small, non-lobulated cells
•	Treatment and Prognosis
 	Low-grade: support, follow.
 	High-grade: be aggressive
78
Q

• Important Things to Know: ALL

A

o Malignant proliferation of lymphoid blasts in blood, bone marrow
o Classified by immunophenotype (B vs. T)
o More common in children
o Prognosis often good; based on immunophenotype classification
• T-lymphoblastic leukemia: Worse prognosis.
aka T-lymphoblastic lymphoma
Teenage male with mediastinal mass
TdT + (only present in lymphoblasts!)
WBC usually ↑↑↑
Bad prognosis
• B-lymphoblastic leukemia: Better prognosis
aka B-cell lymphoblastic lymphoma
Several sub- and sub-subtypes
TdT + (only present in lymphoblasts)
Rarely see Philadelphia chromosome (9:22 translocation); commonly seen in CLL
o Classified by presence of TdT since only found in lymphoblasts
o Treatment
• Chemo ± bone marrow transplant
• Many children are ‘cured’!
Typically 5-10 years without disease
o Prognosis
• Immunophenotype (T is bad; B is good)
• Age (1-10 good)
• WBC (<10,000 good)
• Cytogenetics (hyperdiploidy good!) → more than one set of chromosomes