• 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

1/838

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;

838 Cards in this Set

  • Front
  • Back
Bone marrow and all blood corpuscles
Liver
Spleen
Lymph Nodes
Thymus
Tissues of the Hematopoietic System
These tissues are embryologically related
Hematologic disorders often affect all of these tissues (lymphoma, leukemia, hemochromatosis, myelofibrosis).
Hollow space of bones filled with all blood cell precursors
Infant: Activity in all bones from skull to feet. Cellularity is 100%.
Cellularity decreases by 10% with each decade of life until age 70-80, when cellularity remains at 20-30%
Bone Marrow
Normal myeloid to erythroid ratio is 3:1
Controlled growth with increase in activity based upon demand
Infection
Low tissue oxygenation
Activity is decreased when demand is met
Mediated by growth factors
Increase in production results in greater numbers of mature cells and some young cells, but no immature cells are released.
Bone Marrow: Normal Production
Bone Marrow - What is normal production?
2.4 billion white blood cells
10 billion red blood cells
175 billion platelets
Stromal cells
Fibroblasts
Fat cells
Endothelial cells
Adhesion molecules
Growth factors
Stromal Matrix
Sites of Embryonic Hematopoiesis
First 6 weeks --Yolk sac
6-18 weeks -- Liver
18-30 weeks -- Liver and spleen
30 weeks to birth to 8 week old infant -- Liver, spleen, and bone marrow
>10 weeks -- Bone marrow only
Cell size
-Decreases with maturation
Nuclei
-Always round
-Chromatin condenses with maturation
-N:C ratio decreases with maturation
Cytoplasm
-Basophilia: immaturity
-Magenta: as maturation occurs and hemoglobin accumulates
-No granules
Erythropoiesis
No nucleus
No cytoplasmic organelles
No protein or lipid synthesis
No oxidative phosphorylation
Picks up oxygen from the lungs
Delivers oxygen to the tissues
Picks up CO2 from the tissues
Delivers CO2 to the lungs
Red Cells: Function
Biconcave disk shape  large surface area  good for gas exchange
Highly deformable; allows changes in size
-8 microns in a large vein to 2 microns in a capillary
Red cell function is essential for the function of the rest of the body
Red Cell: Shape
Polys
PMN (Polymorphonuclear neutrophil)
Seg (segmented neutrophil)
3 lobes separated by a thread
Neutrophil
Myeloblast

Promyelocyte

Myelocyte

Metamyelocyte

Band

Neutrophil
Stages of Neutrophil maturation
Granules contain enzymes involved in oxidative and non-oxidative killing of bacteria and fungi
-Particularly bacteria
Circulate in the blood and tumble along endothelium, loosely adhering
-Circulating pool
-Marginating pool
Neutrophil - function
Bi-lobed nucleus
Eosinophilic granules
Parasitic infections
Allergic reactions
Vasculitis
Some hematologic malignancies
Eosinophils
(Mast cell)
Basophilic granules
Release histamine
IgE
Increased in myeloproliferative disorders
Basophils
Circulate in the bloodstream for only 24 hours
Then go into tissues to become macrophages
Ingest fungi, mycobacteria, and play a role in battling pyogenic bacteria
Monocyts
No morphologically distinct stages of maturation
Lymphoblast
Intricate acquisition and loss of surface antigens takes place as cells mature and differentiate in to B, T, and NK cells
Lymphocyte
Produced in the bone marrow
Migrate to other sites of the body to mature and acquire specific properties
T cells (thymus): helper and suppressor cells
B cells (bursa of Fabricius in birds): produce antibodies against foreign antigens (plasma cells)
NK (natural killer) cells: large granular lymphocytes
Lymphocyte: Development
Very little cytoplasm
Nucleus is about the same size as a red blood cell
Life span is years
Peripheral blood:
70% T cells
25% B cells
< 5% NK cells
no plasma cells
Lymphocyte
CD10
CD19
CD20
CD79a
sIg kappa/lambda
B-Cell Antigens
CD16
CD56
NK-cell antigens
CD3
CD4
CD5
CD7
CD8
T-cell antigens
Megakaryocytes are multinucleated
Largest cells in the body
Do not circulate
Filtered out by lung microvasculature
No intermediate maturation stages
Platelets brak off from megakaryocytes
Filters blood
Examines blood cells
and destroys injured
erythrocytes and cells that have been sensitized by IgG and complement
Activates complement
Extremely important in helping clear encapsulated organisms from the blood
-Streptococcus pneumoniae
-Haemophilus influenzae
-Neisseria meningitidis
Spleen - function
During periods of extensive red cell damage and splenic activity, blood may enter the spleen but be unable to exit (sequestration)
~10% of the population will have an accessory spleen
Spleen - function
Spleen:
Lymphocytes around arterioles
White pulp
Spleen:
splenic sinusoids
Red pulp
2 alpha chains (pink)
2 beta chains (yellow)
Aka: non-alpha chains
4 heme molecules (blue balls)
The hemoglobin molecule
A ring structure called protoporphyrin IX
Has an atom of divalent/ferrous iron (Fe+2) attached
One group per chain
Each combines reversibly with one molecule of oxygen
What makes blood red
Heme
2 pairs of polypeptide chains (4 total)
Each chain has ~140 amino acids
Alterations in the amino acid sequences results in different globin chains
Globin chains
Chromosome 16
The alpha globin genes

Humans have two copies of the a and g genes per chromatid for a total of 4 genes per person
Chromosome 11
The beta, delta, gamma glogin genes

Humans have two copies of the a and g genes per chromatid for a total of 4 genes per person
Genes get transcribed to mRNA
mRNA gets translated to a globin polypeptide chain and then released into the cytoplasm from the ribosomes
Each globin chain binds with a heme molecule
Heterodimers are formed between an alpha chain and a non-alpha chain
Two heterodimers combine to form tetramers  hemoglobin
Hemoglobin formation
2 alpha chains and 2 beta chains
The main form of hemoglobin after birth
Hemoglobin A
2 alpha chains and 2 delta chains
Delta chains are not expressed efficiently
Normally only a small amount after birth
Hemoglobin A2
2 alpha chains and 2 gamma chains
In adults, this is only in a few RBCs, called F cells
Hemoglobin F
Have different mobilities in electrophoresis
Hemoglobin
Can also undergo post-translational modification
Reactions can occur with various sugars and the amino groups of the globin chains
-Results in glycated hemoglobin
-Monitor this to monitor diabetic patients
Hemoglobin A
Bind oxygen in the lungs
Transport it to tissues
Unload oxygen in the tissues
Hemoglobin Function
affinity for oxygen depends on the partial pressure of oxygen (pO2)
Hemoglobin
Sigmoid shaped
At low oxygen tension, Hb has a low affinity for oxygen
At high oxygen tension, Hb has a high affinity for oxygen
As each heme group binds oxygen, the avidity increases
Hemoglobin - obtaining oxygen
2,3-bisphosphoglycerate (2,3-BPG)
Normally found in RBCs
With high 2,3-BPG, Hb molecule goes from relaxed and oxygenated to tense and deoxygenated
Need a higher pO2 to saturate the same amount of Hb
Shifts curve to right
Bohr effect: a shift of the curve as a result of pH
Low pH (acidic environment)
Shifts curve to right
Has increased oxygen affinity compared to Hb A
Extracts oxygen from maternal circulation
Doesn’t release oxygen as easily as Hb A; so fetus needs more Hb to adequately oxygenate tissues
Hemoglobin F
Oxygen delivery process does not require energy consumption
Mature erythrocyte does not have a nucleus, mitochondria, and other organelles
Can’t synthesize proteins or lipids
Can’t undergo oxidative phosphorylation
(which forms ATP, which generates energy)
RBC Metabolism
So how does the RBC generate energy for the metabolic pathways that it needs for survival?
Embden-Meyerhof pathway
Anaerobic glycolytic pathway
Glucose enters RBC through facilitated membrane transport system
Glucose gets metabolized to lactic acid
Requires 2 molecules of ATP per glucose molecule
Generates up to 4 molecules of ATP per glucose molecule
Hexose monophosphate shunt (aerobic glycolysis)
Without (enzyme), no NADPH is formed
Without NADPH, glutathione metabolism doesn’t occur – H2O2 doesn’t get converted to water
Results in oxidative damage
(and RBCs carry large amounts of oxygen)
G6PD deficiency is the most common enzyme deficiency in the world
Obtaining blood samples
Venipuncture, port, PICC (peripherally inserted central catheter)
Blood collections
Different tubes have different additives
Some allow blood to clot, others don’t, etc
Automated
Results in minutes
Quantity of each major cell type
Differential:
-Percentage of different types of white cells
“Flags” abnormal values
Complete Blood Count (CBC)
Red cells are highest
At birth (RBC, Hb, Hct)
-30% are Hb F

Number of red cells remains stable until puberty, then male values exceed female
Number of red cells does not decrease as part of aging
Value is measured in grams per deciliter
Tells you how much hemoglobin (in grams) is in one deciliter of blood
Hemoglobin
Value is a percentage
Tells you the volume of packed red blood cells in a given volume of whole blood
Hematocrit
tells average weight of hemoglobin per cell
MCH - Mean corpuscular hemoglobin
Tells average concentration of hemoglobin per cell
MCHC - Mean corpuscular hemoglobin concentration
Tells us the size of the red blood cell
MCV - Mean corpuscular volume
Tells us the how varied the sizes of the red cells are
RDW - Red cell distribution width

If RDW is normal, all cells are similar size (whether they’re all big or all small)
If RDW is increased, cell size is NOT known
Larger than mature RBC; with less central pallor
Not part of CBC; need to order separately
Reticulocyte count

Measures the percentage of reticulocytes circulating in the blood
Methylene blue stain identifies precipitated RNA in young cells
Order of percentage of all white cells
Neutrophils (segs and bands)
Lymphs
Monos
Eos
Basos
Others
Too few platelets
Thrombocytopenia
Too many platelets
Thrombocytosis
Low MCH
Hypochromia
Iron deficiency anemia
Hemoglobinopathies (sickle cell, thalassemias, etc)
Anemia of chronic disease
Copper deficiency
Lead poisoning (decreased heme synthesis)
Low MCV - microcytic anemia
Punctate basophilic precipitation of undegraded RNA
-A sign of ineffective hematopoiesis
-Seen in lead toxicity
Basophilic stippling
Result of redundant red cell membrane/ decreased cell volume
Hemoglobinopathies/thalassemias
Iron deficiency anemia
Drug-induced hemolytic anemia
Liver disease
Target cells - also called sombrero or Mexican hat cells
disseminated intravascular coagulation, TTP
Schistocyte (helmet cell)
Liver disease, artifact
acanthocyte
French for “rolls” as in rolls or stacks of coins
RBCs abnormally adhere to each other due to increased immunoglobulin production
-Multiple myeloma, plasma cell leukemia, infection
-Artifact
Rouleaux formation
Remnants of nuclear chromatin normally removed by the spleen
Seen in surgically or functionally asplenic patients and patients on dialysis
Howell Jolly Bodies
No central pallor
Seen in hereditary spherocytosis
Spherocytes
Bone marrow biopsy and aspirate - where?
Posterior, superior iliac crest (pelvis) or breastbone
Needle/liquid withdrawal
Aspirate
Pulled out like soil sample
Core biopsy
Aspirate sampling
Flow cytometry
FISH
Cytogenetics
Microscope
Core sampling
Look at under microscope
Sodium
Potassium
Chloride
CO2
BUN
Creatinine
Glucose
Calcium
Basic metabolic panel
Sodium
Potassium
Chloride
CO2
BUN
Creatinine
Glucose
Calcium
Total Protein
Albumin
AST
ALT
Alkaline Phosphatase
Total Bilirubin
Comprehensive metabolic panel
Use to separate and quantitate serum protein including hemoglobin
Separates different proteins based upon their physical and chemical properties
Mobility of protein depends on molecular weight and charge
Electrophoresis
Serum Proteins:
Albumin (main component)
Microglobulins:
-Alpha 1 (alpha 1 antitrypsin), Alpha 2
-Beta 1, Beta 2 (predicts change in immunoglobulin production; used to assess disease activity of multiple myeloma)
-mmunoglobulins: IgG, M, A, >>>E, D
Electrophoresis - SPEP
Red cells are lysed, Hb is separated and applied to a gel. A current is applied.
Different hemoglobins migrate at different speeds across the gel
Electrophoresis - Hemoglobin
RBC mixed with reducing agent and observed under microscope for characteristic change in shape.
Sickle cell "screen)
takes place within macrophages outside vascular stream
-Physiological conditions
-Low-grade chronic hemolytic states
Extravascular hemolysis
major red cell lysis occurs within the circulation
-15% of Hb catabolism follows this pathway
-Transfusion reactions
Intravascular hemolysis
Primary site of RBC destruction
In patients without organ macrophages of other organs assume this function
-Liver
-Bone marrow
Spleen
Cells may be retained if:
Shape is less adaptable (spherocytes)
Membrane is less flexible (older cells)
There are inclusions or particles stuck to the membrane
-Heinz bodies (denatured Hb)
-Howell-Jolly bodies (nuclear remnants)
Spleen
Hemoglobin (Hb) binding glycoprotein made in the liver
It is an acute phase reactant
Absent in the newborn
Haptoglobin (Hp)
Low or absent plasma Hp is an indicator of
recent or ongoing intravascular hemolysis
Rapid saturation of Hp and rapid clerance of HB/Hp complex during
Massive intravascular hemolysis
degraded into a.a for recycling
Globin and Hp

The Hp/Hb complex is internalized by the hepatocyte
Catabolized to bilirubin and iron
Heme

The Hp/Hb complex is internalized by the hepatocyte
Free excess of heme is bound to (2 things) and taken by hepatic receptors for catabolism
Hemopexin and methemalbumin
Hb appears in the urine (haemoglobinuria)
-Can precipitate and cause ARF
Massive intravascular hemolysis

Some free Hb is reabsorbed into cells of the proximal tubules
ferritin accumulates in the tubular cells
-Hemosiderin via Prussian blue staining
Low chronic intravascular hemolysis

Some free Hb is reabsorbed into cells of the proximal tubules
Shorter life-span of erythrocytes

Compensatory increase in erythrocyte production (reticulocytosis)
Hemolytic anemia
Intrinsic red cell abnormalities
Extrinsic causes of hemolysis
Immune mediated hemolysis
Non-immune mediated hemolysis
Hemolytic anemia
-Red Cell Membrane Disorders
Hereditary Spherocytosis
Hereditary Elliptocytosis
-Red Cell Enzyme Disorders
G6PD Deficiency
Pyruvate Kinase Deficiency
-Hemoglobin Disorders
Unstable Hemoglobins
Methemoglobinemia
Thalassemia
Sickle Cell Disease
Intrinsic red cell abnormalities
-Immune mediated hemolysis
Warm Reactive Autoimmune Hemolytic Anemia (AIHA)
Cold Agglutinin Disease
Paroxysmal Cold Hemoglobinuria (PCH)
-Schistocytic hemolytic anemia: Mechanical destruction of RBC as they travel
Hemangiomas (Kasabach-Merritt syndrome)
Prosthetic heart valves
Microangiopathic hemolytic anemia (MAHA)
--TTP
--HUS
Extrinsic Causes of Hemolysis
Pallor
Icterus, jaundice
Fatigue
Splenomegaly
Gallstones
Cholecystitis
Dark urine
May present with parvovirus associated aplasia
Family history
Clinical Presentation of Hemolytic Anemia
Increased Nutritional Requirements of Chronic Hemolysis
Greatest requirement is for folic acid and lowest body stores (10 day storage)
Amino acid to make globin chains (normal nutritional status)
B12 (10 years storage)
No increase Fe requirement since it is “recycled” and stored in bone marrow
Purpose: to identify the presence of immunoglobulins or complement on the surface of the red cells.
Sample from the patient is mixed with reagent that has antibodies against human immunoglobulins and complement.
If agglutination forms, the test is +.
Direct Coombs (Direct Antiglobulin Test)
If the test is positive, further tests are done.
Three main patterns:
IgG only on surface of RBC
IgG and C3complement
C3 only on surface cells, antibody on cells may be IgM
Direct Coombs Test
Testing patient’s serum for antibodies
Indirect Coombs
Normal to have antibody to major blood groups but not to minor blood groups unless prior transfusion (or through exposure due to baby’s blood during pregnancy or delivery)
IgG mediated
Extravascular clearance primarily via the reticuloendothelial system (spleen)
May be idiopathic or associated with SLE, lymphoid malignancies, immunodeficiency
Warm Reactive Autoimmune Hemolytic Anemia (AIHA)
IgM mediated
Can be associated with Mycoplasma, EBV
Intravascular lysis
Cold Agglutinin Disease
Acute illness, often after viral URI
-Inciting infections include measles, mumps, varicella, syphilis, Mycoplasma
Caused by cold reactive IgG (Donath Landsteiner Antibody)
Intravascular hemolytic anemia
Paroxysmal Cold Hemoglobinuria (PCH)
Management of hemolytic anemia
Observe growth, development
Determine baseline hemoglobin
Follow for splenomegaly
Educate family regarding risks for gallstones, parvovirus B19 aplastic crisis
Folate supplementation
Erythrocyte transfusions, intermittent vs chronic
Splenectomy
Cholecystectomy if symptomatic gallstones
Whole Blood Phlebotomy
Packed Red Blood Cells
Platelets
Plasma
Apheresis
Same as phlebotomy (except we can collect more)
White Blood Cells
-Granulocytes, Monocytes, T Cells, Stem Cells
Donation of 1 unit of whole blood (WB) yields:
1 unit of packed red blood cells (PRBC)
1 unit of plasma
1 unit of random donor platelets
Transfuse (1 or 2 units) to treat anemia
Hemoglobin (Hb) usually < 8 g/dL
Stored in the refrigerator
PRBC
Dose
-1 unit will increase the Hb by 1 g/dL
-Increase Hematocrit (Hct) by 3%
Pediatric
-10 mL/kg will increase Hb by 2 g/dL
-Increase Hct by 6%
PRBC
replacement of 1 total blood volume (usually 10 units of PRBC) in less than 24 hours
May be complicated by dilutional coagulopathy, hypocalcemia, hyperkalemia, arrhythmia
Massive transfusion
PRBC can be frozen
For ten years
Thrombocytopenia requiring platelet tranfusion
<20,000/microliter
Plasma frozen within 8 hours of phlebotomy
Will have normal levels of Factor VIII
Factor VIII levels degrade over several days
Factor VIII levels are 50-80% normal by 5 days
Fresh Frozen Plasma (FFP)
Plasma frozen within 24 hours of phlebotomy
Used for all plasma orders at Wake Forest
Now used interchangeably with FFP
24 hour plasma
Transfuse (2 units) to treat clotting factor deficiencies
Prolonged Prothrombin time (PT) and activated partial thromboplastin time (PTT)
Frozen – takes about 30 min to thaw
Plasma
Transfused (5 or 10 units) usually to replace fibrinogen
and provides a more concentrated form of:
-Fibrinogen
-Factor VIII
-von Willebrand Factor
-Factor XIII
-Fibronectin
Used to treat bleeding from deficiency of these factors.
-Safer, purified, virus inactivated or recombinant concentrates are now available for Hemophilia A and von Willebrand’s Disease
-It is NOT concentrated FFP
Cryoprecipitated AHF
(Anti-Hemophilic Factor) (Cryo)
present on all human red blood cells. Think of as the “O” antigen.

Is the precursor of the A & B antigens
RED BLOOD CELLS (“H” Antigen) Group O
Red blood cells "A" antigen
Galnac (added on top of the "H" antigen)
Red blood cell "B" antigen
Gal (added on top of the "H" antigen)
Anti-B antibodies
Group A person
Anti-A antibodies
Group B person
No ABO antibodies
Group AB person
Anti-A and Anti-B antibodies
Group O (H antigen) person
A person needs an exposure to foreign, non-self red blood cells to have an antibody response
-Pregnancy
-Blood transfusion
-?? Sharing needles ??
The D antigen
Antibodies to D antigen are NOT “naturally” occurring
test the PATIENT’S red cells for A or B antigen
forward type
test the PATIENT’S serum for the expected antibody
Reverse type
test the red cells for the D antigen
Rh type
check the PATIENT’S serum for other antibodies against red cell antigens
Other than ABO – such as Kell, Duffy, Kidd
Mix the PATIENT’S serum with a sample of DONOR red cells from the exact unit that we wish to transfuse
Crossmatch
If the red cells and serum mixture clumps, then the unit is INCOMPATIBLE
The endpoint of most blood bank tests is AGGLUTINATION... “clumping”
Used to evaluate hemolytic transfusion reactions or autoimmune hemolytic anemia
IgG or C3d
The Direct Antiglobulin Test (DAT) or Direct Coombs Test
Mother = Rh neg
Fetus = Rh pos
Fetal red cells enter maternal circulation
Mother synthesizes Anti-D
Anti-D crosses placenta and hemolyzes fetal red cells
Hemolytic Disease of the Newborn (HDN)
Anemia with possible organ failure, edema (Hydrops), jaundice
Cardiac failure and Liver failure
+DAT
Mother: High titer of Anti-D
Other antibodies to red cells can cause hemolytic disease of the newborn
Bilirubin & Bile Pigments in the amniotic fluid
Peripheral smear of baby
-Spherocytes
-Nucleated red cells
Hemolytic Disease of the Newborn (HDN)
Which is more common for bacterial/septic transfusion reactions - platelets or RBCs?
Platelets b/c not refrigerated
Acute hemolytic
Transfusion-related acute lung injury (TRALI)
Septic (Bacterial contamination of the product)
Anaphylaxis
Life Threatening - Rare transfusion reactions
Febrile non-hemolytic
Urticarial (mild allergic)
Circulatory overload
More common - benign transfusion reactions
Most common RBC reaction
Hemolytic
Most common platelet reaction
Septic
Most common plasma reaction
Transfusion-related acute lung injury (TRALI)
-Signs and Symptoms
Fever and chills – Most common
Renal failure
DIC
Back pain
-Laboratory
+ DAT
Hemoglobinemia
Hemoglobinuria
INCREASED: LDH ; Bilirubin
DECREASED: Hemoglobin / Hematocrit ; Haptoglobin
Peripheral Smear
Schistocytes or Spherocytes
Hemolytic transfusion reactions
Trying to reverse effects of hemolytic transfusion reaction
Early recognition & stopping the transfusion.
Fluid & blood pressure support if needed.
Prevent renal failure / maintain urine output
Intravenous fluids, diuretics, and renal dose dopamine if necessary
What is blood tested for?
HIV
HCV
Bacteria in platelets
West Nile Virus
HBV
HTLV
Syphilis
Chagas Disease
Now a regulatory requirement
Why does the patient need a transfusion?
There is no other alternative therapy
Risks of transfusion
Infectious diseases
HIV, Hepatitis, West Nile virus, many more...
Transfusion reactions
Administrative errors
Rare fatal reactions
Informed consent
Occurs after extrusion of nucleus from orthochromic normoblast
Remains in marrow for approx 3 days
Released into circulation – remodeled with loss of water and membrane
Normal are macrocytic
Reticulocytes
Reticulocyte shift with anemia
Normal (45% hematocrit) - 3.5 days in bone marrow and 1.0 days in blood

When hematocrit drops, reticulocytes stay in bone marrow for a shorter period of time and spend more time in blood
Mechanism unknown
Acetaldehyde
-can induce membrane changes
-Interferes with cellular division
Alcohol as a cause of macrocytic anemia
May be caused by increased lipid deposition on red cell membranes
Target Cells
Liver disease and macrocytosis
Autoimmune thyroiditis associated with antiparietal (think about HCl production in the stomach and the ability to convert protein to absorbable material) cell antibodies
Thyroid disease and macrocytosis
Artifacts that can occur to normal sized RBCs
Clumps of RBCs counted as single cells by automated cell counters
Spurious macrocytosis
National Cancer Institute definition: A group of diseases in which the bone marrow does not make enough healthy blood cells. Also called preleukemia and smoldering leukemia.
Hypolobulated or hypogranular neutrophils
Large and/or abnormally granulated platelets
Monocytosis
Occasional blast forms
Myelodysplastic Syndromes
Pronounced reduction in the number of erythrocytes, all types of leukocytes, and the blood platelets in the circulating blood.
Pancytopenia
Anemia, often pancytopenia, with macrocytic red blood cells and hypersegmented neutrophils due to an impairment in DNA synthesis
Inadequate conversion of deoxyuridylate to thymidylate
Slows DNA synthesis
Delayed nuclear maturation
Nuclear/cytoplasmic dyssynchrony (ie, immature nucleus/mature cytoplasm)
Megaloblastic anemia
Folate deficiency
Cobalamin deficiency
Drugs
-Inhibit absorption of B12 or folate
-Inhibit enzymes required for DNA synthesis
Interference with DNA synthesis
Characterized by defect in DNA synthesis resulting in unbalanced cell growth and impaired division
Immature-appearing nucleus, but mature cytoplasm, and large cells
Most common causes are B12 and folate deficiency
Megalobalstic Anemias
Sources of B12
Fish, eggs, poultry
Sources of folate
Fortified cereals, beef liver, cowpeas, spinach
Cobalamin absorption defect in stomach
Pernicious anemia (lack of IF)
Defect in IF
Partial or total gastrectomy
Gastric bypass
Atrophic gastritis
Acid-blocking drugs
Cobalamin absorption defect in jejunum
Bacterial overgrowth
Parasites
Sprue
Cobalamin absorption defect in ileum
Ileal resection
Crohn's disease
Imerslund-Grasbeck disease
Cbl-F disease
Cobalamin absorption defect in blood vessel
TCII deficiency
Cobalamin absorption defect due to pancreas
Chronic pancreatitis
Intake of cobalamin strictly through food
Strict vegetarianism w/o supplementation
B12 deficiency and MMA and homocysteine levels
MMA high
Homocysteine high
Folate deficiency and MMA and homocysteine
MMA - no
Homocysteine - high
A stage of cellular necrosis in which the fragments of the nucleus fragments and its chromatin are distributed irregularly throughout the cytoplasm.
Nuclear-cytoplasmic dyssynchrony
-Peripheral blood
Hypersegmented neutrophils
Oval macrocytosis with or without anemia
Thrombocytopenia and/or leukopenia with immature forms
Basophilic stippling, leukoerythroblastic changes
-Bone marrow
Hypercellular
Giant bands and metamyelocytes
Nuclear-cytoplasmic dyssynchrony
Open and immature nuclear chromatin pattern
Karyorrhexis
Blood chemistry
-increased indirect bilirubin
-increased lactate dehydrogenase
Hematologic abnormalities in Cobalamin or Folate deficiency
Karyorrhexis
Fragmentation of the nucleus whereby its chromatin is distributed irregularly throughout the cytoplasm; a stage of necrosis usually followed by karyolysis.
Considerable variation in the size of cells that are normally uniform, especially with reference to red blood cells.
Anisocytosis
Characteristic hematologic abnormalities seen in megaloblastic anemia
A, Macro-ovalocytes and marked anisocytosis are seen under low and high power. B, A hypersegmented neutrophil with at least six lobes. C, Megaloblastic pronormoblast (left) and megaloblastic polychromatophilic normoblast (right). D, Megaloblastic “giant” metamyelocyte (left) and band (right).
All proliferating cells exhibit (phrase), including epithelial cells lining the gastrointestinal tract (buccal mucosa, tongue, small intestine), cervix, vagina, and uterus.
However, such changes are most striking in the blood and bone marrow.
Megaloblastosis
Other clinical manifestations:
Glossitis
Secondary malabsorption caused by megaloblastic GI changes
Weight loss or growth failure
Infertility
Thrombosis
Hyperpigmentation
Immune deficiency
Cobalamin and Folate Deficiency
causes neuropsychiatric manifestations:
Peripheral neuropathies
Dorsal column involvement (loss of position and vibratory sense, ataxia)
Subacute combined degeneration of spinal cord
Psychiatric symptoms (dementia, psychosis)
B12 deficiency
Treatment of B12 deficiency
Parenteral 1000 micrograms/day for 1 week and and then 1000 micrograms/wk for 1 month; then 1000 micrograms/month for life for PA

Oral - 1000 micrograms/day for 1 month and then 125-500 micrograms/day for intake deficiency or 1000 micrograms/day for Pa
may normalize MCV and Hgb but allow neurologic manifestions to persist
Folate repletion without B12 repletion
Part 1: Give radioactive cobalamin by mouth. After 1 hour, give injection of unlabelled cobalamin by injection (“flushing dose”) to saturate plasma B12 binders so that labeled cobalamin will not bind but be excreted via the kidney
An individual with pernicious anemia would absorb little or none of the oral B12 since these patients lack intrinsic factor. The amount of radiolabeled B12 recovered in the urine would be less than normal.

Part 2: Give exogenous intrinsic factor with the radiolabeled cobalamin.
In a patient with pernicious anemia, this should correct the absorption and increase the urinary excretion into the normal range.
If still abnormal, then the small intestine can’t absorb the B12-IF complex
Schilling Test – measures cobalamin absorption
Schilling Test Results for B12
1. Prolonged inadequate intake of B12
2. Absent IF (PA)
3. Small-bowel disease interfering with absorption
4. Bacterial competition for B12
With IF Without IF
1. Normal Normal
2. Abnormal Normal
3. Abnormal Abnormal
4. Abnormal Abnormal
Anti-parietal cell antibodies
Anti-intrinsic factor antibodies
Low serum pepsinogen I
High serum gastrin
Schilling test, stage 1 and 2
Pentagastrin-resistant achlorhydria
Endoscopy with biopsy
Diagnostic tests for Pernicious Anemia
Diet lacking fruits and vegetables
Alcoholism
Sprue, Crohn's disease and others
Hemodialysis
Increased cellular proliferation (pregnancy, skin diseae, hemolysis, malignancies)
Drugs (antifolates and anticonvulsants)
Cause of folate deficiency
A reduction in oxygen carryng capacity resulting in decreased tissue oxygenation

Measured as a decrease in RBC, Hgb or Hct
Anemia
Fatigue
Dyspnea
Palpitations
Headache
Dizziness
Decreased Exercise or Work tolerance
Decreased Concentration
Symptoms of Anemia (impaired tissue oxygenation)
Bleeding site(s)
Stools + blood
Blood loss anemia
Jaundice
Splenomegaly
Maxillary hyperplasia
Hemolysis or dyserythropoiesis
Excessive milk intake
Iron deficiency
Pagophagia
Iron deficiency
Pica
Iron deficiency
Neonatal jaundice
Hemolysis
Splenectomy/Cholecystectomy
Hemolysis
Abdominal Surgery (ileal resection)
Vitamin B12 deficiency
Jaundice
Hemolysis
Splenomegaly
Hemolysis
Maxillary hyperplasia/frontal "bossing"
Hemolysis
Koilonychia (spooning of nails)
Iron deficiency
Hepato-splenomegaly
infltrative disorders
Lab tests used to diagnose and evaluate anemia
CBC, reticulocyte count, peripheral blood smear
CBC includes:
Hgb
RBC
MCV
RDW
HCT
MCH
MCHC
A reduction in the size of red cells (erythrocytes)

Measured as a decrease in the mean cell volume (MCV) of red cells.
Microcytosis
associated with defects in DNA synthesis
Macrocytes
associated with defects in Hgb synthesis
Microcytes
What Can Cause a Microcytic Anemia?
Decreased Iron Availability
Disordered Heme Synthesis
Disordered Globin Synthesis
Iron deficiency anemia in infancy in adults
Disorders of iron metabolism congenital / acquired

Sideroblastic anemias congenital / acquired
Thalassemias

Hemoglobinopathies (Hb E)
Causes of Microcytic Anemias
LOW Hb
LOW Hct
LOW MCV
LOWMCH &MCHC
HIGH RDW
CBC IN IRON DEFICIENCY
-IRON PROFILE:
Serum Iron
TIBC / Transferrin
% Iron Saturation of TIBC
Serum Ferritin

-Soluble Transferrin Receptor
-Free Erythrocyte Protoporphyrin
Lab tests in iron deficiency
TIBC
Total iron-binding capacity - an indirect method of determining the transferrin level in serum. Transferrin is saturated by the addition of iron to a serum specimen. Excess iron is removed, and the specimen is analyzed for iron content. The result is the total amount of iron that can be bound by transferrin. This result is helpful in differentiating anemias: high TIBC is associated with iron deficiency, low TIBC is associated with excess iron.
Causes: BLOOD LOSS BLOOD LOSS BLOOD LOSS BLOOD LOSS
usually from large bowel especially males and post-menopausal females
Iron deficiency in adults; other causes include:
Increased requirements (pregnancy and lactation)
Decreased absorption (decreased gastic acidity, small bowel disease)
Low birthweight
Perinatal bleeding
Low hemoglobin at birth
High growth rate
Early cow’s milk and solid food intake
Frequent tea intake
Infants at risk for iron deficiency
CONGENITAL:
X-linked or autosomal recessive MITOCHONDRIAL DISEASE
ACQUIRED:
DRUGS (isoniazid)
TOXINS (lead)
NEOPLASTIC (MDS)
Sideroblastic anemia
Disorders of globin chain imbalance
Any of the globin chains can be affected
Alpha
Beta
Gamma
Delta
The globin chains are structurally normal
Thalassemias
decreased synthesis of alpha globin chain “new” hemoglobins:
“Barts”-Gamma chain tetramer
“H”- Beta chain tetramer
Varied clinical features- determined by the number of alpha genes deleted
Alpha Thalassemias
Types of alpha thalassemia based on gene deletion
One gene deleted: Silent Carrier
Two genes deleted: “thalassemia minor”
Three genes deleted: hemoglobin “H” disease
Four genes deleted: Hydrops fetalis
MICROCYTIC HYPOCHROMIC RED CELLS
“TARGET” CELLS PRESENT
DECREASED MCV
NORMAL RDW
MILD ANEMIA
AlphaThalassemia Trait (thalassemia minor 2 alpha genes deleted)
MODERATELY SEVERE HEMOLYSIS
ELEVATED RETICULOCYTES

RED CELL INCLUSIONS ON SUPRAVITAL STAINING
HEMOGLOBIN H DISEASE (3 alpha genes deleted)
Incompatible with life thalassemia
HYDROPS FETALIS (4 alpha genes deleted)
DECREASED SYNTHESIS OF BETA CHAINS (usually due to mutations not deletions)
HETEROZYGOTE- (condition) minor (condition trait)
HOMOZYGOTE (or compound heterozygote) (condition) major (Cooley’s anemia)
Beta thalassemia
MILD MICROCYTIC HYPOCHROMIC ANEMIA.
ELEVATED Hb. A2
AUTOSOMAL DOMINANT
Low MCV
Normal RDW
B-THALASSEMIA MINOR
VERY LOW HEMOGLOBIN
TRANSFUSION DEPENDENT
AUTOSOMAL RECESSIVE
BONE MARROW TRANSPLANT CAN BE CURATIVE
B-THALASSEMIA MAJOR
Maxillary hyperplasia
Hepatomegaly
Splenomegaly
Marrow hyperplasia
"Hair on End" on X-ray
B-THALASSEMIA MAJOR
B-CHAIN MUTATION (B26lys..glu)
PREVALENT IN S.E. ASIA
MICROCYTIC HYPOCHROMIC
INTERACTS WITH B-THALASSEMIA
Hemoglobin E
COMPOUND HETEROZYGOTES FOR HEMOGLOBIN E AND BETA THALASSEMIA
MODERATELY SEVERE ANEMIA
SPLENOMEGALY
GROWTH DELAY
TRANSFUSION REQUIREMENT
Rule of threes
RBC x 3 =
Hb x 3 =
Hb
Hct
LOW MCV & NORMAL RDW
LOW MCV & HIGH/NORMAL RBC
Think thalassemia
LOW MCV& HIGH RDW
LOW MCV & LOW RBC
Think iron deficiency
Mentzer index - MCV/RBC
>13.5 =
<11.5 =
Iron deficiency
Thalassemia trait
Decreased Hb/Hct
Other cell lines decreased? Yes
Consider bone marrow exam
Decrased Hb/Hct
Other cell lines decreased? No
Evaluate RBC indices -
microcytic
normocytic
macrocytic
Iron Deficiency Anemia (IDA)
Anemia of Chronic Disease (ACD)
Iron excess/sideroblastic states
-Hemosiderosis
-Hemochromatosis
-Sideroblastic anemias
-Lead poisoning (Plumbism)
Porphyrias - heme synthesis disorders
Heme synthesis: iron metabolism disorders
Thalassemias
Hemoglobinopathies: HbC, HbE
Globin synthesis disorders
Iron Deficiency
Anemia of chronic disease
Thalassemias
Sideroblastic anemia
Microcytic anemias
Macrocytic Anemia
Defective DNA synthesis
Asynchrony between nuclear and cytoplasmic maturation
Gigantic cells with immature chromatin: megaloblasts
Macrocytic-normochromic red cells (macro-ovalocytes)
Granulocytes are hypersegmented
Megakaryocytes are abnormal resulting in thrombocytopenia
Vitamin B12 Deficiency
Folate Deficiency
Inherited Megaloblastic Anemias
Drug-Induced (dilantin, sulfa, AZT, methotrexate)
Other (Alcoholism, Hypothyroidism, Liver Disease, MDS, Reticulocytosis)
Macrocytic anemias
Acute hemorrhage
RBC enzyme defects, e.g. G6PD deficiency
RBC membrane defects, e.g. Hereditary spherocytosis
Bone marrow disorders (aplastic anemia, leukemia)
Hemoglobinopathies: HbS
Autoimmune hemolytic anemia
Anemia of chronic disease
Normocytic anemias
MCV formula =
Hct/RBC
Mean volume of all erythrocytes
MCV<80 fL
Microcytic
MCV > 100 fL
Macrocytic
MCH formula =
Hb/RBC
Amount of hemoglobin in an average RBC

Normal range: 26 - 33 pg
MCH > 34
this is considered to be too high because of macrocytic anemia (thus fitting more hemoglobin)
MCH < 26
this is considered too low. The MCH level can be too low because of blood loss over time, too little iron in the body, or microcytic anemia.
MCHC formula
Hb/Hct

Normal range: 32-36 g/dL
MCHC > 36 g/dL
Hyperchromic
MCHC < 32 g/dL
Hypochromic
RDW formula =
Standard deviation RBC volume x10/mean MCV

Measure of anisocytosis – variation/range in cell volume

Normal range: 12-16%
The MCV can be normal while individual RBCs vary in volume
Useful in early nutritional deficiency anemias, e.g. IDA
Increased/bimodal distributions: agglutination, fragmentation, transfusions, recently treated nutritional deficiency, reticulocytosis
Reticulocytosis
normally ↑ % in response to anemia
Reticulocytopenia
abnormal ↓ % in response to anemia
soluble protein-iron complex (apoferritin and Fe+3-phosphate core)
Synthesis stimulated by the presence of iron
Ferritin
insoluble protein-iron complex
Formed by lysosomal digestion of ferritin
Hemosiderin
-Present in food as ferric hydroxide and ferric-protein complexes
Meat and liver is good source of dietary iron
Average western diet contains 10-15 mg
Daily requirement 1-2 mg per day
-5-10% is absorbed in duodenum and jejunum
Facilitated by acid and reducing agents (citrates, ascorbate) in Fe+2 form
Inhibited by tannins, phytates
Increased absorption with demand (pregnancy, growth) and excessive loss due to acute or chronic hemorrhage
Iron in diet
Synthesized in liver, serum half-life of 8-10 days
Each molecule binds two iron atoms
Normally only about 30% saturated
Erythroblasts have transferrin receptors, CD71
Transferrin
-Primary cause of defective heme synthesis
Most common cause of anemia worldwide
About 20% of women, 50% of pregnant women; and 3% of men

-Etiology is age-related
Infants/children – dietary insufficiency
Adults – chronic blood loss, malabsorption, menstruation, blood donation, hemoglobinuria, etc.

-Clinical Features
1. Insidious, slowly progressive
2. Fatigue, irritability, dizziness, headache, breathlessness
3. Pica – craving/ingestion of unusual substance
4. Impaired neuromuscular activity
5. Brittle, pitted nails
6. Atrophy of lingual papillae, burning/sore mouth
7. Dysphagia, gastritis
Iron deficiency anemia
Hematologic Findings:
Hb usually < 8 g/dL
Reduced indices (MCV, MCH, MCHC)
RDW elevated
3. Reticulocytosis - mild
4. Thrombocytosis – may be twice normal (reactive)
5. BM – erythroid hyperplasia mild/moderate

Diagnostic Labs (Fe Studies):
1. Serum Ferritin decreased
2. TIBC increased
3. Saturation transferrin reduced
<16% supply to marrow below minimal requirement for heme production
4. Serum transferrin receptors increased
5. BM Fe stores depleted
iron deficiency anemia: diagnostic approach
Green leafy vegetables, beans, legumes, whole grains, oranges (heat labile)

Serum normal levels >3.7 ng/mL
Liver stores 20 – 70 mg
Sufficient for only 3 – 5 months


Polyglutamate deconjugated in GI/bile for absorption in jejunum
Circulates unbound (5-methyl THF)
Requires Vitamin B12 for entry

Essential for:

Purine/pyrimidine synthesis

Methionine synthesis

Methylation transfer reactions
Folate
Dietary insufficiency
Common in alcoholic, drug addicts
Low SE status
Chronic liver/kidney disease

Increased requirement
Pregnancy - supplemented prior to and during pregnancy
Cause neural tube defects in utero
Infancy
Hematologic diseases w/ rapid cellular proliferation, e.g. sickle cell anemia, leukemias

Defective absorption, e.g. Tropical/celiac sprue malabsorption

Drugs
Methotrexate (chemotherapy drug that is a folate antagonist)
Alcohol
Oral contraceptives
Others drug-induced folate deficiency (dilantin, sulfasalazine),
Mechanisms of Folate deficiency
synthesized by bacteria, found in meat, fish, dairy (heat stabile)

Normal levels 10-1000pg/mL;
Liver stores 3000 – 5000 mg
Sufficient for 2-5 years

Released by gastric acid
Binds to R-binder which is subsequently degraded by pancreatic enzymes
Binds to IF
Complex adheres to brush border of ileum (pH and Ca dependent)
TCI/III delivered to liver
TCII delivers to liver, BM

Essential for synthesis of:
Methionine
Succinate
Vitamin B12
Dietary lack (vegetarians)

Increased requirement

Defective absorption
Pernicious Anemia
Most common syndrome
Gastric parietal cell atrophy: ↓ IF
F>>M; disease of late adulthood
Severe atrophic gastritis
Neurologic problems

Gastrectomy

Blind loop syndrome: intestinal bacterial overgrowth

Fish tapeworm (competes for B12)

Other: Crohn’s disease, Zollinger-Ellison, Tropical/celiac sprue, Imerslund Syndrome (familial selective B12 malabsorption), hemodialysis, HIV

Rare causes: defective transport, disorders of metabolism
Vitamin B12 deficiency
Insidious onset

Moderate to severe fatigue, malaise

Lemon-yellow skin

Mucosal atrophy: tongue, vagina, GI with pain, malabsorption

Neurologic deficits/peripheral neuropathy (methionine loss)

Posterior and lateral columns of spinal cord
Paresthesias, numbness, tingling, ↓ vibration sense, ataxia, symmetric paralysis


CNS deficits
Megaloblastic madness (paranoia, depression)
Macrocytic anemia
-Moderate to severe anemia
MCV range from 100 – 150 fL
MVC > 120fL generally diagnostic, except for drug sulfasalazine
MCHC normal
Circulating macrocytes, minute RBC fragments, basophilic stippling, Howell-Jolly bodies
RDW usually markedly elevated
Reticulocytopenia
-Hypersegmentation of neutrophils
Early sign
≥ 6 nuclear lobes (or significant % with > 5)
Megaloblastic anemia diagnostic approach
Bone Marrow:
Hypercellular, with erythroid and myeloid hyperplasia
Increased mitoses, apoptosis
Large cells, immature nuclei with mature cytoplasm, multinuclearity
Giant myelocytes, bizarrely nucleated metamyelocytes
WARNING: May be mis-diagnosed as leukemia
Meagaloblastic anemia diagnostic approach
Defective DNA synthesis
Asynchrony between nuclear and cytoplasmic maturation
Gigantic cells with immature chromatin:
Macrocytic-normochromic red cells (macro-ovalocytes)
Granulocytes are hypersegmented
Megakaryocytes are abnormal resulting in thrombocytopenia
Morphologic Classification: Macrocytic Anemias
Vitamin B12 Deficiency
Folate Deficiency
Inherited Megaloblastic Anemias
Drug-Induced (dilantin, sulfa, AZT, methotrexate)
Other (Alcoholism, Hypothyroidism, Liver Disease, MDS, Reticulocytosis)
Macrocytic Anemias
Vinca alkaloids
Taxanes
interfere with what part of the cell cycle?
Mitosis
Anti-metabolites
Epipodophyllotoxins
Camptothecins
interfere with what part of the cell cycle?
DNA synthesis
Alkylating Agents
Anthracyclines
interfere with what part of the cell cycle?
Cycle nonspecific
More Common:
Cyclophosphamide* (CTX, Cytoxan®)
Ifosfamide
Platinums
-Cisplatin (CDDP)
-Carboplatin
-Oxaliplatin
Nitrosureas
-Lomustine* (CCNU)
-Carmustine (BCNU)
Less Common:
Mechlorethamine
Melphalan* (Alkeran®)
Chlorambucil* (Leukeran®)
Busulfan* (Myleran®)
Thiotepa
Procarbazine*
Dacarbazine (DTIC)
Temozolamide*
Alkylating agents - cycle non-specific
Alkylating agents also work on:
Mucosa, GI, hair
Template being replicated is misread or mispaired during DNA synthesis
Cross-linking prevents DNA strands from unwinding
Single or double-strand breaks in DNA occur
Potential outcomes of alkylating agents
Parent drug is converted by hepatic microsomal enzymes to 4-hydroxycyclophosphamide (4-HCP); liver impairment will affect dosing of the patient
4-HCP is converted to acrolein and phosphoramide mustard
Phosphoramide mustard alkylates DNA
Acrolein is responsible for causing hemorrhagic cystitis
Mechanism of Action Cyclophosphamide
Parent drug is converted to acrolein and ifosforamic mustard
There is more acrolein formed with (drug); more hepatic cystitis
Acrolein is responsible for causing hemorrhagic cystitis
Mechanism of Action Ifosfamide - Prodrug
Clinical uses of (drug):
leukemia, lymphomas, solid tumors
bone marrow transplant
treat graft-versus-host-disease
Rheumatic disorders & autoimmune nephritis
Cyclophosphamide
Clinical uses of (drug):
Solid tumors
Ifosfamide
Cyclophosphamide side effects
Myelosuppression (dose-limiting)
Hemorrhagic cystitis
Nausea & vomiting
Alopecia
Syndrome of inappropriate anti-diuretic hormone (SIADH) – get low sodium; increases risk for seizure and vomiting
Ifosfamide side effects
Hemorrhagic cystitis (dose-limiting)
Myelosuppression
CNS toxicity (can be increased when administered with other cyp450 drugs)
Nausea & vomiting
Alopecia
Pulmonary & cardiac toxicity
Caused by accumulation of acrolein
-Binds to thiol in bladder wall
Hematuria, urinary frequency & irritation
Prevent with vigorous hydration (≥2 L/day) & MESNA
Treat with bladder irrigation, alum irrigation, and other therapies
Heme test urine while on therapy
Hemorrhagic cystitis
Uroprotectant containing sulfhydryl group
-Binds to acrolein in the bladder to form a nontoxic compound
Not systemically absorbed so does not interfere with cytotoxic activity – goes straight to the bladder
Use with cyclophosphamide >2 g/m2/dose, ifosfamide ALWAYS
Effective in prevention only
MESNA
(mercaptoethane sodium)
-Cisplatin
Filtered by glomerulus & concentrated in renal tubules; incompletely cleared
Nephrotoxicity – ↓ GFR, electrolyte losses (Mg, K), and renal failure
Prevent with aggressive hydration (NaCl)
-Carboplatin
Not concentrated in the renal tubules; more efficiently cleared
Dosing based on area under the curve (AUC)
Dose = AUC ( GFR + 25 )
Option for those with problems with renal function
-Oxaliplatin
Platinum chemotherapeutic drugs
Clinical uses of (drug):
-Testicular, ovarian, metastatic bladder, lung and other solid tumors
Cisplatin – most active single agent in cervical cancer
Oxaliplatin – colorectal cancer
-Non-Hodgkin’s lymphoma
Clinical uses of platinum
Cisplatin toxicities
Vomiting – need antiemetic*
Nephrotoxicity*
Peripheral neuropathy
Neurotoxicity
Ototoxicity
Carboplatin toxicities
Myelosuppression*
Neurotoxicity
Vomiting
Oxaliplatin toxicities
Peripheral neuropathy (can’t drink cold drinks for almost 2 weeks)*
Myelosuppression
chemoprotectant agent that is metabolized to an active free thiol which binds to cisplatin and prevents damage to normal tissue
free radical scavenger
side effects include hypotension and nausea and vomiting
also used to prevent radiation-associated xerostomia
Amifostine (Ethyol) – administered with cisplatin to reduce some cytotoxic effects
-Chlorambucil (Leukeran)
Used for the treatment of chronic lymphocytic leukemia
-Busulfan (Myleran)
Used for treatment of leukemia and transplant
Also available IV form (used more for transplant)
toxicity is decreasing seizure threshold (require seizure prophylaxis)
-**Melphalan (Alkeran)
Used for the treatment of multiple myeloma
8 mg/m2 PO daily day 1-4 repeat every 28 days
All three of these medications come as 2 mg tablets (watch dispensing by brand name)
-Temozolomide (Temodar)
Used for treatment of brain tumors
150 mg/m2 PO daily days 1-5 a cycle repeat cycle every 28 days
Oral alkylating agents
Doxorubicin (Adriamycin®, hydroxy-daunorubicin)
Daunorubicin
Idarubicin
Epirubicin
Mitoxantrone
Anthracyclines
Mechanism of action (drug):
Inhibition of topoisomerase II

Intercalation between DNA base pairs, interfering with DNA synthesis

Formation of free radicals that damage DNA and cell membranes
Anthracyclines
Clinical use of (drug)
Breast cancer
-Most active agent
Sarcomas
GI tumors
Lymphoma
Doxorubicin and Epirubicin
Clinical use of (drug):
-acute leukemia treatment
Daunorubicin
Idarubicin
Mitoxantrone
Anthracyline Toxicities
Myelosuppression*
Cardiotoxicity*
Extravasation injury – leaks out of vein into surrounding tissue*
-Treat with Wydase and cold
Nausea and vomiting
Mucositis
red/orange urine discoloration
Acute:
Arrhythmias (free radicals causing damage to cardio tissue)
within 24 hours of administration
Related more to peak concentrations
Chronic:
cardiomyopathy
secondary to free radical formation
cumulative doses > 550 mg/m2
Anthracycline cardiotoxicity
Chronic cardiotoxicity with anthracycline is function of:
Total dose
Probability of CHF – probably should not administer to those with a poor ejection fraction (EJ)
-Dexrazoxane (Zinecard®)
Disrupts iron-anthracycline complex
Prevents free radical formation without interfering with cytotoxic activity
Used in leukemia with patients who have underlying (x) dysfunction
-Liposomal doxorubicin (Doxil®)
Liposomal delivery system not as readily taken up by tissue
Used in breast, ovarian cancer
How to mitigate the cardiotoxicty of anthracycline
Similar ring structure to anthracyclines
Similar mechanism of action, with decreased tendency for free radical formation
Decreased cardiotoxicity and extravasation
Decreased nausea and vomiting
Blue-green urine discoloration
Mitoxantrone
Anthracycline for:
Used in testicular cancer, Hodgkin’s Disease
Test dose needed only for Hodgkin’s disease
Watch for pulmonary toxicity and N/V
Bleomycin
Anthracycline for:
used in gastrointestinal tumors
used intravesicularly in bladder cancer
Sent to OR for shake and bake (placed directly in GI during operation – drug warmed and a med student rocks the patient back and forth to get the drug into small spaces)
Mitomycin C
Antifolates
Purine analogs
Plyrimidine antagonist
Antimetabolites
Taken up intracellularly by cancer & healthy cells
Inhibits dihydrofolate reductase decreases tetrahydrofolate decreases purine & thymidylate
Lack of purines & thymidylate prevents DNA synthesis
Leucovorin rescue
Reduced folate that bypass (drug) inhibition of tetrahydrofolate synthesis
Uptake healthy cells > cancer cells
Methotrexate
Clinical uses of (drug):
Hematological and solid malignancies
ALL, non-Hodgkin’s lymphoma
Breast and bladder CA
Osteosarcoma (high-dose)
Non-oncologic uses
Methotrexate
Toxicities of Methotrexate:
Myelosuppression and mucositis*
Nephrotoxicity (crystallization of MTX)*
-Avoid nephrotoxic meds (NSAIDs, sulfa)
Neurotoxicity (with intrathecal therapy)
Photosensitivity, eye discomfort
Pneumonitis
Hepatotoxicity
How to reduce toxicities of methotrexate
prevent renal damage by alkalinizing the urine with sodium bicarbonate solutions
avoid drugs that can interfere with excretion of methotrexate: Bactrim, NSAIDS, etc.
leucovorin rescue with high doses (yellow card)
can accumulate in fluid and leach out over time causing serious toxicity – ensure patient has no fluid collections (ascitis, pleural effusions, etc.)
Make sure CXR is obtained prior to dose
needed when administering high-doses of methotrexate > 100-500 mg/m2
directly converted into tetrahydrofolate without the need for dihydrofolate reductase
begin 24 hours after methotrexate given
should be given until methotrexate level is < 0.05 micromolar (5 x 10-8M)
Refer to yellow card
May give carboxypeptidase (from NCI)
Needs IRB approval prior to dose
Leucovorin rescue
inhibits multiple enzymes involved in folate metabolism and DNA synthesis
used for malignant pleural mesothelioma, non-small cell lung cancer
cutaneous reactions – prevent with dexamethasone 4 mg bid day -1, 0, +1 (folate is very important for skin growth)
give folic acid 350-1000 mcg daily and vitamin B12 1000 mcg IM q 9 weeks starting week before initiation and for 21 days after therapy to prevent hematologic and gastrointestinal toxicity
Pemetrexed (Alimta)
Standard of care in acute leukemias

Arabinose analog of cytosine

Phosphorylated to active component within cancer cells

Inhibits DNA polymerase
Cytarabine (Ara-C)
Clinical uses of drug:
Acute leukemias
Non-Hodgkin's lymphoma
No significant activity against solid tumors
Cytarabine (Ara-C)
Cytarabine (Ara-C) toxicities
Myelosuppression (100 mg/M2/day)
Alopecia
Gastrointestinal
Rash—plantar-palmer syndrome
High dose toxicities
(3 g/M2 q12h)
-nausea
-CNS toxicity
-chemical conjunctivitis, acral erythema
MOA & structure similar to cytarabine
Intermittent dosing more effective than continuous dosing
Effective for solid tumors
Pancreatic cancer
NSCLC
Achieves intracellular concentrations 20x greater than cytarabine
Gemcitabine (Gemzar)
Gemcitabine toxicities
Myelosuppression
Generalized rashes
Fever and flu-like symptoms
Peripheral edema
Nausea and vomiting-mild
NOT Neurotoxic
Clofarabine (Clolar)
Relapsed pediatric ALL
Peds 52 mg/m2 IV daily x 5 days ($36,000)
Adults 30-40 mg/m2 IV daily x 5 days
AE-skin toxicity-rash to desquamation

Nelarabine (Arranon)
Tcell ALL or Tcell lymphoblastic lymphoma
Peds 650 mg/m2 IV daily for 5 days
Adults 1500 mg/m2 IV Day 1,3,5
AE-neurotoxicity
Other pyrimidine antagonists
prodrug that is metabolized to FdUMP in order to be active
FdUMP binds to thymidylate synthase (TS)
prevents conversion of uracil (RNA) to thymidine (DNA)
stabilizes TS & FdUMP in the presence of leucovorin (given with leucovorin to treat colon cancer)
Mechanism of action of Fluorouracil
-fluorinated analog of uracil
Clinical uses of (drug):
Treatment of solid tumors including breast, colorectal and other GI tumors

Non-oncologic uses: actinic keratoses and noninvasive skin cancers
Fluorouracil
Leukovorin enhances the efficacy of this drug by locking it onto thymidylate synthase
Fluorouracil
Myelosuppression (bolus)*
Bloody diarrhea (CI)*
Mucositis (CI)*
Dermatologic (rings on your fingernails – like tree trunks)
Ocular
Nausea and vomiting (mild)
Cardiotoxicity (rare)
Toxicity of fluorouracil
Oral prodrug of fluorouracil
Metabolized to active component in tumor tissue
Use in metastatic colorectal & breast cancer
Take BID with food (↓ N/V)
Diarrhea, palmar-plantar rash
Capecitabine (Xeloda)
-Mercaptopurine (6-MP)
Metabolized by xanthine oxidase (same as allopurinol)
↓ dose x 75% if used with allopurinol
-Thioguanine (6-TG)
No dose reduction required with allopurinol
-Fludarabine & cladribine
Immunosuppressive → risk of opportunistic infections
Purine analogs
Inhibit de novo purine synthesis
-Vinca alkaloids
Vincristine (Oncovin®)
Vinblastine (Velban®)
Vinorelbine (Navelbine®)
-Taxanes
Paclitaxel (Taxol®)
Docetaxel (Taxotere®)
Mitotic inhibitors
“Spindle poisons” which bind to tubulin
MOA of mitotic inhibitors
Inhibit microtubule assembly
Interfere with formation of mitotic spindle
Cells accumulate in mitosis
Vinca alkaloids (plate spaghetti)
Promote microtubule assembly
Interfere with microtubule disassembly
Taxanes (hard spaghetti)
Neurotoxicity
Constipation
Vesicant
Extravasation
SIADH
Do not give Intrathecally
Should never be greater than 2 mg – can’t reverse
Vincristine toxicities
Myelosuppression
Vesicant
Extravasation
Vinblastine/vinorelbine
semi-synthetic vinca alkaloid
used for lung, breast, ovarian, lymphoma
toxicities:
myelosuppression
neuropathy
nausea and vomiting
extravasation
alopecia
Vinorelbine (Navelbine)
Myelosuppression
Mucositis
Peripheral neuropathy (cumulative) – tends to occur 18 months after therapy
Alopecia
Hypersensitivity reactions* (due to the mixing of this compound with oil)
Nausea and vomiting (rare)
Toxicities of Taxanes
Myalgia
Bradycardia
Cremaphor EL (oily solution)
Paclitaxel toxicity
Fluid retention
Palmar-plantar rash
Polysorbate-80 (oily solution)
Docetaxel toxicity
Semi-synthetic analog of epothilone B
Binds directly to ß-tubulin on microtubules, leading to suppression of microtubule dynamics
Some binding sites overlap with paclitaxel
-accounts for its activity in taxane resistant patients
Ixabepilone (ix-a-BEP-i-lone)Ixempra® (ix-EM-pra)
Treatment of patients with metastatic or locally advanced breast cancer:
In combination with capecitabine in patients resistant to treatment with an anthracycline and a taxane
Monotherapy in patients whose tumors are resistant or refractory to anthracyclines, taxanes, and capecitabine
Ixabepilone: Indication
Neurotoxicity about 65% overall in studies
Neutropenia incidence 65% but febrile neutropenia incidence only 3-4%
Premeds:
H1 blocker – diphenhydramine 50 mg
H2 blocker – ranitidine 50 mg IV
Ixabepilone: Adverse effects
Etoposide (VP-16) & teniposide (VM-26)
Inhibit topoisomerase II
Toxicities:
-Myelosuppression*
-Mucositis (BMT)
-Hypotension (alcohol-based diluent)
Clinical uses
-VP16 – AML, NHL, BMT, solid tumors (IV or oral)
-VM26 – ALL, SCLC
Epipodophyllotoxins
Irinotecan & topotecan
Inhibit topoisomerase I
Clinical uses:
Ovarian cancer
Lung cancer
CML, MDS
Cervical, ovarian cancer
Colorectal cancer
Camptothecins
Topotecan toxicity
myelosuppression
Irinotecan toxicities
Severe diarrhea (20%)
Acute (≤ 24 hours)
Facial flushing, abdominal cramping
Treat with scopolamine or atropine
Prevent with 5HT-antagonist & antihistamines
Chronic (~11 days)
Secretory → life threatening dehydration
Treat with high-dose loperamide
Degrades asparagine found in the serum
In lymphoid malignancies the lymphocytes are unable to produce asparagine due to a lack of or low levels of asparagine synthetase and rely on serum asparagine for its needs.
Without the serum asparagine the cells are unable to grow and reproduce
Used for ALL
Adverse Events
pancreatitis (check amylase)
*decreased fibrinogen < 100mg% (clotting problems) if low give cryoprecipitate
*hypersensitivity reactions
L-asparaginase
Used for CML
Causes myelosuppression
Doses 50 mg/kg/d (aprx 500 mg PO BID & titrate to WBC effect)
Hydroxurea
Selective, reversible inhibitor of the proteasome -
Proteasome: multi-enzyme complex in all cells; degrades proteins and regulates cell-cycle progression
Adverse events: peripheral neuropathy, fatigue, malaise, weakness, GI effects, thrombocytopenia
Used for Multiple Myeloma, NHL, ? leukemias
Bortezomib (Velcade)
Used with APL
Matures promyelocytes blasts inducing a CR
May cause (drug) syndrome that needs to be treated with dexamethasone
Dose: 45 mg/m2/d (round to nearest 10 mg) PO divided BID with food up to 90 days
Give with cytarabine and daunorubicin
(Drug) syndrome (maturation syndrome – cytokine release)-
-fever, dyspnea, pleural effusion, peripheral edema, hypotension
-treat-dexamethasone 10 mg IV BID x 3 days
All-trans retinoic acid (Vesinoid)
arsenic trioxide (Trisenox) – used for APL
retinoic acid syndrome (differentiation syndrome)
QTC prolongation
thalidomide – for multiple myeloma
increase risk for thromboembolism
drowsiness
peripheral neuropathies
pts / prescribers / dispensers must enroll in STEPS
Very teratogenic
Lenalidomide (Revlimid)-for MDS (myelodysplastic syndrome) and MM
Myelosuppression
Dose for MDS: 10 mg po day (patients are already myelosuppressed)
Dose for MM: 25 mg po day
Cells need methyl groups to grow
Removes methyl groups leading to cancer cell death
Well tolerated drugs
Hypomethylating agents
Used for MDS
Given 75-150 mg/m2 SQ or IV daily x 7 days
SQ route has local reaction
Azacitadine (Vidaza)
Hypmethylating agent
Used for MDS
15 mg/m2 IV every 8 hours x 9 doses every 6 weeks
20 mg/m2 IV daily x 5 days every 4 weeks
Decitabine (Dacogen)
Hypomethylating agent
Cancer cells can have too much (x) which allows the cell to grow unregulated (unable to die)
If inhibit (x), then allows the cell to develop normally and complete cell life
Ultimate goal is cell death through normal cell regulation
Histone Deacetylators (HDACs)
Histone: “spools” around which DNA wind
Histones contain lysine-rich amino-terminal tails that are responsible for conformational change by DNA
Remove acetyl group to lysine tail, restores charge, increases attraction between histones and DNA → condensation of chromatin → represses transcription
Histone Deacetylators (HDACs)
Some tumor cells produce excess amounts of histone deacetylase (HDAC), leading to a closed chromatin structure and prevention of DNA transcription
HDAC inhibitors have also been shown to:
Cause cell cycle arrest
Induce apoptosis
Inhibit angiogenesis
Clinical Use
Cutaneous manifestations in patients with cutaneous T-cell lymphoma (CTCL) who have progressive, persistent or recurrent disease on or following two systemic therapies
No dosing recommendations for hepatic or renal impairment
Patients should drink at least 2 liters/day of fluid to prevent dehydration
Vorinostat (Zolinza)
-Hematologic abnormalities
Anemia
Thrombocytopenia
-Gastrointestinal symptoms
Diarrhea, nausea
Taste disorders
-May prolong QTc interval
-Serious but rare
Pulmonary embolism
Squamous cell carcinoma
Anemia
-Laboratory abnormalities
Increased serum creatinine
Hyperglycemia
Proteinuria
Vorinostat (Zolinza) toxicities
MOA: Inhibition of mTOR blocks translation of mRNA and halts progression from G1 to S phase
Treatment of advanced renal cell carcinoma (came about approx. 3 years ago)
Dose
25 mg IV over 30-60 minutes once a week
Premedicate with antihistamine (i.e. diphenhydramine)
Hold for ANC < 1,000/mm3, platelet < 75,000/mm3, grade 3 AE’s
Restart when toxicities resolve to grade 2
Reduce dose by 5 mg/week (minimum dose 15 mg)
mTOR inhibitor:
Temsirolimus (Torisel)
Hypersensitivity reactions (9%)
Hyperglycemia / Hyperlipidemia
-plays role in glucose and lipid metabolism
Immunosuppression
-Infections and impaired wound healing
Bowel Perforation
-Fatal in 1 patient
Renal Failure
Interstitial lung disease (2%)
Temsirolimus: adverse effects
Destroys tumor cells through a number of possible mechanisms, including activation of complement and antibody-dependent cell-mediated cytotoxicity
Useful as means of targeting cytotoxic radioisotopes, toxins, or drugs to tumors, enhancing their delivery to tumors while minimizing systemic exposure
Animal (murine/equine), human or chimeric (from two species) derived
Monoclonal Antibodies (-mab) MOA
Monoclonal Ab naming:
momab:
zumab:
ximab:
radiolabeled
human
chimeric with murine and human
Infusion-related toxicity (65-80%): SOB, temp, chills, nausea, asthenia, and HA
-premedications—acetaminophen, diphenhydramine, hydrocortisone
Hypotension (10%)-recommend holding anti-hypertensives
MAB toxicities
-Rituximab (Rituxan) – called Vitamin R by patients; #5 drug used with all cancer patients at WFUBMC
Anti-CD-20 antigen found on B lymphocytes
Used for B-cell non-Hodgkin’s lymphoma
-Gemtuzumab ozogamicin (Mylotarg)
Anti-CD-33 antigen linked to ozogamicin
Used for Acute melogenous leukemia (AML)
Profound bone marrow suppression
-Alemtuzumab (Campath) – “liquid AIDS”
Anti-CD-52 antigen found on B and T lymphocytes
Used for B-cell chronic lymphocytic leukemia
Profound immunosuppression
1st generation MAB
-ibritumomab (Zevalin)
directed against CD-20
given with rituximab
used in follicular non-Hodgkin’s lymphoma
-tositumomab (Bexxar) - $20,000
directed against CD-20
used in follicular non-Hodgkin's lymphoma
Radiolabelled MAB
Regulates cellular proliferation, differentiation, function, & survival

Receptor & non-receptor enzymess
FLT3, VEGF, ABL, c-KIT, etc.

Activity tightly controlled in normal cells
Tyrosine Kinase (TK)
-Small molecule inhibition
Blocks ATP binding to (enzyme) domain
Stops intracellular signaling pathways
Cellular apoptosis
-Monoclonal antibodies
Target receptor (enzymes) or the ligand
Interrupt (enzyme) signaling
Antibody-mediated cytotoxicity
TK inhibitors
Imatinib (Gleevec®)
Gefitinib (Iressa®)
Erlotinib (Tarceva®)
Sunitinib (Sutent®)
Sorafenib (Nexavar®)
Small molecule TK inhibitors
Cetuximab (Erbitux®)
Trastuzumab (Herceptin®)
Bevacizumab (Avastin®)
Monoclonal antibodies TK inhibitors
binds to the extracellular domain of the human epidermal growth factor receptor 2 protein (HER-2) found on some breast cancers
used for metastatic breast cancer whose tumors overexpress the HER-2/neu protein
can cause congestive heart failure (therefore used as maintenance therapy after chemo)
trastuzumab (Herceptin)
antibody against vascular endothelial growth factor (VEGF)
used for metastatic colorectal cancer
inhibits blood vessel formation (do not give within a month of surgery)
causes hypertension
bevacizumab (Avastin)
antibody against epidermal growth factor receptor (EGFR)
used for metastatic colorectal cancer
causes acneform rash
cetuximab (Erbitux)
Patients with EGFR-expressing, metastatic colorectal carcinoma with disease progression on or following one or more regimens containing:
Fluoropyrimidine, Oxaliplatin, or Irinotecan
6 mg/kg IV every other week
Premedications are necessary
Toxicities:
Pulmonary fibrosis
dermatologic toxicity
infusion reactions
Hypomagnesemia
N/V/constipation
Panitumumab (Vectibix)
inhibits epidermal growth factor receptor (EGFR) tyrosine kinase

used as salvage treatment of non-small cell lung cancer
causes acneiform rash (can be a marker of actually having clinical effect), diarrhea, interstitial lung disease
erlotinib (Tarceva)
inhibits epidermal growth factor receptor (EGFR) tyrosine kinase
used for non-small cell lung cancer in patients who are benefiting or have benefited from gefitinib
skin rash, ocular symptoms, pulmonary symptoms
Off-market b/c so few patients respond to this
gefitinib (Iressa)
Skin rash (72%)
Diarrhea (35%)
Nausea/vomiting
Myelosuppression
Pulmonary symptoms (SOB, cough, fever) with acute onset or worsening
TK inhibitor toxicities
inhibits Bcr-Abl (enzyme)
-Bcr-Abl is the abnormal gene product that is caused by the Philadelphia chromosome in chronic myeloid leukemia (CML)
-also inhibits (enzyme) for platelet derived growth factor (PDGF), stem cell factor (SCF) and c-kit
Tyrosine Kinase Inhibitors: CML
Musculoskeletal pain
Fluid retention
QT prolongation
Myelosuppression
GI Bleeding
Dyspnea
Cardiac failure
Diarrhea
Headache
Dizziness
Constipation
Pyrexia
Fatigue
Skin Rash
Nausea/Vomiting
Cough
Anorexia
Pain
Neuropathy
CML TK inhibitor
used to treat Philadelphia chromosome + CML and Kit-positive gastrointestinal stromal tumors (GIST)
Dose: 400 to 800 mg daily
There are a lot of mutations that may be overcome except T315I – have to have transplant if you have this mutation
Imatinib (Gleevec)
Adults with chronic, accelerated, or myeloid or lymphoid blast phase chronic myeloid leukemia with resistance or intolerance to prior therapy including imatinib
Don’t take antacid 2 hrs prior to or after dose
Major drug interactions:
With CYP3A4 inhibitor, decrease dose to 20-40 mg daily
Consider increase in dose if given with CYP3A4 inducer
Dasatinib (Sprycel) or
Nilotinib (Tasigna)
Tyrosine Kinase Inhibitors: renal cell cancer
Sorafenib (Nexavar)
Advanced renal cell carcinoma in adults
Sunitinib (Sutent)
Gastrointestinal stromal tumors (GIST) after disease progression or intolerance to imatinib
Advanced renal cell carcinoma in adults
DECREASED OXYGEN CARRYING CAPACITY (RBC,Hb,Hct)

FUNCTIONAL DEFINITION DECREASED CAPACITY FOR OXYGEN DELIVERY
Anemia
What does statistical anemia imply?
People might normally lie outside 2 SD - they might not really have problems
MACROCYTIC
(x) HEMOGLOBIN
ALTERED ENZYME ACTIVITIES
SOME WILL HAVE TARGET APPEARANCE
ALTERED MEMBRANE ANTIGEN EXPRESSION
Fetal erythrocytes
NEONATAL PERIOD Birth – 1 month
Erythropoiesis
MARKED ERYTHROPOIETIC ACTIVITY AT BIRTH
INCREASED RETICULOCYTES
PREDOMINANCE OF FETAL ERYTHROCYTES
INCREASED ERYTHROCYTE MCV (mean cell volume)
EARLY INFANCY 2 – 6 months Erythropoiesis
“PHYSIOLOGIC” (adaptational) ANEMIA
HEMOGLOBIN NADIR @ 8-10 WEEKS OF AGE
HEMATOPOIETIC RECOVERY WITH INCREASED RETICULOCYTES AT END OF “PHYSIOLOGIC” ANEMIA.
PHYSIOLOGIC ANEMIA DEVELOPS EARLIER AND HAS A LOWER NADIR.
INCREASED RISK FOR IRON DEFICIENCY ANEMIA – LESS IRON STORES
Erythropoietic effects of prematurity
LATE INFANCY 6 – 24 months erythropoiesis
RAPID SOMATIC GROWTH, INCREASED IRON REQUIREMENTS.
NUTRITIONAL IRON DEFICIENCY ANEMIA PREVALENT
“ADULT-TYPE” ERYTROCYTES PREDOMINATE
CHILDHOOD erythropoiesis
“ADULT-TYPE” ERYTHROCYTES PREDOMINANT.
WBC DIFFERENTIAL SHOWS INCREASING NEUTROPHILS AND DECREASING LYMPHOCYTES.
Adolescence erythropoiesis
RAPID SOMATIC GROWTH.
INCREASED NUTRITIONAL IRON REQUIREMENT.
GENDER DIFFERENCES APPEAR IN HEMOGLOBIN LEVELS (androgenic effect).
HEMORRHAGE
Feto-Maternal Bleeding (look at mother’s blood for the presence of fetal cells)
Feto-Fetal Bleeding
HEMOLYSIS:
Inherited Red Cell Disorders
Alloimmune
Common Neonatal Anemias
IgG (IgM too big) CAN CROSS PLACENTA AND HEMOLYZE FETAL ERYTHROCYTES IF MOTHER IS IMMUNIZED TO A RED CELL ANTIGEN INHERITED FROM THE FATHER

Positive coombs' test (DAT)

Spherocytes
ALLOIMMUNE HEMOLYTIC ANEMIA of the NEWBORN
Hemolysis
Red Cell Disorders
MEMBRANE: HEREDITARY SPHEROCYTOSIS HEREDITARY ELLIPTOCYTOSIS
ENZYME

HEMOGLOBIN
MOST COMMON HEMOLYTIC ANEMIA DUE TO A MEMBRANE DEFECT.
THE DEFECTS ARE MUTATIONS IN PROTEINS THAT LINK VERTICALLY WITH THE LIPID BILAYER.
Hereditary Spherocytosis

SPLENOMEGALY
NEONATAL JAUNDICE
HEMOLYTIC ANEMIA
CHOLELITHIASIS
AUTOSOMAL DOMINANT (75%)
RESPONSE TO SPLENECTOMY
INCREASED MCHC – can be diagnostic
INCREASED OSMOTIC FRAGILITY
MEMBRANE INSTABILITY
ANKYRIN SPECTRIN BAND 3 BAND 4.2
Hereditary Spherocytosis
COMPLICATIONS OF HERDITARY SPHEROCYTOSIS
“APLASTIC CRISES” (parvovirus B19) – fifth disease
FOLIC ACID DEFICIENCY
INCREASED HEMOLYSIS
CHOLELITHIASIS
LEG ULCERS – because of decreased oxygen delivery
AUTOSOMAL DOMINANT
PROTEIN 4.1 SPECTRIN DEFECTS
MANY VARIANTS
CAN BE ASYMPTOMATIC
Elliptocytosis
Membranopathies
Spherocytosis
Elliptocytosis
Pyropoikilocytosis
Stomatocytosis
Most common glycolytic pathway enzymopathy
Autosomal Recessive
Neonatal Jaundice
Chronic Severe-Moderate Hemolysis
Transfusion dependant
Variable response to splenectomy
Very high reticulocyte count
Pyruvate Kinase Deficiency
Most common enzyme deficiency
X-Linked inheritance
Neonatal Jaundice
Infection induced hemolysis
Drug induced hemolysis
Fava bean hemolysis
Heinz body hemolytic anemia
"Blister" cells
G6PD Deficiency
MULTIPLE ENZYME VARIANTS
Congenital Nonspherocytic Hemolytic Anemia Chronic Hemolysis

B- variant (Mediterranean) unstable enzyme Fava bean hemolysis
A- (African) paroxysmal oxidant hemolysis. Hb down and reticulocyte up with drugs like primaquine due to lower oxygen levels.
G6PD Deficiency
CONGENITAL PURE RED CELL APLASIA (DIAMOND-BLACKFAN ANEMIA)

HEMOLYSIS Inherited Red Cell Disorders
Early Infancy anemias
CONGENITAL PURE RED CELL APLASIA
TYPICAL “FACIES”
THUMB ANOMALIES
FETAL RED CELLS (high MCV – low Hb, low reticulocyte, low RBC, low hct)
RPS 19 mutation (25%)
ELEVATED RBC ADENOSINE DEAMINASE
DIAMOND-BLACKFAN ANEMIA
HEREDITARY OROTIC ACIDURIA

PEARSON SYNDROME

TRANSCOBALAMIN II DEFICIENCY (leads to intracellular vitamin B12 deficiency)
PEDIATRIC ANEMIAS (UNCOMMON)
NUTRITIONAL IRON DEFICIENCY ANEMIA.
TRANSIENT ERYTHROBLASTOPENIA OF CHILDHOOD (T.E.C.)
Late infancy anemias
NORMOCYTIC NORMOCHROMIC (normal MCV)
LOW RETICULOCYTES
VERY LOW HEMOGLOBIN
SPONTANEOUS RESOLUTION
TRANSIENT ERYTHROBLASTOPENIA OF CHILDHOOD
SECONDARY TO OTHER DISEASE (anemia)
Childhood anemias
PANCYTOPENIA WITH CONGENITAL ANOMALIES
BONE MARROW HYPOPLASIA
RADIAL/THUMB ANOMALIES
ALTERED SKIN PIGMENT (HYPER OR HYPO PIGMENTED)
INCREASED RISK FOR MALIGNANCIES
Fanconi's Anemia

Fanconi’s Anemia is the most common type of inherited aplastic anemia. It is inherited as an autosomal recessive and has a carrier frequency of about 1:300.
Associated physical abnormalities include:
Skin hyperpigmentation and/or heterochromia (62%)
Short stature (59%)
Skeletal anomalies, esp. of the thumb (48%)
Hypogonadism in males (42%)
Renal anomalies- “horseshoe kidney” (24%)
Microcephaly or micrognathia (26%)
Mental retardation (13%)
Ear anomalies +/- deafness (10%)
Fanconi's Anemia
IRON DEFICIENCY ANEMIA

SECONDARY TO OTHER DISEASE

SPORTS ANEMIA
Adolescence anemias
IgG (warm) OR IgM (cold) MEDIATED
Paroxysmal Cold Hemoglobinuria (PCH)….IgG that behaves like IgM
Autoimmune hemolytic anemia
FIBRIN DEPOSITION IN CAPILLARY BEDS CAUSES FRAGMENTATION OF RED CELLS AND PLATELET TRAPPING
HEMOLYTIC UREMIC SYNDROME (HUS)
THROMBOTIC THROMBOCYTOPENIC PURPURA (TTP)
DISSEMINATED INTRAVASCULAR COAGULATION (DIC)
MICROANGIOPATHIC HEMOLYTIC ANEMIA
Hb. S is most common in U.S., followed by C and E.
Hb. S and E are most common world wide.
HEMOGLOBINOPATHIES

Electrophoresis or chromatography identifies variants with charge change.
Hb. S/S
Hb. S/C
Hb. S/B-thal. (+/0)
HEMOLYSIS
VASO-OCCLUSION
SICKLE CELL DISEASE

Splenic sequestration
Avascular necrosis
Stroke
Dactylitis
Acute chest syndrome
EARLY DIAGNOSIS (NEONATAL SCREENING)
HYPOSPLENIC FUNCTION
PNEUMOCOCCAL VACCINE
PROPHYLACTIC PENICILLIN
DACTYLITIS (HAND FOOT SYNDROME)
SPLENIC SEQUESTRATION
STROKE PREVENTION (HTN meds?)
SICKLE CELL DISEASE IN CHILDREN
UNSTABLE HEMOGLOBINS
MUTATIONS AFFECT HEME BINDING OR CONTACT POINTS.
NOT RECOGNIZED BY ELECTROPHORESIS.
OFTEN NEW MUTATIONS.
HEINZ-BODIES (DENATURED HEMOGLOBIN) FORMED.
EXCESSIVE MILK INTAKE

PICA -A perverted appetite for substances not fit as food or of no nutritional value; e.g., clay, dried paint, starch, ice.
Iron deficiency
NEONATAL JAUNDICE
FAMILY HISTORY OF SPLENECTOMY OR CHOLECYSTECTOMY
Hemolysis (congenital)
SHORT STATURE
THUMB & RADIAL ANOMALIES
HORSESHOE KIDNEY
MICRO-OPHTHALMIA
Fanconi's anemia
THUMBS
FACIES
DIAMOND-BLACKFAN ANEMIA
LACTIC ACIDOSIS
FAILURE TO THRIVE
Pearson syndrome
LOW MCV & NORMAL RDW
LOW MCV & HIGH/NORMAL RBC
Think thalassemia
LOW MCV& HIGH RDW
LOW MCV & LOW RBC
Think iron deficiency
High MCHC
Spherocytes
High MCV
? Marrow disease
?Reticulocytosis
High RDW
Check blood smear
Disc shaped, 2-4um, anuclear
Blue gray on Wright's stain with reddish-purple granules
Normal counts 150-450K
Circulate for 7-9 days
2/3rds in blood, 1/3 in spleen
Platelets
What drives platelet production?
Thrompoietin

The level of free eTPO in the blood drives platelet production
Platelet levels, in turn, regulate eTPO levels via binding and clearance by TPO-Rs on the platelets
Normal platelet levels allow more eTPO to be bound, leaving less eTPO available to bind to hematopoietic cells
With less eTPO binding to hematopoietic cells, fewer platelets are created by megakaryocytes
Normal platelet counts are maintained
In general thrombocytopenia, platelet levels are low
Low platelet levels result in less eTPO bound by TPO-Rs
Serum levels of unbound eTPO are increased
More eTPO is available to bind to progenitor cells and megakaryocytes; platelet production increases
More platelets are released into circulation, allowing platelet levels to return to normal levels
The width of the blue arrow represents the serum level of TPO, with wider arrows representing higher concentrations
a glycoprotein that binds to its receptor on platelets and megakaryocytes
produced at a constant rate by the liver
inverse relationship between serum levels and platelet mass
concentration regulated by the total mass of PLTs/megakaryocytes available to bind and degrade the protein
Thrombopoietin (TPO)
What is involved in all phases of differnentiation and maturation of platelets
Thrombopoietin
Beta-thromboglobulin
Factor V
Factor XI
Protein S
Fibrinogen
vWF
Platelet factor 4
Platelet-derived growth factor
Alpha granules of Platelets
ADP (activate neighbors)
ATP
Calcium
Serotonin (vasoconstrictor)
Dense bodies in platelet granules
Main function – formation of mechanical plugs during the normal haemostatic response to vascular injury
-If absent, spontaneous leakage of blood through small vessels may occur
Local release of vasoconstrictors (serotonin) to decrease blood flow to the injured area
Catalysis of reactions of the soluble coagulation cascade leading to fibrin clot formation
Initiation of the tissue repair process
Regulation of local inflammation
Platelet function
small blood cells produced by megakaryocytes. In the event of blood vessel injury, these tiny blood cells are rapidly recruited to the area of damage, where they effectively seal off the injured site to prevent blood loss. This is achieved through the execution of a series of functional events beginning with adhesion, followed by spreading and aggregation, leading to thrombus (clot) formation.
Platelets
Platelets roll and cling to non-platelet surfaces

Reversible,
seals endothelial gaps, requires vWF in arterioles
Platelet adhesion
platelets cling to each other

Irreversible, platelet plugs form, secretion of all platelet contents, requires fibrinogen
Platelet aggregation
discharge the contents of their granules

Irreversible,
occurs during aggregation, essential to coagulation
Platelet secretion
Following blood vessel injury, platelets adhere to the exposed subendothelial connective tissues.
Under the influence of shear stress, platelets move along the surface of vessels until the platelet engages collagen.
Following adhesion, platelets extrude long pseudopods which enhance interaction between adjacent platelets.
Platelet activation is then achieved by glycoprotein IIb/IIIa binding fibrinogen to produce platelet aggregation.
Events of primary hemostasis
Defects in GPIb result in:
Bernard-Soulier disease
Defects in GPIIbIIIa result in:
Glanzmann Thromboasthenia
Thombin
TXA2
ADP
Collagen
PAF
Inducer of aggregation
Consists of a series of reactions (coagulation cascade) that are triggered at the same time as platelet plug formation, resulting in the generation of cross-linked fibrin that encases and interlaces the platelet plug. Once endothelial injury has healed, the physiologic thrombus is no longer required. The endogenous fibrinolytic system results in the production of plasmin that degrades the cross-linked fibrin into cross-linked fibrin degradation products. The degraded thrombus is washed
Secondary hemostasis
Defined as platelet count <150,000/ul
Consequences
Bleeding following surgery or trauma with PLT counts < 50K
Spontaneous hemorrhage with PLT counts < 10K
Transfusion threshold
Thrombocytopenia
Cutaneous (petechiae, purpura, ecchymosis, venipuncture sites)
Mucosal (epistaxis, menorrhagia, hemorrhagic bullae in mouth - blood blisters, gastrointestinal bleeding)
Central nervous system (intracranial bleeding is most feared)
An “ooze” rather than a “gush”
Sites of bleeding with platelet disorders
Often see petechiae, ecchymoses, subconjunctival hemmorhage in what platelet disorder
Immune-mediated thrombocytopenic purpura
Cutaneous hemorrhage and pupura in what platelet disorder
drug-induced thrombocytopenia
Failure of platelet production
Increased consumption of platelets
Abnormal distribution of platelets
Dilutional Loss
Causes of Thrombocytopenia
Selective megakaryocyte depression
Rare congenital defects
Drugs, chemicals, viral infections

Part of general bone marrow failure
Cytotoxic drugs
Radiation
Marrow infiltration
HIV infection
Failure of Platelet Production
Absent radii syndrome
Associated with thrombocytopenia
Lack of dense granules in platelets
Gray platelet syndrome
Increased consumption of platelets
Immune
Autoimmune/idiopathic (ITP)
Infections: HIV, malaria
Drug-induced
Heparin (HIT)
Post-transfusional purpura
Disseminated intravascular coagulation (DIC)
Thrombotic thrombocytopenic purpura (TTP)
Immune Thrombocytopenic Purpura
Acute
Abrupt onset of bruising, petechiae, mucosal bleeding in a previously healthy person
May follow an infection, usually a nonspecific URI or GI virus
Majority recover without treatment
Chronic
Less responsive to therapy
Found in: Increased platelet turnover Myeloproliferative disorders Myelodysplastic disorders
Giant platelet
Morphology:Platelet larger than a normal red cell.
A Disease of Accelerated Platelet Destruction and Suboptimal Platelet Production
ITP pathophysiology
Treatment of ITP
Target the Immune System
Steroids
Splenectomy
Immune Globulin (gives the macrophage something else to chew on)

Increase Platelet Production
Thrombopoietin Receptor Agonists stimulate megakaryocytes
IgG Abs directed against heparin-platelet factor 4 complex
Suspect if platelet count falls to <100,000/ul or <50% of baseline value 5 to 15 days after heparin therapy is started
Venous, arterial, and microvascular thrombosis threatens life and limb
Heparin Induced Thrombocytopenia
Devastating disorder; fatal if untreated
Clinical pentad (FatRN):
Fever
Anemia
Thrombocytopenia
Renal dysfunction
Neurologic deficits
Blood film: schistocytes + few platelets
TTP-Thrombotic Thrombocytopenic Purpura
The absence or impairment of ADAMTS13 (vWF cleaving enzyme) allows for the persistence of the ultralarge “sticky” forms of vWF, which trap platelets and cause thrombi in vessels, thus leading to end-organ damage, and the appearance of the pentad of clinical features.
Pathophysiology of TTP - thrombotic throbocytopenic purpura
Thrombocytopenia Due to Dilution
Thrombocytopenia occurs in patients receiving massive transfusions (10-20 units) of PRBCs over a brief time frame due to the absence of viable platelets in stored PRBCs
Fibrin strands/clots
Give falsely low platelet counts
Normally, what percentage of platelets are circulating and what percentage are in the spleen?
70% circulating/30% spleen
Defined as a PLT count > 500K
Mechanisms
reactive : Cytokine driven (>80% of cases)
autonomous/clonal/neoplastic (Essential Thromboctyosis or thrombocythemia – these are “stupid” platelets)
Complications
Thrombosis in 15-20%, Bleeding in 3-5%
Neither typical in patients with reactive thrombocytosis
Thrombocytosis-Too Many Platelets
Obligate intracellular organisms
Utilize host cell machinery to replicate
Tropism for specific cell type(s)
Therapeutic window typically small
Latency/chronic infection frequently part of the natural history of infection
General Principles of Viral Infection
Occurred only once in a lifetime
Could be life threatening, profound anemia
Transfusions life saving and patients spontaneously recovered
Often preceded by a nonspecific ‘viral’ illness
Appeared to be outbreaks and even transmission to ‘roommates’ in the hospital
Aplastic Crises in Patients with Chronic Hemolytic Anemia Historical Context
-DNA virus, discovered in 1974, but only associated with disease in the early 1980’s
-Now clearly shown to be the causative agent of:
hydrops fetalis
erythema infectiosum (fifth disease)
aplastic crises in hemolytic subjects
seronegative RA syndrome
chronic anemia in immunocompromised hosts
Parvovirus B-19
about half of all adults are antibody positive
-prevalence rises rapidly during school years
20-60% of children in an outbreak situation will be symptomatic
virus shed in respiratory secretions
-patients with aplastic crisis and virtually no transmission once symptomatic (Erythema infectiosum)
Parvovirus B-19 Epidemiology
-Infects erythroid progenitor cells
thus marked anemia out of proportion to reductions in other cell lines w/infection
-Acute infection causes mild illness in most immunocompetent children (Erythema infectiosum)
onset of rash about the time of seroconversion
fever, myalgias, HA precede rash by a few days
No/little anemia due to RBC life span of 120 days
Parvovirus B-19 Pathogenesis
Arthropathy - mimics acute onset of RA, but seronegative; occurs with acute infection in ~60% of adults, but only 10% of children, women > > men (many think RA is infectious b/c this looks so similar)
Aplastic crisis: due to high reticulocyte counts in chronic hemolytic anemias
Chronic anemia due to inadequate immune response (Dx requires PCR, no Ab detectable); primarily seen in advanced HIV
Parvovirus B-19
Additional Clinical Syndromes
18 yo college freshman comes in on Monday after attending his first big rush party that weekend. Doesn’t remember much, but he’s been feeling poorly for several weeks which he related to being away from home for the first time, 8 o’clock classes, rush,etc. Major complaints now are sore throat, chills, fatigue. Noticed some swelling in his neck and difficulty swallowing

Atypical monocytes on smear

1st sexual encounter just a few weeks before
Epstein Barr virus/mono
-Cytomegalovirus
particularly in allogeneic BMT patients
-Varicella Zoster or Herpes simplex
usually as part of dissemination
-Community acquired
Respiratory Syncytial Virus (RSV)
Influenza
Parainfluenza
Viral Causes of Pneumonia in Immunocompromised Hosts
Idiopathic pneumonia
ARDS
Alveolar Hemorrhage
Leukemic Infiltration
Lymphoma
Pulmonary Emboli
Aspiration
Drug Induced Lung Injury:
bleomycin
cyclophosphamide
busulfan
ifosfamide
methotrexate
BCNU
doxorubicin
Non-infectious Causes of Fever and Pulmonary Infiltrates in Immunocompromised Hosts
Seropositive status (donor or recipient)
older age
conditioning regimens with agents other than cyclophosphamide
GvHD!!!!!!!
Idiopathic is unlikely infectious b/c of lack of variation in severity with level of immuno compromise
Risk Factors of CMV in BMT Recipients
Diagnosis is usually clinical, but BAL cytology and culture helpful
Very often found in presence of another pathogen
-(condition) immunosuppressive in and of itself
-fungi (including PCP) and Pseudomonas most common
CMV pneumonia
IV GCV (ganciclovir) 5 mg/kg q 12h (dose adjusted for renal failure) is cornerstone
-duration unclear; personal style - - -> bid x 14 d, then qd x7d, then off
-Can substitute valganciclovir 900 mg po bid as equivalent if tolerating/absorbing po meds
Foscarnet for salvage/intolerance
unclear if (virus) Ig has any role in SOT, perhaps in unresponsive disease, but
Either (virus) Ig or IVIg is absolutely necessary for treatment in HSCT recipients
Therapy of CMV disease
-Prophylaxis
anti-(viral) therapy when either donor or recipient is (virus) seropositive
easier to write into a protocol
no need for surveillance
may expose many to risks of drug for benefit of a few
-Preemptive Therapy
more targeted therapy for brief periods given with:
‘induction’ therapy
ALA (anti-lymphocyte antibody) therapy
defined lab evidence of infection
fewer patients exposed
may improve reconstitution of immunity
requires sensitive and predictive lab tests
Two Major Strategies to Reduce the Risk of CMV Disease
-Anemia
DRUGS
neoplasia (Kaposi Sarcoma, lymphoma)
Folate/B12 deficiency
Infection:
MAC
MTb
CMV
Chronic Parvovirus B19
Fungi (histo, cocci)
-Thrombocytopenia
ITP:
most common
responds to therapy for HIV
can Rx with steroids, IVIG, etc, but short-term help
TTP:
also may be initial manifestation of HIV
DIC
Hematologic Manifestations of HIV
68 yo man (diagnosed late 50s) with CLL treated with Chlorambucil/prednisone followed by fludarabine intermittently, but reasonably well controlled
admitted to the CCU with gradual onset of left sided chest pain
became very severe, no associated symptoms
exam demonstrated marked left sided chest tenderness
Ruled out for MI by enzymes/EKGs
on rounds, medical student notices a rash under his left arm
vesicular, erythematous base
by the time the attending gets there two hours later it has spread to the left chest, but does not cross the midline
Herpes Zoster
Frequent in patients with lymphoma, lymphoid leukemias and after bone marrow or PBSC transplantation (up to 50% in some series)
increased risk of dissemination, but still rare
Probability of (virus) increased with graft vs. host disease
Vaccine NOT indicated (yet) in immunocompromised
treatment options not compared in RCTs:
IV acyclovir 10mg/kg q 8h
PO Acyclovir 800 mg 5x/day
PO Valacyclovir 1000 mg tid
PO famciclovir 500 mg tid
Herpes Zoster
Do peripheral blood counts decrease with age?
Not significantly

Marrow can sustain normal peripheral blood counts throughout the human lifespan, but…

Diminished reserve capacity in times of stress
Common causes of anemia in the elderly
Iron deficiency 20%
Anemia of chronic disease (ACD) or anemia of inflammation 20-25%
Chronic kidney disease
B12 or folate deficiency
Relationship between hemoglobin and mortality
<12 g/dL increased risk of mortality (HR >1)
12-16 g/dl have (HR <1)
Increased mortality
Increased cardiac disease (CHF, MI)Decreasedmuscle mass/ strength
Increaseddisability
Increased falls and fractures
Associated with cognitive impairment
Clinical associations of anemia in elderly
May be the first sign of underlying serious illness (ie. Colon cancer)

May be an independent cause of morbidity and mortality
Iron Deficiency Anemia
Longer duration of carcinogen exposure
Decreased DNA repair ability
Increased genomic instability
Decreased tumor suppressor activity
Decreased immune surveillance
Theories of Carcinogenesis in Aging
Do elderly patients benefit from aggressive chemotherapy?
Selected elderly patients can benefit from aggressive treatments

Study compares aggressive tx versus supportive care for older adults with AML
Population statistics consistently demonstrate decreased survival in adults >65
Patients >65 are 16 times more likely to die of disease1
Older adults experience increased toxicity related to treatment
As a group, older adults experience inferior outcomes
Outcome disparity for older cancer patients is multi-factorial
Treatment disparity:
-research bias and under-treatment
Tumor characteristics:
-tumor biology
Host characteristics:
-physiologic changes
-impairment in physical function
-comorbidities
Research bias
Only 1/3 of patients on NCI sponsored trials were >65 years of age1

Very few adults >75 years of age are enrolled on clinical trials2

Poor generalizability due to selection bias
Up front “dose attenuation” results in inferior outcomes when treating for cure in:
Aggressive NHL1
Small cell lung cancer2
Breast cancer3
Dose reduction may not be doing older adults any favors
Why are older adults treated differently?
Concern for increased toxicity
Question effectiveness of treatment
Lack of referral
Social marginalization
Patient preference?
Lack of clinical trial data
Age related physiology
Older adults tend to have decreased reserve capacity in times of stress
Decreased intestinal absorption
Decline in renal excretion

Changes in volume of distribution

Altered metabolism by cytochrome P450
? Bioavailability

Increased toxicity?

Increased toxicity due to increased free drug

Impaired activation or elimination?
What outcomes are most meaningful to your patients?

Survival?
Avoidance of Disability?
Maintenance of functional independence?
“Quality of life”?
Treatment Decisions in Elderly Cancer Patients
Myelosuppression: Use prophylactic colony stimulating factors in patients >65 years old receiving myelosuppressive combination therapy
Renal : Consider adjustment of renally excreted drugs based on GFR
Mucositis: Nutritional support, early hospitalization if dysphagia/diarrhea develops
Neurotoxicity: monitor neurotoxic regimens closely (ex. hearing loss, neuropathy, cerebellar toxicity)- consider alternatives if possible

Cardiac: careful pretreatment assessment, avoid cardiotoxic regimens if possible
Emerging Guidelines to Minimize Toxicity in Elderly Patients
Aging is not a disease but does decrease physiologic reserve
Blood counts do not decrease with normal aging
Anemia is common and should be evaluated
Malignancy is more common in the elderly
Considerations of treatment of malignancies in older adults should be individualized on the basis of multiple factors including functional status, comorbidities, and goals of treatment
Take home points about aging and cancer treatment
% of all people with cancer will receive radiation at some point during their cancer treatment.
50-60%
The “mother of Radiation Oncology”
Marie Curie. The Curie’s were the first to use ionizing radiation as a potential therapy. The Curie’s received the Nobel Prize early in the 20th century for their work.
Attacks reproducing cells, it does not distinguish between cancer cells and normal tissues.

The damage to normal cells can result in side effects.

Therapy involves a balance between destroying the cancer cells (in order to cure or control the disease) and sparing the normal cells (to minimize undesirable side effects).
Radiation oncology principles
is thought to work by damaging the DNA in cells.
Radiation
TYPES OF RADIATION USED TO TREAT CANCER
Electromagnetic radiation, (x-rays and gamma rays). Particulate radiation (electrons, protons, neutrons, alpha particles, and beta particles) are all forms of ionizing radiation
uses high-energy photons from radioactive sources such as cobalt, cesium or a machine called a linear accelerator.
The most common type of radiation therapy
GOALS OF RADIATION THERAPY
Radiation is considered a local treatment because only cells in the area being treated are affected.
Radiation may be used in early stage cancers in an attempt to cure or control the disease.

Radiation may be used before surgery to shrink the tumor or following surgery to prevent the cancer from coming back.
It may be used in combination with surgery and/or chemotherapy.
is the first part of treatment planning.
Simulation (sometimes referred to as a marking session)
At simulation, the size or volume of the tumor is determined. Potential route of spread and normal tissues in the treatment area are assessed. Also consider dosing lymph nodes due to likelihood.
The radiation dose will be decided based on a number of factors and by the ability of the normal tissue to tolerate the radiation; dose is normally based on dose to normal tissue – side effects.
Treatment planning for radiation oncology
One Gray is equal to
100 rads and one cGray (cGy) = one rad.
the radiation is focused from a source outside the body onto the area affected by the cancer. It is much like getting a x-ray, but for a longer time. This type of radiation may be given by machines called linear accelerators.
External beam radiation
is also known as brachytherapy. Brachytherapy means short-distance therapy. The two main types of brachytherapy are interstitial radiation and intracavitary radiation.
Internal radiation therapy
several techniques used to deliver a large precise radiation dose to a small tumor volume. The term surgery may be confusing since no incision is actually made. The most common site being treated with this technique is the brain. (Gamma Knife)
Sterotactic surgery or sterotactic radiation therapy
A genetic alteration occurs in an immature hematopoietic cell
Resulting in clonality, and excess growth
ie: neoplastic transformation
ie: cancer
Usually occurs with myeloid or lymphoid cells but can involve other cells, including cells in the erythroid or megakaryocytic lineage
Leukemia pathogenesis
“Acute” Leukemias
Describes which cell is in excess
Describes when people present
Describes rate of growth
Describes type of treatment
Acute Myelogenous Leukemia
Acute Nonlymphocytic Leukemia
Synonyms for Acute Myeloid Leukemia
AML: What do these blasts do?
Grow uncontrollably
Signal to grow is always on
Signal to stop growing or die gets turned off
Maturation (differentiation) is halted
Inhibit growth of normal cells in the marrow
Quickly becomes a life-threatening disease
Prior chemotherapy
Alkylating agents and epipodophyllotoxins
Prior ionizing radiation
Particularly prenatal (less relevant now)
Exposure to benzenes
Abnormal genetics:
Down syndrome
Neurofibromatosis
Schwachman syndrome
Bloom syndrome
Familial monosomy 7
Kostmann syndrome
Fanconi anemia
AML: Risk Factors
How do we diagnose AML?
Bone marrow biopsy
There must be >=?% of blasts to have diagnosis of AML
20%
What is definitive for being a myeloid cell?
Auer rods
What do we do with the aspirate?
Flow Cytometry
-This helps us determine lineage
-This is how we tell an AML from an ALL –always do this for confirmation
Cytogenetics
Fish
What do we do with the core?
Look at the cellularity
AML with t(8;21)(q22;q22), (AML1/ETO)
AML with abnormal bone marrow eosinophils and inv(16)(p13q22) or t(16;16)(p13;q22), (CBFb /MYH11)
Acute promyelocytic leukemia with t(15;17)(q22;q12), PML/RAR-alpha and variants
AML with 11q23 (MLL) abnormalities
AML with recurrent genetic abnormalities
Following MDS or MDS/MPD
Without antecedent MDS or MDS/MPD, but with dysplasia in at least 50 percent of cells in two or more myeloid lineages
AML with multilineage dysplasia
Alkylating agent/radiation-related type
Topoisomerase II inhibitor-related type
Other
AML and myelodysplastic syndromes, therapy related
AML, minimally differentiated
AML without maturation
AML with maturation
Acute myelomonocytic leukemia
Acute monoblastic/acute monocytic leukemia
Acute erythroid leukemia (erythroid/myeloid and pure erythroleukemia variants)
Acute megakaryoblastic leukemia
Acute basophilic leukemia
Acute panmyelosis with myelofibrosis
Myeloid sarcoma
AML, not otherwise categorized
AML: Treatment
Don’t use surgery
Don’t use radiation
Use chemotherapy – circulates in the blood like the leukemia cells do
Chemotherapy is nonspecific:
Kills the good cells as well as the bad cells
First treatment patients get
Called so because we are trying to induce a remission (this is our goal)
Standard of care:
Cytarabine (or ara-C) 100-200 mg/m2/d IV CI on days 1-7
Daunorubicin 45-90 mg/m2/d IV on days 1-3
(and sometimes we add…)
Etoposide 100 mg/m2/d IV on days 1-3
“7+3+3”
AML: Treatment: Induction chemotherapy
So if the chemo only takes 7 days to give why do we make patients stay in the hospital for 4 to 6 weeks?
Supportive care (during time of pancytopenia)
Need RBCs
Need platelets
Preventing and treating tumor lysis syndrome (uric acid, phosphorous, calcium goes to kidneys, potassium – these are chemicals released from the lysed cells)
Monitoring for and treating side effects of the medications:
Nausea, vomiting, diarrhea, mucositis
Cardiotoxicity (daunorubicin), hepatic toxicity, renal dysfunction/failure, etc.
AML - How do we know our chemo is working?
Repeat a marrow on day 14 (nadir marrow)
If no leukemia is visible, await count recovery – takes about another two weeks
If leukemia still present, need additional chemo – either “5+2+2” or different chemo regimen
This resets the clock and extends hospital stay
Recovery marrow taken before patient leaves after induction therapy
To make sure that the only cells that came back were the good ones.
If we can’t see any leukemia cells, the patient is in remission, and we have achieved our first goal
Sludging” symptoms:
Chest pain, shortness of breath, headaches, blurry vision
Usually occurs when patients have WBCs >75K with majority of those cells being blasts
Need emergent leukopheresis
Central line is placed and blasts are filtered out of blood by a pheresis machine (give hydroxyurea to impair cell cycle)
WBC can drop from 300K to 150K, for example, in a matter of hours
Leukostasis associated with AML induction chemotherapy
Involves three rounds “booster treatments”
Round: high dose ara-C: 3 grams/m2 IV q12 hrs on days 1, 3, and 5
Patients go home after chemo has finished running in (six day hospitalization) – but they still need supportive care
Discharge patients on antibiotic pills
Arrange for outpatient transfusions as needed
~50% of patients will have to return to the hospital because of fevers while neutropenic, to get IV antibiotics
AML Treatment: Consolidation
Increased age
(In)Ability to tolerate chemotherapy
Higher incidence of MDR (multi-drug resistant) abnormalities
Higher incidence of antecedent hematologic disorders (MDS, etc.)
Secondary AML (toxin-induced)
Certain genetic abnormalities
-5, -7, 11q23 (MLLgene)
<10% CR rate
Not obtaining a remission after induction chemotherapy
AML: Bad Prognostic Factors
APL: acute promyelocytic leukemia t(15:17)
AML FAB M3
>80% CR at 5 years
Inv(16) or t(16;16)
Some AML FAB M4
~60% CR at 5 years
t(8;21)
Some AML FAB M2
>40% CR at 5 years
AML: Good Prognostic Factors
Normal cytogenetics and AML?
Considered to be an intermediate risk factor.
1st CR if pt has poor cytogenetics
At the time of relapse – but need to get them into CR again
AML and transplant
Comprises ~10% of AMLs
In > 90% of cases involves t(15;17) – is diagnostic
Patients are younger at the time of diagnosis
Associated with risk of DIC
High incidence of early fatal hemorrhage (10-20%)
7% of patients will die of intracranial hemorrhage
AML M3: Acute Promyelocytic Leukemia
Treatment is different than AML
ie: we don’t use 7+3+3
Instead, we use 7+4+ATRA
“7”: 7 days of cytarabine, as before
“4”: 4 days of daunorubicin, instead of 3
ATRA: all-trans retinoic acid
APL: Treatment
t(15;17)
Chromosome 15 has PML gene (promyelocytic leukemia)
Chromosome 17 has RARalpha gene (retinoic acid receptor alpha) – normal promoters don’t work with the translocation
Retinoic acid promotes normal promyelocyte differentiation
When the translocation occurs, supra-physiological doses of retinoic acid are needed to promote differentiation of the promyelocytes
ATRA all-trans retinoic acid (Vitamin A)
Helps decrease bleeding complications associated with APL and DIC
Improves CR rate
Works by a different mechanism, we skip the day 14 marrow and just do a recovery marrow
APL: Treatment: ATRA
-Hyperleukocytosis
Why cytotoxic chemo follows (drug) by 2 days
-(Drug) syndrome
AKA: differentiation syndrome
Fever, pulmonary infiltrates, hypotension, dyspnea
Thought to be due to cytokines from granules
Treat with 2 to 3 day course of decadron
APL: Treatment: ATRA
CALGB 9710:
Arsenic trioxide improved overall survival and event-free survival

Overall treatment is for 2 years, much longer than other AML regimens
APL: Treatment
What is the least common type of leukemia in children?
CLL
What is relative incidence of leukemia as a cancer in children?
30%
What is the most common leukemia in children?
ALL
predominance of very immature WBC precursors; these cells proliferate, and lack differentiation.
Acute leukemia
Proliferation of relatively mature WBC’s; often indolent; more commonly seen in adults than children
Chronic leukemia
peak incidence at age 2-5 years
whites > blacks
males > females
ALL epidemiology
80% of all childhood leukemias are ALL
fatigue
pallor
bruising, bleeding
fever
lymphadenopathy
hepatosplenomegaly
mediastinal mass
pain (musculoskeletal)
Clinical manifestations of ALL
leukocytosis or leukopenia (high or low white blood cell count, respectively); may see “blasts” on the blood smear
anemia (low hemoglobin/hematocrit)
thrombocytopenia (low platelets)
may see chemical abnormalities consistent with “tumor lysis” (increased uric acid, phosphorus, potassium, creatinine)
Blood findings of ALL
Infection (especially EBV, other viruses)
Immune thrombocytopenic purpura (ITP)
juvenile rheumatoid arthritis
aplastic anemia
other malignancies (e.g. neuroblastoma)
ALL differential diagnosis
>25% lymphoblasts in bone marrow (usually upwards of 80%)
lumbar puncture also required for evaluation of CNS disease (loves to go to the brain)
Diagnosis of ALL
Diagnosis/classification of ALL
Morphology
Cytochemical stains
Immunophenotyping
Hyperdiploidy (>50 chromosomes per leukemia cell)
t(12;21) translocation (TEL-AML1 fusion gene, aka ETV6-RUNX1)
Trisomies of chromosomes 4, 10, and 17
Cytogenetic findings associated with favorable ALL prognosis
Hypodiploidy (<44 chromosomes per leukemia cell)
t(4;11) translocation (MLL-AF4 fusion)
t(9;22) translocation (BCR-ABL fusion or Philadelphia chromosome)
Cytogenetic findings associated with an unfavorable ALL prognosis
Classification of ALL
-Acute lymphoblastic leukemia (L1/L2 morphology)
Precursor B cell (B-lineage, pre-B, early pre-B)
Precursor T cell (T cell)
-Acute lymphoblastic leukemia, B-cell (L3 morphology) (mature B-cell, Burkitt cell leukemia)
-Acute leukemia, biphenotypic
associated with:
Males > Females
older age (5-12 years)
high WBC count
bulky adenopathy, mediastinal mass, hepatosplenomegaly
CNS disease
Precursor T cell (T cell) ALL
ALL emergencies
Sepsis (infection)
Bleeding (from thrombocytopenia)
Tumor lysis syndrome
Hyperleukocytosis (very high WBC count)
Tracheal compression/SVC syndrome
ALL - treatment
Combination chemotherapy
4 components of therapy:
1. Remission induction (~1 month)
2. Intensification (consolidation) (~6 months)
3. CNS treatment (throughout all phases)
4. Continuation (“maintenance”) (2-3 years)
Commonly used drugs for ALL
Steroids (prednisone, dexamethasone) – don’t give unless you know what you are treating (can mask “real” problems) – only for lymphoblastic
Vincristine
Asparaginase
Doxorubicin/daunorubicin (antharcycline)
Methotrexate
Mercaptopurine
Cytarabine
Cyclophosphamide
Imatinib for ALL
BCR-ABL (Philadelphia t (9;22))positive
Nelarabine for ALL
T-cell
Rituximab for ALL
CD20-positive
Clofarabine for ALL
All subtypes
ALL - CNS treatment
Intrathecal chemotherapy (delivered by lumbar puncture (LP)) – start on first day of therapy and continue throughout treatment duration (18-20 LPs over ~3 years)
radiation therapy (e.g. CNS positive at diagnosis or relapse, T-ALL)
Only 5-20% patients currently get CNS radiation
Can it be omitted for all patients?
high dose IV methotrexate - penetrates CNS
ALL - treatment timeframe
Treatment generally lasts 2.5 - 3 years (exception is B-cell (Burkitt’s) ALL, which is treated intensively for only about 5 months)
stem cell/bone marrow transplants generally reserved for refractory disease or very high risk patients
Pediatric ALL - Prognosis
Depends on multiple factors (age, WBC at diagnosis, etc. – see next slide) but prognosis is generally GOOD, with ~80% overall event-free survival (Unlike adult ALL, where prognosis is relatively poor - only 30-50% of adults are cured)
ALL - risk assessment
Best done by combining the following data for each patient:
presenting clinical features (age, WBC count)
blast cell immunophenotype (T-cell vs. B-cell) and genotype (cytogenetics, other DNA tests)
early responsiveness to treatment
Patients are currently classified as low, standard, high, or very high risk
Late effects of ALL therapy (and primary culprits)
Neurocognitive delay (CNS therapy)
Endocrinopathies (CNS therapy and steroids)
Gonadal failure/sterility (alkylating agents)
Cardiac dysfunction (antharcyclines)
Musculoskeletal disease (steroids)
Second malignancies (chemotherapy and radiation therapy)
Relapsed ALL
Longer first remissions are better than shorter first remissions
In general, prognosis for relapsed ALL is 30-50%
if long first remission - chemotherapy alone
if short first remission - stem cell transplant
CNS, testicles (“sanctuaries”) are a relatively common sites of extramedullary (outside bone marrow) relapse
– there are some mature cells
Cancer of the white blood cells
Malignant cell is relatively immature stem cell
Result is excess production of mature cells of multiple lineages and bands
RBCs are not elevated – could be anemic
CML
How do we diagnose CML?
Find the Philadelphia chromosome
t(9;22)
Chromosome 9: abl gene (Abelson leukemia virus)
Chromosome 22: bcr gene (breakpoint cluster region)
How does the t(9;22) work in CML?
The abl protein is a tyrosine kinase, which is an enzyme involved in signal transduction
When the t(9;22) is present, the tyrosine kinase is always phosphorylated (ie always “on”)
This provides a constant signal to certain pathways that results in cell growth that exceeds apoptosis
Chromosome 22 can break in different regions within the bcr gene, resulting in different sizes of bcr-abl protein products
p190 (190 kDa protein): seen in ALL
p210 (210 kDa protein): seen in CML
Chronic phase of CML
<5% blasts in the marrow
Accelerated phase of CML
Many different criteria
5-20% blasts in the marrow
Blast crisis in CML
>20% blasts in the marrow
Just like any other acute leukemia
Can be myeloid or lymphoid (flow cytometry)
Treat like a new AML crisis
How do you treat CML?
Gleevec (imatinib mesylate)
TKI (tyrosine kinase inhibitor) – remember that the ABL is always phosphorolated and TK always on
STI (signal transduction inhibitor)
Small molecule inhibitor
Pill taken once a day
Side effects:
Mild nausea and vomiting
Periorbital edema
Pleural effusions
Well-tolerated
Binds to c-kit
On GISTs
(GI stromal tumors)
Binds to PDGFR-alpha
Seen in hypereosinophilic syndrome
Gleevec
Gleevec resistance
Some via T315I mutations
Dasatinib
Nilotinib
Neither are effective against the T315I mutation – go back to caveman tx of CML
2nd generation tyrosine kinase inhibitors
Acclerated phase of CML can be treated with
Higher doses of Gleevec
Blast crisis in CML is treated like:
acute leukemia:
Myeloid blast crisis: “7+3” cytarabine x 7 days with daunorubicin x 3 days
Lymphoid blast crisis: Complex multi-agent chemotherapy regimen; exactly what we would use for de novo ALL
Cancer of the white blood cells
Malignant cell is more differentiated than CML
Results in excess numbers of mature-appearing lymphocytes
Continuum with SLL (small lymphocytic lymphoma – cancer of lymphocyte – type of NHL) that has no circulating neoplastic cells and resides in lymph nodes
Chronic lymphocytic leukemia
What will the CLL CBC look like?
Elevated total white cell count
Differential is primarily lymphocytic
Hemoglobin and platelets are normal
Except in advanced stages of disease when Hb and platelets can be low
How do we diagnose CLL?
Peripheral blood sample for flow cytometry
Do not need a bone marrow for diagnosis
(+) CD5 (normal t-cell), CD19, CD23 (normal b-cell)
(+/-) CD20 (weak expression)
(+) surface immunoglobulin
Light chain restriction
Only kappa or lambda, not both
Send chromosomal studies to get information on prognosis
When do you start treatment of CLL?
Need to have symptoms:
Symptomatic lymphadenopathy
Symptomatic splenomegaly
“Symtomatic” counts
Anemia or thrombocytopenia as a result of progression of CLL in the marrow
Stage III or stage IV disease
Note that the absolute white cell count is NOT listed as an indication to treat
Immune dysregulation (do not say functionally neutropenic)
Insufficient immune system:
Difficult to fight infection
Often need prolonged courses of antibiotics
Hypogammaglobulinemic
Quantitative immunoglobulins often reveal patients to be pan-hypoglobulinemic
If patients have persistent infections or infections severe enough to require hospitalization, will treat with IVIG
Seen in CLL
Overactive immune system
Inappropriate destruction of “self” cells
Immune thrombocytopenic purpura
Labs are the same as with any other:
Elevated LDH, bilirubin
Low Hb, haptoglobin
Can be Coombs positive (a test for antibodies, the so-called anti–human globulin test using either the direct or indirect)
Generally treat with steroids
Don’t need a bone marrow to make this diagnosis
AIHA in CLL
How can we tell if a CLL patient’s thrombocytopenia is from ITP or stage IV disease?
Do a bone marrow - if it is stage IV, there will be no room in the bone marrow for megakaryocytes to make the plateletes
Development of diffuse large B cell lymphoma arising from one CLL clone
May have B symptoms, one area of lymphadenopathy out of proportion to others
PET scan will show transformed sites
CLL is not PET avid (slow growing)
DLBCL is very PET avid (fast growing – diffuse large B-cell lymphoma)
Must document with biopsy to prove transformed disease
Treat with DLBCL regimen such as R-CHOP
CLL chemo is ineffective
Pts are still left with underlying CLL after treatment complete
Richter's transformation in CLL
CLL Treatment
-Chemotherapy - purine analog based
Fludarabine based
Pentostatin based
-Patients are at significant risk for tumor lysis syndrome with the first cycle of treatment
-Hydration and frequent lab monitoring is important
Comprises 2% of all leukemias
Very slow-growing
B cell malignancy (CD19, 20, 22)
Has aberrant expression of T cell marker CD103
TRAP-positive (tartrate-resistant acid phosphatase), a stain – diagnostic for HCL, only present on HCL
Clinically, notable for very large spleens, and dry taps on bone marrows (lot of fibrosis/scaring of bone marrow)
Hairy Cell Leukemia
In the bone marrow, cells look like fried eggs
Marrow also has lots of fibrosis
Treatment is also with purine analogs: Cladribine – might only need one treatment in a lifetime because this is so slow-growing
Hairy Cell Leukemia
-Rolling, tight adhesion, spreading, diapedesis, chemotaxis
-phagocytosis
-degranulation
-release of antimicrobial products (O2 dependent and O2 independent)
Neutrophil functions
Definition: absolute increase in number of leukocytes in peripheral blood
Without reference to cell type or level of maturity
In adults, greater than 10,000-11,000/mm3
Majority of cases of leukocytosis are due to an increase in neutrophils
Leukocytosis
Leukocytosis caused by:
Infection
Inflammation
Stress
Drugs
Trauma
Hemolytic anemia
Normally responding bone marrow
Leukocytosis caused by:
Acute leukemias
Chronic leukemias
Myeloproliferative disorders
Abnormal bone marrow
An excessive white blood cell response (i.e. 50,000 white blood cells per cm3) associated with a cause outside the bone marrow
May be neutrophilic, eosinophilic, lymphocytic, monocytic
Usually caused by relatively benign processes (i.e., infection or inflammation)
An underlying malignancy is the most serious but least common cause.
Down syndrome babies can have – resolve spontaneously
Leukemoid reaction
Decrease emigration of neutrophils from blood into the tissues
Increase release of mature neutrophils from the bone marrow (release of storage pool)
Decrease margination of neutrophils inside vasculature
Corticosteroids
Defined as absolute neutrophil count of 8,000/mm3 or higher
Mechanisms of:
-Increased production by bone marrow
Infection
Inflammatory stimulus
Hemolysis and chronic blood loss
Exogenously administered hematopoietic growth factors (G-CSF and GM-CSF)
-Increased mobilization from storage pool or marginated pools (pseudo(term)) - White cell count not over 15,000/mm3
Vigorous exercise
Epinephrine
Labor and pregnancy
-Failure to exit the circulation
Anatomic or functional asplenia
Adhesion deficiency syndromes (decreased endothelial adhesion and migration) – LAD (leukocyte adhesion deficiency)
Neutrophilia
Partial or total deficiency of CD11/CD18
Number of circulating neutrophils is increased
Associated with severe and fatal bacterial infections
Delayed detachment or prolonged healing of umbilical stump
Leukocyte adhesion deficiency
Absolute lymphocytosis:
>9,000/mm3 in infants and small children (2-3 yrs)
7,200/mm3 in older children
>4,000/mm3 in adults
seen in children due to rapid tissue growth and development of the immune system
Physiologic lymphocytosis
Causes of absolute lymphocytosis
Acute infections: CMV, EBV, pertussis, hepatitis, toxoplasmosis
Chronic infections: TB, brucellosis
Lymphoid malignancies: CLL
Causes of relative lymphocytosis
Normal in young children
During viral infections
Splenomegaly
Benign Reactive Lymphocytosis
Pronounced :
Pertussis
Acute Infectious Lymphocytosis (Coxsackie)
Infectious Mononucleosis (EB)
Allergic events
Parsitic infections
Dermatologic conditions
Scarlet fever, cholera, leprosy, GI infections
Immunologic disorders: RA, SLE, eosinophilia-myalgia syndrome
Malignancies: NHL, Hodgkin's disease
MPD
Adrenal insufficiency
Sarcoidosis
Pleural and pulmonary conditions
Etiology of eosinophilia
Infections: varicella, chronic sinusitis
Inflammatory conditions: IBS, chronic airway inflammation
MPD
Endocrinologic: hypothyroid, ovulation, estrogens
Alteration of marrow and RE compartments
Basophilia
Decrease in the absolute neutrophil count (ANC) below accepted norms for age
Ethnic and racial groups < 900
Benign ethnic neutropenia (some African and Asian)
Altitude (lower ANC above 5,000 ft)
Neutropenia
Age related “lower limits of normal” for neutrophils
Term newborn (up to 1 week) < 3,000
Infant (1 week – 2 years) < 1,100
Child, adolescent, adult < 1,500
immune neutropenias, hypersplenism, infection
Normal marrow reserve
congenital neutropenias, marrow failure syndromes, chemotherapy, etc
Decreased marrow reserve
Low normal neutrophil count
No history of infections
Most due to increased neutrophil margination along blood vessel wall
Entry into circulation and exit from vascular pool are normal
Rapid mobilization of neutrophils from marginal pool with exercise, epinephrine
Pseudoneutropenia
Common during viral infections, usually transient
Proctated can be seen with mononucleosis, hep B and HIV
Mechanisms:
Increased use
Up regulation of adhesion and migration due to complement and cytokines
Marrow suppression
Infection induced neutropenia
More than 100 drugs implicated
Immune mediated or direct destruction of granulocyte precursors
Drug induced neutropenia
Antibodies directed to neutrophils or their precursors
Can be associated with other immune cytopenias (hemolytic anemia and immune thrombocytopenia) or autoimmune disorders
Chronic benign neutropenia: occur in children and adults. Infections are infrequent despite low ANC (how do you diagnose? – challenge test?).
Majority resolve spontaneosly within few yrs from diagnosis
Immune neutropenia
-Cyclic neutropenia:
Autosomal disorder
Marked neutropenia every 21 days, nadir last 3-7days
Patients are subject to recurrent severe infections
-Kostmann syndrome
Present at birth
ANC < 200/ul
Predispose to leukemia and preleukemic conditions
Congenital neutropenia
Absolute lymphocyte count (ALC) of <1,000/mm3 in adults or <1,500/mm3 in children
ALC of less than 1000/mm3 in an infant is highly abnormal
Decreased Production
Immunodeficiency Diseases: SCID (severe combined immunodeficiency) or AIDS, aplastic anemia
Increased destruction: acute stress (steroid administration), irradiation, immunosuppressive agents, chemotherapeutic agents
Increased loss: congenital lymphatic abnormalities
Lymphopenia
Stimulation of chemoreceptor trigger zone (CTZ) (outside BBB – then transmits inside BBB to vomiting center)
Peripheral mechanisms
- Damage of gastrointestinal (GI) mucosa
- Stimulation of GI neurotransmitter receptors
Cortical mechanisms
- Direct cerebral activation
- Indirect (psychogenic) mechanisms
Vestibular mechanisms
Potential mechanisms of chemotherapy induced nausea/vomiting
Central Mechanism
Chemotherapeutic agent activates the CTZ located in the area postrema in the brainstem
Activated CTZ invokes release of various neurotransmitters, which activates brainstem vomiting center
Peripheral mechanism
Chemotherapeutic agent causes GI irritation and damage to GI mucosa, resulting in release of neurotransmitters
Activated receptors mediated by vagal afferents send signals to brainstem vomiting center
Neurotransmitters may act independently or in combination to induce vomiting
Proposed pathophysiology of chemotherapy induced n/v
Acute phase of CINV
Most common
Begins 1 to 2 hours, peaks 4 to 10 hours, resolves within 12 to 24 hours
Usually associated with high frequency and severity
Serotonin antagonists are most effective
Neurokinin antagonists are also effective
Delayed phase of CINV
Begins 1 to 5 days after chemotherapy, peaks 48 to 72 hours
Less severe than acute N/V, longer duration
Associated with high-dose cyclophosphamide, mitomycin-C, cisplatin, doxorubicin, and ifosfamide
Neurokinin-1 antagonists are most effective
Anticipatory phase of CINV
Not associated with a time frame
Conditioned response
Usually prior history of severe N/V
Stimulated from environment
Occurs in up to 25% of patients
Often refractory to therapy
Blocks serotonin both centrally and peripherally
Effective for acute N/V but not any more effective for delayed N/V than other therapies
Most effective when given 30 minutes prior to administration of chemotherapy
Increase efficacy when given with corticosteroids (90% effective with corticosteroid/60% w/o)
Serotonin (5HT3) Receptor Antagonists
5HT3 Receptor Antagonists Adverse Events
Headache (migraine patients don’t have enough serotonin)

Constipation

Diarrhea

EKG changes – concern only with underlying arrhythmias
Ondansetron (Zofran®) (5 cents)
8-32 mg po/iv daily (either daily or divided)
8 mg oral dissolving tablets (ODT) available
Granisetron (Kytril®)
1-2 mg po daily
10 mcg/kg IV daily
Granisetron (Sancuso) ($250/patch – lasts 7 days)
Dolasetron (Anzemet®)
50-100 mg po/iv daily
Palonosetron (Aloxi®)
0.25 mg IV x 1 – may last 3-5 days
Only indicated for highly emetogenic chemotherapy as oral agent – starting to be used more generally due to availability of generics
5HT3 Receptor Antagonists
-setron are serotonin receptor antagonists
Inhibits Substance P
Indicated for the prevention of acute and delayed CINV in combination with serotonin antagonists and corticosteroids
Part of combination regimen with a 5HT3-receptor antagonist plus dexamethasone:
Day 1 125 mg PO 1 hour prior to chemotherapy
Days 2-3 80 mg PO in morning
Neurokinin-1(NK-1) Receptor Antagonists Aprepitant (Emend)
Aprepitant (Emend) Adverse Events
Asthenia/fatigue (18%)
Nausea (13%)
Hiccups (11%)
Diarrhea (10%)
Somnolence
Steroids should be decreased by 50% when given IV or 25% when given PO
Should be used in caution when administering agents that are metabolized by CYP3A4
Inhibition of CYP3A4 by aprepitant could result in elevated plasma concentrations of the following agents:
Etoposide* - Irinotecan
Ifosfamide (increase in CNS toxicity) - Paclitaxel*
Vinorelbine* - Docetaxel*
Vinblastine - Vincristine
Imatinib - Warfarin
*The company has provided data to say that these agents can be administered safely together.
Drug interactions with Aprepitant (Emend)
Phenothiazines
prochlorperazine (Compazine®), promethazine (Phenergan®) – you sleep with these
Butyrophenones
droperidol (Inapsine®)
Substituted benzamide
metoclopramide (Reglan®)
Dopamine Receptor Antagonists (use with other antiemetics)
Effective for delayed nausea/vomiting

Adverse events
Akathisia (shifting in seat; restlessness) – lorazepam can help
Dystonia (tremor) – diphenhydramine or benztropine can help
sedation – more common with promethazine (will become tolerant to this)
Phenothiazines
Prochlorperazine (Compazine®)
5-10 mg PO/IV/PR q 4-6 hours
promethazine(Phenergan®)
12.5-50 mg PO/IV/PR q 4-6 hours

Prochlorperazine is a more potent antiemetic in cancer patients but has a higher incidence of akathisia and dystonia
Phenothiazines
Blocks dopamine in the CTZ and peripherally
increases esophageal sphincter tone
improves gastric emptying
increases transit through small bowel
Also blocking serotonin at high dosing – that’s why you low dose dopamine and high dose serotonin
Adverse events: EPS, restlessness, sedation, fatigue, nausea and diarrhea (dexamethasone may decrease diarrhea)
Substituted benzamides
Metoclopramide (Reglan)
10 mg PO q6h (useful for mild nausea)
0.5 mg/kg IV q6h (blocks serotonin)
Possesses amnesic (retrograde), anxiolytic and sedative properties

Used for anticipatory nausea

Not effective for preventing emesis and is usually given with other antiemetics

Lorazepam (Ativan) 1-2 mg IV q4h prn
Benzodiazepines
Increases appetite, improve mood and sense of well being
Immunosuppression
May increase the differentiation of WBC and therefore is not given in acute myelogenous leukemia (AML) - demargenation
Most information with dexamethasone 10-20 mg po/iv daily
Adverse events: mood changes, anxiety, euphoria, headache, metallic taste, abdominal discomfort
Corticosteroids
Less effective than metoclopramide but more effective than phenothiazines
Not effective with highly emetogenicity
Adverse events: mood changes, dysphoria, memory loss, hallucinations, blurred vision, hypotension, tachycardia
Beneficial with younger patients
Dose is 2.5-5 mg po TID
Can increase appetite
Cannabinoids Dronabinol (Marinol)
Beneficial for nausea associated with movement (works through vestibular system)

Scopolamine 1.5 mg patch changed every 3 days (transderm patch for seasickness)
Anticholinergics

Can't see or spit, urinary retention and constipation
Regulate hematopoiesis
Proliferation
Differentiation
Maturation
Effects of CSFs on mature cells
Increase chemotaxis
Enhance phagocytosis
Increase cytotoxic killing
Improve responsiveness to antigens
Enhance eosinophil function
Growth factors
Granulocyte colony-stimulating factor (G-CSF)
Filgastim (Neupogen)

Pegylated filgastim (Neulasta)
Granulocyte-macrophage colony stimulating factor (GM-CSF)
sargramostim (Leukine)
Erythropoietin (EPO)
Procrit or Epogen
Megakaryocyte CSF
Oprelvekin (IL-11) (Neumega)
Supports proliferation of neutrophils

Stimulates neutrophil function

No effect on mature eosinophils and macrophages

Cancer patients receiving:
Myelosuppressive chemotherapy
Bone marrow/stem cell transplantation

Peripheral blood stem cell collection

Severe chronic neutropenia
Activity of G-CSF Filgrastim (Neupogen)
Filgrastim (Neupogen®) Adverse Events
Bone Pain
Occurs in the lumbar, sternal and pelvic areas
Common during initiation of therapy and times of rapid growth
May reflect increased bone marrow activity
Pain responsive to acetaminophen or NSAIDs (be careful of these b/c they will suppress any fever should it occur)
Only attributable adverse event of G-CSF
Pegylated (peg means big) Filgrastim (Neulasta)
This larger chemical structure makes clearance slower and therefore allows administration once after chemotherapy
Cleared by neutrophils so as the white blood cells recover then they are able to clear the drug (patient dependent)
Stimulates CFU-GM and CFU-GEMM to increase neutrophils, macrophages, monocytes, eosinophils
No clinically significant activity on other cell lines
Autologous bone marrow transplant
Treat of BMT failure or engraftment delay (auto or allo BMT)
Neutrophil recovery following chemotherapy for AML
Mobilization of PBPC
Peripheral stem cell transplantation
May increase response to antifungal therapy because of macrophage involvement
Activity of GM-CSF Sargramostim (Leukine®)
Sargramostim (Leukine®) Adverse Events
Constitutional Symptoms
Fever, headache, myalgias, arthralgias
Fever responds well to antipyretics
May have less fever with SQ administration
Bone pain
Skin reactions
Pleural and pericardial effusions
First-dose effect
Enhances RBC production (inc HCT)
Decreases need for RBC transfusions (so these have gone up so that patients can feel better and not receive blackbox drug)
Patients should have Hgb < 10 g/dL or HCT < 30%
Monitor iron stores (need the iron for binding!)
Requires 2-6 weeks to see response
This unfortunately increased the risk of death – only use in non-curable patients

End stage renal disease
AIDS patients treated with zidovudine
Anemia associated with cancer chemo
Anemia of prematurity
Myelodysplastic syndrome
Bone marrow/stem cell transplant
Preoperative blood collection
Activity of EPO Erythropoietin (Procrit/Epogen) and ESAs (Erythropoietin stimulating agents)
Adverse Events of EPO
Hypertension (24%)
Headache (16%)
 tendency to clot vascular access (more RBCs to clot)
Iron deficiency anemia
Seizures
Achiness and cold sensation in long bones
Pyrexia (fever) (38% of AZT-treated patients)
Larger molecule with more carbohydrate groups to allow for slower elimination
Dose: indicated 2.5 mcg/kg SQ weekly or 500 mcg SQ every 3 weeks (this is given less frequently)
Should be used in patients receiving chemotherapy with no intent for cure
New FDA guidelines state that all patients should receive medication guide prior to therapy
Darbopoietin (AraNesp)
Activity
Promotes megakaryocyte production
Prevents severe thrombocytopenia – this has only prevented a minimal number of platelet transfusions
Dosage
50 mcg/kg SQ daily
Continue until plt > 50,000
Clinical use has not been shown to prevent transfusions of platelets because transfusion of platelets does not occur until count < 10,000
Activity and Dosage of IL-11 Oprelvekin (Neumega)
Adverse Events of IL-11 (no reason to give this dinosaur)
Tachycardia (19-30%)
Atrial arrhythmias (60-75%)
Peripheral edema (60-75%)
Headache (41%)
Dizziness (38%)
Insomnia (33%)
Fatigue (30%)
Fever (35%)
Rash (25%)
Dyspnea (48%)
Diseases treatable by HSCT (human stem cell transplant)
Leukemias and lymphomas
AML, ALL, CML, CLL
Hodgkin's lymphoma
Non-Hodgkin's lymphoma
Multiple myeloma and other plasma cell disorders

Myelodysplastic/
myeloproliferative disorders
Myelodysplastic syndrome
Chronic myelomonocytic leukemia
Agnogenic myeloid metaplasia/myelofibrosis

Other malignancies
Testicular lymphoma
Renal cell
Bone marrow failure syndromes:
Severe aplastic anemia
Fanconi anemia
Paroxysmal nocturnal hemoglobinuria (PNH)
Pure red cell aplasia
Amegakaryocytic thrombocytopenia

Immunodeficiency states:
Severe combined immunodeficiency (SCID)
Wiskott-Aldrich syndrome

Hemoglobinopathies:
Beta thalassemia major
Sickle cell disease

Inborn metabolic disorders:
Hurler's syndrome (MPS-IH)
Adrenoleukodystrophy
Metachromatic leukodystrophy
collected from and infused into the same person
Autologous
collected from an identical twin
Syngenic
collected from another member of the same species
Related - member of same family, usually a sibling
Unrelated - general population (NMDP)
Allogeneic
The most important cells are the cells earliest in development, that can then differentiate into WBC, RBC, platelets. The hematopoetic stem cells!
It is now more common to utilize peripherally collected HSC’s.
After GCSF priming (+/- preceding chemotherapy) the peripheral blood WBC is monitored. After it increases an outpatient procedure called apheresis is performed
Peripheral HSCT collection
nothing more than glorified high dose chemotherapy, or chemotherapy with a cellular rescue.
Autologous HSCT
Tumors for which we would consider (treatment) as a therapy have steep dose response curves to chemotherapy.
Increasing the doses of chemotherapy continues to increase the numbers of cancer cells killed.
The dose limiting toxicity to even more effective doses of chemotherapy is myelosuppression
Is a mechanism to get around the dose limiting toxicity
Autologous HSCT
To be successful, you need to have a chemosensitive disease
You need to be able to harvest a healthy graft
Ex- not contaminated by active tumor
Ex- can not use in bone marrow failure states such as aplastic anemia, myelofibrosis, etc
Autologous HSCT
Common indications for autoHSCT
Multiple myeloma
Large cell lymphoma in first relapse
Hodgkins disease in relapse
AML with high risk cytogenetics in first CR if not a candidate for allogeneic HSCT due to lack of donor or underlying patient status
High risk peripheral T cell lymphomas in first CR
Guarantee of a clean graft
Can be used in marrow failure states
Largest difference is this is an immunologic intervention, hoping to harness a graft versus tumor (leukemia in the past) effect
Much of our immunity is based on our blood cells (we think of this in infection). Cellular immunity also is involved in cancer surveillance
Donor lymphocytes in absence of chemotherapy can induce regression of leukemias
Higher rate of relapse in syngeneic than allogeneic HSCT in AML 52 vs 16% at three yrs
Benefits: Allogeneic vs AutoHSCT
Need to find an HLA matched donor, most often a sibling.
Graft versus host disease- essentially grafted cells recognizing patient as “non-self” and attacking. Primarily a T cell mediated process
Acute vs chronic. Need to give additional immunosuppressive drugs to prevent this, which further increases risk of infection
Largely due to GVHD and increased infections, TRM is much higher- 10-20% by day 100.
Negatives Allo vs AutoHSCT
Who is a compatible donor? Immunology and HLA typing
Histocompatibility typing - “tissue typing”
HLA: human leukocyte antigens= MHC (Major histocompatibility complex) in humans
Chromosome 6 has the genes that determine the compatibility antigens ( Class I and Class II)
Gene Loci are designated as:
A, B, C (Class I)
D (Class II)
C appears to have little role in “compatibility”
6 main antigens expressed ( 2 copies of each A, B, D) one from each chromosome 6
Immediate access to stored stem cells after search
Are relatively immune incompetent in mounting normal allogeneic response
Has been shown to have less GVHD
Donors are less allo-immunized
Can tolerate wider HLA disparity
Less likely to be CMV+, hence less CMV infections
Eurocord reported 0.3% positive for CMV by PCR, compared to > 40% positive in adult BM
20% are collected from ethnic minorities
ethnic minorities are underrepresented in most marrow donor registries
Umbilical Cord Blood
Risk factors for acute GVHD – can happen any time post transplant

Occurs in 10-50% of all allogeneic stem cell transplant pts
Increasing age of host
Prior exposure of donor to other blood groups (including pregnancy)
Donor and recipient gender disparity – relatively minor
CMV status of donor and host
Intensity of the transplant conditioning regimen
Peripheral blood stem cell versus bone marrow transplantation
Acute GVHD prophylactic regimen used
Most common site is skin, often manifesting as an erythematous maculopapular rash at the time of engraftment
Can also be involved in chronic (condition) leading to sclerodermatous changes
GVHD - Skin
Liver is second most common site
Often first increased serum levels of conjugated bilirubin and alkaline phosphatase . This reflects the pathology associated with liver (condition): damage to the bile canaliculi, leading to cholestasis
DDx includes, VOD (veno-occlusive disease), viral hepatitis, drug toxicities
Diagnosis best made by biopsy, which is technically difficult due thrombocytopenia. Often done via transjugular biopsy to minimize blood loss.
Acute GVHD- Liver
GI tract involvement is also very common
Most commonly manifests as diarrhea and abdominal cramping. Often very severe and can cause over 10L stool output per day
DDx: includes reaction to preparative regimen, antibiotics, infection –c.diff
Generally requires endoscopy and biopsy
GVHD - GI
Note apoptotic bodies in the crypts and “popcorn lesions”
GVHD Colon
Use the best matched donor
T cell depletion: ATG, alemtuzumab; but increased risk of tumor recurring
Chemotherapy ex- Methotrexate, immunosuppressants :Steroids, cyclosporine, mycophenylate. Planned gradual reduction of immunosuppression
Reduced intensity allografting with gradually increased cellular dose of transplanted cells
Prevention - Treatment of GVHD
Additional immunosuppression
First step is pulsed steroids
Increased immune suppression
Cyclosporine, tactolimus, mycophenylate
ATG
Other experimental measures
GVHD: Treatment
Obviously this represents a very aggressive therapy, and patients have to be fairly healthy
Remember for most patients this is not the first step, but the final step
Preserved cardiac, pulmonary, liver function
Chemosensitive disease at a minimum, preferably disease in remission
No substantial ongoing infections
HSCT Host factors
Evaluation of the Patient with Possible Infection The 3 Preliminary Questions to Always Ask
What type of host is being evaluated? Is the patient immunocompetent or is he/she immunocompromised (or immunoincompetent or immunodeficient)?
If the patient is immunocompromised, what is the nature of the major defect in host defenses?
Can the major defect be identified or predicted?
If more than 1 defect is likely present, is 1 dominant?
How long has the defect been present (i.e., Is a discrete time of onset identifiable?)?
An ICH has impaired resistance to infection that is secondary to:
Combination of underlying disease and treatment
Impaired resistance to infection is a consequence of disease or Rx-induced alterations in one or more of the basic host defenses:
-Neutropenia (ANC <500)
-decreased antibody production
-decreased CMI (CD4 <200 cells/cu mm)
-disrupted barriers
-hypgammaglubinemia (IgG level <800 mg/dl)
Since the treatment of established infection in the ICH still is associated with suboptimal outcomes, the primary goal of management should be:
The prevention of infection
Effective prevention of infection in the ICH is predicated upon a knowledge of the
Origin of infecting pathogens (exogenously acquired vs endogenously present)
For those organisms that are exogenously acquired, it is imperative that the route of transmission to humans be known (ie, direct contact, airborne, etc) so attempts can be made to disrupt transmission or minimize risk of exposure
Organisms native to host & present on admission (e.g. - resident flora of GI tract including Candida species)
Account for ~50% of infections
Suppressive measures
Frequent washing of skin surfaces
Administration of prophylactic antimicrobials
EPIDEMIOLOGY OF INFECTION IN THE ICH Endogenous Organisms
Organisms not part of the host’s resident microbial flora on admission (e.g. - Aspergillus)
Account for ~50% of infections
Sources of acquisition would include hands of personnel, air, food, fomites, and medical devices
Measures to reduce acquisition
Strict handwashing
Protective isolation
Special air handling systems (ie, HEPA filtration)
Special diets
EPIDEMIOLOGY OF INFECTION IN THE ICH Exogenously Acquired Organisms
less sensitive markers for infection in the overtly neutropenic.
Rubor, Calor, Tumor and Dolor

However, Fever is a reliable marker!
The most frequently occurring and reliable sign of infection in the neutropenic host
FEVER! FEVER! FEVER!

Infection may begin and progress in the absence of fever
and
Fever is not specific for infection (30% to 45% of febrile episodes may be noninfectious in etiology)
NONINFECTIOUS CAUSES OF FEVER IN ICHs Special Considerations
Underlying Diseases
Malignant neoplasms
Rheumatic disorders including vasculitis
Hypersensitivity
Drug, blood products, diagnostic agents, prostheses
Metabolic
Gout, hyperthyroidism, adrenal insufficiency
Cardiac and Vascular
Thrombophlebitis, myocardial infarction, pericarditis, arteritis
Pulmonary
Emboli, atelectasis, infarcts
Central Nervous System
Infarct, hemorrhage, post-neurosurgical inflammation
FEVER IN THE ICH
Single temperature > 38.3°C (101°F)
or
Temperature > 38°C (100.4°F) for > 1 hr
or
Temperature > 38°C (100.4°F) on 2 or more occasions within 24 hrs
Skin GI tract
Cutaneous lesions Esophagitis
HEENT Perianal infections
Eye disease CNS
Oral lesions Meningitis/encephalitis
Sinusitis Blood
Lungs Septicemia (? catheters)
Pneumonitis FUO (fever of unknown origin)
COMMON “CLINICAL SYNDROMES” IN THE IMMUNOCOMPROMISED HOST
FEVER, RESPIRATORY SYMPTOMS, and ABNORMAL CXR IN THE ICH

% infectious vs. % noninfectious causes
75% vs. 25%
Geman helmets
Seen on pneumocystis smear
ICH patients

---Fever + cough = Pneumonia
---Fever + headache = CNS infection
When you are evaluating an ICH with fever, try to identify the clinical syndrome that the patient is manifesting as that offers important insight into potentially causative organisms. The patient has fever + ……….
Pneumonia in GRANULOCYTOPENIC PATIENTS WITH ACUTE LEUKEMIA RECEIVING REMISSION INDUCTION THERAPY
Pseudomonas aeruginosa
Klebsiella pneumoniae
Staphylococcus aureus
Aspergillus fumigatus and flavus
Agents of mucormycosis
Anorectal lesions in GRANULOCYTOPENIC PATIENTS WITH ACUTE LEUKEMIA RECEIVING REMISSION INDUCTION THERAPY
Pseudomonas aeruginosa
Escherichia coli
Pharyngitis in GRANULOCYTOPENIC PATIENTS WITH ACUTE LEUKEMIA RECEIVING REMISSION INDUCTION THERAPY
Mixed flora
Normal oral flora
Gram-negative bacilli
Candida sp.
Staphylococcus aureus
Esophagitis in GRANULOCYTOPENIC PATIENTS WITH ACUTE LEUKEMIA RECEIVING REMISSION INDUCTION THERAPY
Candida sp.
Cytomegalovirus
Herpes simplex
Gram-negative bacilli
Skin lesions in GRANULOCYTOPENIC PATIENTS WITH ACUTE LEUKEMIA RECEIVING REMISSION INDUCTION THERAPY
Gram-negative bacilli
Staphylococcus aureus
Normal skin flora
Corynebacterium sp.
Staphylococcus epidermidis
UTI IN GRANULOCYTOPENIC PATIENTS WITH ACUTE LEUKEMIA RECEIVING REMISSION INDUCTION THERAPY
Stool flora
E. coli
Pathogens in the ICH
Extracellular pathogens
Pyogenic bacteria (S. aureus, GNRs)
Filamentous fungi (Aspergillus, Zygomycetes)
Candida sp.
Phagocyte cell deficiency
Pathogens in the ICH
Intracellular pathogens
Viruses (HSV, VZV, CMV)
Parasites (Toxoplasma, Pneumocystis)
Mycobacteria
Fungi (Candida, Cryptococcus)
Selected bacteria (Legionella, Nocardia)
CMI deficiency
Pathogens in the ICH

Encapsulated bacteria
S. pneumoniae
H. influenzae
Antibody deficiency
Heterogeneous group of disorders
Many (over 40)
Due to multiple stages of normal lymphocyte development
Genetic abnormalities result in uncontrolled proliferation of neoplastic lymphocytes at a particular developmental stage
Lymphomas
Primary Lymphoid Tissues
Bone marrow
Thymus
Secondary Lymphoid Tissues
Lymph nodes
Spleen
Tonsils
Clusters of lymphoid tissue in the GI and pulmonary tracts
Mature in the bone marrow
Enter the peripheral blood circulation and migrate to secondary lymphoid tissues
Normal B Cell Development
is made up of spherical clusters of B-lymphocytes called follicles
Cortex of lymph node

If the "virgin" B-cells are unexposed to antigen, they compose homogeneous primary follicles. If the B-cells have been stimulated by antigen and are in the process of proliferating and transforming themselves, they form a pale-staining germinal center surrounded by a mantle zone of smaller, darker B-lymphocytes. The whole assembly is called a secondary follicle
Lymph node area:
T cells
Antigen presenting dendritic cells
High endothelial venules
Paracortex
Lymph node region:
Plasma cells
Medullary sinuses
Medulla
Lymph node region:
Macrophages, histiocytes that capture antigen and process it
Sinuses
Naive B cells are located here
Primary follicle of lymph node
B cells that are proliferating after encountering an antigen
Naïve B cells get pushed to periphery and form the mantle zone
Have germinal centers
Dark zone: centroblasts (grow and divide)
Light zone: centrocytes (more mature and differentiating)
Tingible body macrophages: destroy B cells with “wrong” antibodies
Secondary follicles in lymph node
Normal T Cell Development
Lymphoid stem cells migrate to thymus (still a lot not known) via the peripheral blood circulation
Occurs even after puberty
Normal T Cell Development in lymph node
Cortex
Thymic epithelial cells interact with lymphocytes to help them differentiate
Physical and chemical interactions
Rapid proliferation (look like lymphoblasts) and move in toward medulla
Medulla
Final development occurs
here
Look like resting
lymphocytes
Only 5% of the cells in the
cortex make it this far
(why?)
Thymic epithelial cell (TEC) has an MHC (major histocompatibility complex) molecule on its surface
TEC presents a peptide produced from processing an antigen
Thymocyte recognizes the MHC protein (self) and the antigenic peptide (nonself) via the T cell receptor
Signal for spontaneous apoptosis is turned off
Positive selection in normal T cell development
Central tolerance:
Any thymocyte that has a high affinity for the self MHC molecule and a peptide found on the antigen presenting cells in the thymus gets apoptosis induced
Peripheral tolerance:
Many tissue specific antigens are not present in the thymus
Similar mechanism that occurs outside the thymus, except:
Cells do not undergo apoptosis, but anergy (unresponsiveness)
Why is this helpful?
Fewer autoimmune problems
Molecular mimicry may play a role – sometimes the T-cell will end up destroying same cells b/c it looks like a non-self cell – how autoimmune occurs
T cell development and negative selection
T cells primarily stay in thymus if
T cell receptor is produced from alpha and beta genes
T cells primarmily migrate to various places in the body, such as the epithelium of the GI tract (to help with mucosal defenses) if
T cell receptor is produced from gamma and delta genes
Burkitt’s Lymphoma/Leukemia
Pre-B cell ALL/Lymphoma
Pre-T cell ALL/Lymphoma
Adult T cell Lymphoma
(HTLV-1)
High grade (Highly Aggressive)
NHL
Diffuse large B cell lymphoma
Anaplastic large cell lymphoma
Mantle cell lymphoma
Intermediate grade (Aggressive) NHL
CLL/SLL
Lymphoplasmacytic lymphoma
Plasma cell myeloma
Follicular lymphoma
Mantle cell lymphoma
Marginal zone B cell lymphoma
T cell large granular lymphocyte leukemia
Mycosis fungoides
NK cell LGL
Low grade NHL
Most common low grade non-Hodgkin lymphoma
22% of all new NHL diagnoses
Incidence increases with age
Median age 60-70 years of age
Follicular Lymphoma
“Small cleaved cells”
Mature-appearing
Flow cytometry:
CD20+
CD10+
bcl-2+
CD5-
Follicular Lymphoma
B cell lineage (think of the CD20+)
consist of centrocytes,
the small cleaved cells,
and centroblasts, larger
cells that divide more

The larger the number
of centroblasts, the
more aggressive
Follicular lymphoma
0-5 centroblasts/hpf
Grade 1 Follicular Lymphoma
6-15 centroblasts/hpf
Grade 2 Follicular Lymphoma
centrocytes present OR
solid sheets of
centroblasts
Acts more like
intermediate grade
lymphomas
Grade IIIa/Grade IIIb
follicular lymphoma
Overall lymph node architecture is recognizable but…
Mantle zone is lost
Follicles start to merge together
Polarization of germinal center is lost
Paracortex is lost
Follicular lymphoma
Approximately 85% of patients with (condition) will have t(14;18)
Follicular lymphoma
Not curable
But is very treatable, ie: responsive to chemotherapy
“Reset the clock”, “mow the grass”
No definite standard of care
Treatments may range from watchful waiting to stem cell transplantation
Follicular lymphoma: treatment
FLIPI
5 adverse prognostic factors:
Age > 60 years
Ann Arbor Stage III-IV
Hb < 12 g/dl
Number of nodal areas >4
LDH > upper limit of normal
Follicular Lymphoma International Prognostic Index
Most common of the intermediate grade lymphomas
Comprises ~30% of all new NHL diagnoses
Diffuse large B cell lymphoma
CD19+, CD20+
B cell lineage
Cells are larger than a normal lymphocyte
No standard cytogenetics
Normal architecture is usually effaced
Often very responsive to chemotherapy
Diffuse Large B Cell Lymphoma
Standard treatment is R-CHOP:
DLBCL
R: rituximab (Rituxan)
C: cyclophosphamide (Cytoxan)
H: Hydroxy-doxorubicin (doxorubicin)
O: Oncovin (vincristine)
P: Prednisone
One of the fastest growing tumors that exist
# of person’s cells doubling time is 24 to 48 hours
Burkitt’s Lymphoma (Leukemia!)
3 types:
African: affects jaw or facial bone
“American”, or endemic: affects lymph nodes in abdomen, GI tract
Immunodeficiency-associated
Burkitt’s Lymphoma/Leukemia
Infection with malaria causes excess production of B cells, which are infected with EBV
Tumor cells originate from a single EBV infected B cell
African Burkitt’s Lymphoma

Noted that areas of high rates of Burkitt’s also had high rates of malaria
Children with sickle cell trait were mostly free of both malaria and Burkitt’s lymphoma
Usually 4-7 years of age
Male : female 2:1
Incidence is 50 times higher than in US
Involves bones of the jaw and other facial bones; kidneys, GI tract, other extranodal sites
EBV is almost always found
African Burkitt’s Lymphoma
Usually what we see in the US
Occurs worldwide regardless of climate
Accounts for 1-2% of lymphomas in adults and up to 40% of lymphomas in children
Involves the abdomen, ovaries, kidneys, omentum, Waldeyer’s ring, and other extranodal sites
15-30% of cases will be EBV(+)
Sporadic Burkitt’s Lymphoma
Primarily occurs in patients affected with HIV – this then becomes AIDS-defining
Also seen in allograft recipients, congenital immunodeficiency states
Accounts for 30-40% of all of NHL in HIV (+) patients
Other AIDS-defining malignancies:
Kaposi’s sarcoma
Systemic NHL, primary effusion lymphoma, CNS lymphoma
Cervical cancer
Immunodeficiency-Associated Burkitt’s Lymphoma
Diagnose with tissue
Starry sky pattern – big time board question
~100% Ki-67 staining
CD20(+), CD10(+), CD5(-)
Burkitt’s Lymphoma
All have a cytogenetic abnormality involving chromosome 8: c-myc
t(8;14): Ig heavy chain gene on chr 14
t(2;8): Kappa light chain gene on chr 2
t(8;22): Lambda light chain gene on chr 22
Burkitt's Lymphoma
Treatment must begin immediately
These patients are at extremely high risk for spontaneous tumor lysis syndrome
These like to go to CNS – must give intrathecal, so that CNS does not become a sanctuary site; testicles are other sanctuary
Burkitt's Lymphoma
Clonal proliferation of a cell line derived from the myeloid stem cell
Myeloproliferative Disorders
vHL protein targets HIF-1a
for destruction by
proteosomes
(ubiquitination)
No HIF-1a/HIF-1b complex
Epo gene not transcribed
Regulation of Normal Red Blood Cell Production - normoxia
HIF-1a is not degraded
HIF-1a/HIF-1b bind to Epo gene
Stimulate transcription of Epo
Regulation of Normal Red Blood Cell Production - hypoxia
Appropriate causes of erythrocytosis
Hypoxia:
COPD
R to L cardiac shunt
Sleep Apnea
High altitude
Increased affinity for Hb:
Chronic CO poinsoning
New kidneys keep secreting Epo:
After renal transplant
Clonal disorder
RBC production is independent of erythropoietin and its receptor
Do not need Epo to form RBCs
Blocking Epo receptor does not “turn off” RBC production
The Epo receptor has no mutations
Epo level will be low
Polycythemia Vera
>80% of (condition) patients
Valine substituted for phenylalanine at amino acid position 617 of JAK-2 (Janus activating kinase-2)
Results in constituitively active tyrosine kinase activity
Promotes cytokine hypersensitivity, or cytokine independent growth
Causes erythrocytosis
Polycythemia Vera: JAK-2
Elevated hemoglobin and hematocrit
Elevated RBC mass

~60% of patients will have a platelet count > 400K
~40% of patients will have a WCC > 12K
Why?

Bone marrow overall cellularity is increased
Polycythemia Vera: Lab values
“Congestion”:
HA, visual changes
Dizziness
Paresthesias
Facial plethora
Pruritis after a warm bath
Bleeding, bruising
Thrombosis:
MI, DVT, PE, CVA, Budd-Chiari syndrome
Hepatosplenomegaly
Erythromelalgia – painful red hands
PV: Signs and Symptoms
Increased red blood cell mass
Isotopic studies
“Very much” increased Hb and Hct
Other causes of polycythemia are ruled out
And one or more of the following:
Platelet count > 400K
WCC > 12K
Low Epo levels
Bone marrow biopsy:
Prominent erythroid and megakaryocytic proliferation
Fibrosis
Polycythemia Vera: WHO Criteria for Diagnosis
Polycythemia Vera: Natural History
Thrombotic events:
MI, CVA, DVT, PE
Risk increases with age and white cell count (marker)
Risk of transformation:
Myelofibrosis
AML
Depending on age and previous treatments
Polycythemia Vera: Treatment
Phlebotomy
Goal Hct < 45% for males; < 42% for females
Hydroxyurea
Aspirin 81 mg
Transient processes
Acute blood loss
Recovery from thrombocytopenia
Acute infection or inflammation
Response to exercise
Drug reactions
Sustained processes
Iron deficiency
Hemolytic anemia
Asplenic state
Chronic inflammatory or infectious diseases
Cancer
Thrombocytosis: General
Clonal disorder
Independent of thrombopoietin or its receptor (c-Mpl)
TPO levels are normal or elevated
Decreased clearance
JAK2 mutation seen in ~50% of patients
Essential Thrombocytosis
Sustained platelet count ≥450K
Hyperplasia of megakaryocytes on bone marrow biopsy
Absence of t(9;22)(CML) and other causes of secondary thrombocytosis
Essential Thrombocytosis: Diagnosis
ET: Blood Counts
Elevated platelet count
Normal white blood cell count
Normal hemoglobin
ET: Natural History
Bleeding due to abnormal platelet function
Thrombosis
CVA, TIA, MI, priapism
Splenomegaly
Erythromelalgia
Risk for progression to myelofibrosis or AML
ET: Treatment
Hydroxyurea
Aspirin
Clonal disorder
Excess number of circulating eosinophils
Some patients will respond to Gleevec (imatinib)
Chronic Eosinophilic Leukemia
“Scarring” of the bone marrow
Reticulin and/or collagen fibrosis
Decreased cellularity of bone marrow
Often have “dry taps”
Chronic Idiopathic Myelofibrosis: Bone Marrow
Marked splenomegaly – trying to be hematopoietic
Hepatomegaly present as well - hematopoietic
Extramedullary hematopoiesis can be found in unusual places:
Pleural effusions
Pericardial effusions
Ascites
Central nervous system
Chronic Idiopathic Myelofibrosis: Clinical Presentation
Chronic Idiopathic Myelofibrosis: Blood Counts
Leukoerythroblastic picture:
Pseudo-Pelger-Huet cells
Giant platelets
All signs of marrow replacement
Patients are usually anemic
WCC and platelet count may be high or low
Chronic idiopathic myelofibrosis:
factors
JAK2 mutations also seen in ~50% of patients
Risk of leukemic transformation
Usually myeloid
Can be lymphoid, erythroid, megakaryocytic, or mixed lineage
Chronic Idiopathic Myelofibrosis: Treatment
Palliative
Hydroxyurea
Splenectomy
Appropriate acute leukemia treatments with transformation (prognosis worse than de novo leukemia patients)
Ineffective hematopoiesis
Cells do not progress through the normal stages of maturation
Peripheral blood: cytopenias
Bone marrow: hypercellular, with abnormal cells
How do you think patients will present?
Myelodysplastic Syndromes
MDS: Clinical Presentation
Recurrent infections
Fatigue, pallor
Bleeding
Usually don’t have splenomegaly (unlike myeloproliferative disorders)
Bone marrow biopsy and aspirate
Look for dysplastic cells
Look for an increased number of blasts
< 5% blasts: Normal
>20% blasts: Acute leukemia
6-19% blasts: (condition) (does not need to be present but is diagnostic)
Also evaluate chromosomes (with cytogenetics and sometimes FISH)
Helps with prognosis
MDS: diagnostic
Two greatest risk factors for developing AML
from MDS are:
Age
IPSS score
Cytogenetics: (MDS type)
Clinical course tends to be relatively more benign
Overall more responsive to certain treatments
Thalidomide
Lenalidomide
MDS: 5q- Syndrome
MDS: Treatment
Supportive care
Antibiotics, transfusions
Growth factors: EPO, GCSF
Iron chelators (Exjade [deferasirox])
Binds iron which gets excreted in urine and bile
Chemotherapy
Different from AML chemotherapy
Monitor for transformation to AML
Remember outcomes are worse for patients with AML arising from MDS
Genetic mutation results in increased numbers of (condition cells) with increased amounts of antibody production
Excess antibodies can cause end organ dysfunction
Get continuum of disorders depending on amount of excess protein present
Plasma cell dyscrasias
Plasma Cell Dyscrasias: A Continuum
MGUS (monoclonal gammopathy of undetermined signifance) to
Mutliple myeloma to
Plasma cell leukemia
Plasma Cell Dyscrasias: Diagnosis
To measure the number of plasma cells:
Bone marrow biopsy and aspirate
To measure the amount of protein (antibody)
Comprehensive panel
Total protein elevated in excess of albumin
Serum quantitative immunoglobulins
Serum protein electrophoresis
Serum immunofixation
Serum free light chains
Tells us how much immunoglobulin a patient has
IgG 3404 mg/dl
IgA 260 mg/dl
IgM <12 mg/dl (for example)
Can we tell if this is a monoclonal excess of IgG by this information? No
Quantitative Immunoglobulins used in concert with SPEP
Clonal disorder
One narrow peak
All the proteins are the same so they travel the same distance on the electrophoresis gel
Quantitates the M spike
0.40 g/dl, for example
Can we tell if this is IgG (or IgA or IgM)? No
SPEP: Monoclonal Gammopathy used in concert with quantitative immunoglobulins
Run patient’s serum on gel
Stain for different antibodies using specific reagents
Confirms clonality
Tells us which antibody is in excess
IgG kappa, for example
IEP: Monoclonal Gammopathy
Get quantification of amount of light chains in the serum
Free kappa, serum: 1720.0 mg/L
Free lambda, serum: 1.8 mg/L
Free kappa/lambda ratio: 15.9 (normal 2:1)
For example
Serum Free Light Chains
MDS End Organ Damage: Bones
Increased osteoclast activation
Leads to lytic lesions in the bones
Calvarium
Spine
Ribs
Pelvis
Long bones
MDS End Organ Damage: Kidneys
Deposition of Ig
Cast nephropathy "Myeloma kidney"
Infitration of kidney by plama cells
*Think Bence-Jones proteinuria
MDS End Organ Damage:
Bone Marrow
Increased numbers of plasma cells in the marrow
Normochromic normocytic anemia
Circulating plasma cells usually only seen with plasma cell leukemia
Classic changes seen on peripheral smear - Rouleaux formation due to extra monoclonal proteins
MDS End Organ Damage: Electrolytes
Hypercalcemia
Due to osteoclast activation
Contributes to renal dysfunction
“Stones, bones, groans, and moans” (applicable to all hypercalcemic states)
Stones: kidney stones
Bones: increased bone rebsorption
Groans: constipation
Moans: psychiatric issues
Plasma Cell Dyscrasias: Treatment
Depends on where the patient falls along the continuum
Observation
Oral chemotherapy
Intensive IV chemotherapy
Transmembrane protein
Found everywhere
Except in the bloodstream
Deletion is lethal (can create mouse with 1% tissue factor, but can’t live w/o any)
Cofactor for Factor VIIa activity
Factor X activation – static system
Factor IX activation – flowing system
TF-VIIa is inhibited by TFPI
Trigger is Tissue Factor
TF-VIIa is inhibited by
TFPI - Tissue factor pathway inhibitor
Major coagulation effector enzyme
Converts Fibrinogen to Fibrin
Activates Factor XIII
FXIIIa crosslinks Fibrin

Activates Platelets
Thrombin
Soluble, digested by plasmin
Fibrinogen
Weak clot, easily digested by plasmin
Fibrin
Strong clot, can be digested by plasmin
Gamma crosslinked Fibrin
Linear linking between fibrin segments with Factor XIIIa
Strong clot, resists plasmin digestion
Alpha crosslinked Fribrin
3-D linking between fibrin segments with Factor XIIIa
Vitamin K-based clotting factors and anti-coagulant factors
II, VII, IX, X
Protein C and Protein S
Thrombin and AT3 in the presence of what creates what?
UF Heparin
Thrombin anti-thrombin
FXa and AT3 in the presence of what creates what?
UF and LMW Heparin
X anti-thrombin
FIXa and AT3 in the presence of what creates what?
nothing
IX anti-thrombin
Thrombin and Thrombomodulin combine to create (part of protein C system)
TM IIa

Loses the ability to activate factor V, X, platelets and fibrinogen to fibrin
TM-IIa is used to convert what?
Protein C to aPC
What does aPC do?
aPC kill the amplifier FVa to FVi in the presence of Protein S (and some VIIIa)
Triggered by aPC

Plasmin is the effector enzyme
Relatively non-specific serine protease

Fibrin specificity is conferred by activation mechanism
Fibrinolysis
aPC goes to aPC receptor on endothelial cell - endothelial cell releases
tPA
converts Plasminogen to Plasmin
Plasmin converts what to what
Fibrin to FDP (d-dimers)
Severe deficiency of a single factor
Hemophilia
Immune inhibitors
Combined deficiency of many factors
Vitamin K deficiency (2, 7, 9, 10)
Liver disease (everything but 8)
Anticoagulation
Warfarin
Heparin
Argatroban, Refludan, Angiomax
Excess Fibrinolysis
Liver disease
Thrombolytic therapy
Decreased thrombin/plastin ratio - defective hemostasis
Flow velocity
Laminar versus turbulent
Vessel diameter
Blood viscosity
Shear forces
Pressure difference across the vascular defect
Pressure gradient
Hydraulic forces
In a closed space, as bleeding continues, external pressure rises to match internal pressure
Effectiveness depends on pressure difference
If external pressure rises above a critical value all blood flow ceases and other structures are compromised giving a “compartment syndrome” – particularly in the limbs
Tamponade
If external pressure rises above a critical value all blood flow ceases and other structures are compromised giving a
“compartment syndrome” – particularly in the limbs
Shear forces increase as
Linear velocity increases
Activated coagulation factors are washed away
Viscosity increases
Vessel radius decreases
Flow changes from laminar to turbulent
High flow rates
Vessel irregularities
Virchow's triad
Alteration in the vessel
Damaged endothelium
Exposes tissue factor
Exposes collagen/VWF
Alteration in the blood
Increased thrombin generation
Decreased plasmin response
Stasis
Valve pockets
Area of hypoxia
Atherosclerotic plaques as focus
Tissue factor exposure by smooth muscle cells (at base of ulcerated plaque)
Platelets are central
High shear setting
vWF involved
Coagulation involved in thrombus growth
Arterial thrombosis
Inherited
Protein C, Protein S, AT3 deficiencies
FV Leiden, G20210A mutations
Congenital
Factors II, VII, VIII, IX, XI, VWF, homocysteine
Acquired
Age
Lupus anticoagulant, DIC
Obesity, sedentary lifestyle, smoking
Malignancy, surgery, pregnancy
HIT/HITT
Thrombophilic Factors
TF on circulating monocytes
Activation by lipopolysaccharide – sepsis
Disseminated intravascular coagulation - DIC
AT3 is adequate to control the coagulation cascade
But:-
Factors V and VIII are being activated to FVa and FVIIIa by thrombin
Procoagulant effectiveness increases
More thrombin produced for a given stimulus
Circulating platelets are activated
Risk of thrombosis is increased
DIC Stage 1
DIC Stage 1 (The Hypercoagulable State – this would almost be considered pre-DIC) Treatment
Control coagulation to regulate process
Low dose heparin
Amplifies AT3 effectiveness
Shuts down thrombin generation
Reduces risk of thrombosis
Anti-platelet agents
GP IIb/IIIa inhibitors block platelet aggregation
ASA (aspirin), Clopidogrel (Plavix) block platelet activation
AT3 has been consumed
Excess thrombin is produced
The protein C system becomes activated
aPC inactivates FVa and FVIIIa
FV and FVIII deficiencies result
Activated platelets are removed from the circulation
Thrombocytopenia
Risk of bleeding is increased (FV and FVIII deficiency and lack of platelets)
DIC Stage 2 (Early or Mild DIC/Consumption Coagulopathy)
DIC Stage 2 (Early or Mild DIC/Consumption Coagulopathy) Treatment
Replace missing coagulation factors V and VIII
Cryoprecipitate
FVIII and Fibrinogen
Platelets
FV
Replace AT3
Concentrate or bank plasma
Then low dose heparin to control thrombin generation
Increases risk of bleeding
Antiplatelet agents
Increases risk of bleeding
aPC inhibitor has been consumed
Fibrinolysis has been activated
Free Plasmin
Dissolves hemostatic plugs
Reduces the thrombin/plasmin ratio
Digests most coagulation factors
Digests Fibrinogen
Digests platelet surface receptors
Defective hemostasis and rebleeding of old wounds
DIC Stage 3 (Late or Severe DIC/Consumption Coagulopathy)
DIC Stage 3 (Late or Severe DIC/Consumption Coagulopathy) Treatment
Need to replace coagulation factors
Plasma
Concentrates
Cryoprecipitate
Platelets
AT3
Prothrombin complex
Need to inhibit Thrombin
Heparin and friends
High risk of fatal bleeding
Need to inhibit plasmin
AMICAR
High risk of fatal thrombosis
What is the most frequent site of bleeding in hemophilia patients?
Joints/Hemarthrosis
What do you do to stop the bleeding in the psoas muscle of a patient with severe congenital Factor VIII deficiency?
Use rFVIII
Most common hemophilia?
Hemophilia A/Factor VIII deficiency
Factor VIII gene located near the tip of the long arm of X chromosome
Missense or frameshift mutations, deletions or inversions
Defect is an absence or low level of Factor VIII
Incidence is 1 in 5,000 live male births
Hemophilia A
Recurrent bleeds leading to persistent inflammation, soft tissue, bone destruction
Recurrent pain, chronic arthritis
Decreased range of motion and mobility
Eventual physical impairment, long-term disability, psychosocial consequences
Hemophilic Arthrophathy
synovitis and increased blood flow to the joint may lead to
epiphyseal overgrowth and limb length discrepancy
Factor IX deficiency
Same clinical manifestations as
Hemophilia A
Less common (1 in 25,000 births)
X-linked mutation/deletion in Factor IX gene (first described in Christmas family)
Hemophilia B
Bleeding Time/PFA-100 – normal (tests skin, vessels and primary hemostasis)
Platelet Count - normal
Prothrombin Time – normal (extrinsic pathway)
Activated Partial Thromboplastin Time – long
Factor VIII Assay
Severe <1%
Mild >5%
LABORATORY FINDINGS IN HEMOPHILIA A
Hemophilia Treatment Centers
Plasma derived or Recombinant factors to replace the missing factor
Clotting factor concentrates are given to prevent bleeding and to limit existing hemorrhage
Synovectomies, joint replacements/fusions
PREVENTION, PREVENTION, PREVENTION
Treatment of Hemophilia
-purified from the cell culture of transfected mammalian cell lines
-require no further viral attenuation
Recombinant human factor VIII and IX
-large starting pool of carefully screened donor plasma
-Affinity chromatography using monoclonal antibodies
-Viral inactivation procedures (pasteurization, solvent-detergent treatment, ultrafiltration) are effective against HIV and hepatitis viruses.
Plasma-derived concentrates
Development of inhibitors (alloantibodies) to Factor VIII and less commonly, Factor IX (about 10-20%)
Infectious
Hep B (70-90% prior to vaccine)
Hep C (>90% in pts treated prior to 1985)
HIV (By 1984, >90% of severe Hemophilia A)
CMV, Parvovirus B19
Complications of hemophilia treatment
development of neutralizing and clearing anti-factor VIII (FVIII) antibodies in individuals without a preexistent congenital FVIII deficiency.
uncommon - incidence of 0.2 to one cases per million population per year
Older adults
Postpartum
50% with underlying autoimmune DO (lupus, rheumatoid arthritis), malignancy
50% idiopathic
Acquired Hemophilia
Heterogeneous group of inherited or acquired bleeding disorders
Bleeding due to reduced level or abnormal function of (name)
The most common bleeding disorder
Most types have autosomal dominant inheritance
von Willebrand's Disease
Promotes platelet adhesion to damaged endothelium and to other platelets

Carrier molecule for Factor VIII (increases half-life of Factor VIII)
Hemostatic function of vWF
DDAVP = 1-deamino-8-D-arginine vasopressin = synthetic analog of ADH L-vasopressin
Releases stored Factor VIII and VWF from endothelial cells (you get tachyphylaxis – lose drug effect)
Humate P – Intermediate purity factor VIII concentrate which contains both VWF and factor VIII
Treatment of vWD
Drug that stabilized fibrin clot- stops fibrinolysis
Amicar
behavior pattern of overwhelming involvement with obtaining and using a drug. Will occur in < 1% of patients receiving pain therapy with opioid analgesics.
Addiction
withdrawal symptoms appear upon stopping drug
Physical dependence
when a particular dose loses its effectiveness
Tolerance
Therapeutic principles of pain managment administration
Select appropriate drug
Select appropriate dose
Select appropriate interval and route
Prevent persistent pain and relieve breakthrough pain
Titrate dose
Use adjuvant measures
Prevent, anticipate, and manage side effects
Defined as 1-2,3-4 on a 0-10 point VAS
Acetaminophen
Non steroidal anti-inflammatory drugs (NSAIDs)
Mild opioids combinations (Darvocet)
Mild pain
Defined as 5-6 on a 0-10 VAS
Small doses of morphine or oxycodone alone
Combination agents may work
Moderate pain
Defined as 7-10 on 0-10 VAS
Morphine is drug of choice
Sever/debilitating pain
Step 1 analgesic, co-analgesic
Analgesic and antipyretic effects
Mechanism: inhibits prostaglandins in CNS and peripherally blocks pain impulse generation
Onset of action is 30 – 60 minutes
Duration of action is 4 hours
Metabolized in liver, excreted in urine
Well tolerated without common toxicities expected with NSAIDs
Available in many different dosage forms
Max daily dose is 4 grams
Acetaminophen
Analgesic, antipyretic and anti-inflammatory effects
Inhibits cyclo-oxygenase (COX)
enzyme that releases PG known to sensitize or activate peripheral nociceptors
Vary in COX-2 selectivity
Dose-response curve plateaus
All the drugs in this class exhibit an analgesic ceiling effect
Particularly helpful in bone pain
NSAIDs

Limitations:
Gastropathy
Anti-platelet effect
Renal toxicity
Drug interactions
Patients where an NSAID is indicated with:
History of GI ulcers
Elderly
Low platelet count < 50K
Receiving anticoagulation
Receiving corticosteroids
Coagulopathy
Prior intolerance to non-selective NSAID
Cox-2s
Celecoxib (Celebrex)
Familial adenomatous polyposis (FAP)
400 mg PO BID
Many drug interactions that should be watched (warfarin, CYP450)
Caution in pts with sulfonamide allergy
Binds to the mu-opiate receptors but also binds to norepinephrine and serotonin
50-100 mg PO every 4-6 hr (max 400 mg/day)
Needs to be adjusted in renal dysfunction (CrCl < 30 ml/min)
Limited by N/V, constipation, and dizziness; may lower seizure threshold
Tramadol (Ultram)
Gold standard for pain management
Multiple dosage forms
PO (IR and SR), IV, PR (rectal)
Active metabolite
morphine-6-glucuronide
Well tolerated
Release of histamine
Adverse events: sedation, urinary retention, decreased respirations, CONSTIPATION (must put them on a laxative)
Initial dosing is based on severity of pain usually 1 mg IV q4h as needed
Inexpensive
Morphine
Slightly more potent than morphine
Fewer dosage forms
PO (IR and SR)
Multiple combination products
Milder side effect profile relative to morphine
Street value must be considered (Hillbilly heroin)
Oxycodone
Approximately 6 times more potent than morphine
Usual starting dose is 0.2 mg IV q3h prn
Available as both IV and PO product
No PO SR dosage form
Useful alternative in liver/renal failure patients
No active metabolite
Less nausea/vomiting and pruritis relative to morphine
Hydromorphone (Dilaudid)
Available: IV, PO (IR & ER); 10x more potent than morphine
Changing from IV to PO
10 x IV dose divided in two doses
Changing between immediate release and extended release
2 times the IR dose
Really only used in pain clinic or by oncologists
Oxymorphone (Opana)
100 times as potent as morphine; titrate slowly
1 mg IV morphine = 10 mcg
Unique dosage forms
IV, transdermal (absorbed more under heat), lozenge
Least likely to induce histamine release
It is not known whether the dose requires adjustment for renal or hepatic failure; Renally eliminated and hepatically metabolized
Fentanyl
Cheap form of pain management
Dosing is complex because of longer half lives with administration
Should only be dosed with advisement of practitioner with experience (pain control or palliative care)
Methadone
Major Adverse Effects of Narcotics
CNS
Respiratory System
Urinary Effects
CV Effects
CONSTIPATION!!!!
Must give bowel stimulant not just stool softener
Sennakot with docusate 2 tablets at bedtime or
Miralax at bedtime
complexes with Antithrombin III accelerating its ability to inactivate factors IIa, Xa, and IXa.
Heparin
Monitoring:
check aPTT (activated partial prothrombin time) at 6 hours and adjust dose to keep aPTT within the therapeutic range
Recheck aPTT every 6 hours for 1st 24 hours then every morning unless outside therapeutic range
Monitoring of unfractionated heparin
Improved bioavailability and more predictable pharmacokinetics
Do not have to monitor aPTT
Longer half life
Good for treatment of home DVT or uncomplicated PE
Use in caution in obese patients and those with renal insufficiency
LMW Heparin
Enoxaparin (Lovenox)
Dalteparin (Fragmin)
Fondaparinux (Arixtra)
LMW Heparin choices - all administered SQ
inhibits the production of vitamin K dependent clotting factors (II, VII, IX, X)
It will take at least 5-7 days to reach steady state
Most bridge therapy with heparin product when treating
If load patients with (drug) – protein C and protein S will be gone (natural anti-coagulants)  could produce hypercoagulable state
Skin necrosis – bruising on buttocks, thighs, penis, breasts
Adverse events: bleeding
Warfarin
Check international normalized ratio (PT-INR)
The goal INR depends on the indication
A fib goal 2-3
Mechanical Heart valves
aortic position  2.0—3.0
mitral position  2.5 – 3.5 or 2.0—3.0 + ASA 80-100 mg
additional RFs, or systemic embolism despite adequate anticoagulation  2.5-3.5 + ASA 80-100mg
Bioprosthetic heart valves
Aortic or mitral position  2.0—3.0 for 3 months followed by ASA 162 mg daily
History of systemic embolism  2.0—3.0 for 3-12 months followed by ASA 162 mg daily
Monitoring of Warfarin
Drug Interactions with Warfarin
Highly protein bound
H2 blockers/PPIs
Antibiotics
Antiepileptic Agents
Barbiturates
Carbamazepine
Phenytoin
Lipid lowering agents
Antifungal agents
Leukotriene Inhibitors
Antidepressants
MOA: inhibits cyclooxygenase which inhibits formation of
Thromboxane-TXA2 (vasoconstrictor; irreversible) inhibits platelet aggregation and activation
Increases risk for bleeding
prostacyclin (PC; vasodilator; reversible)
Major Adverse effects: GI Bleed, renal insufficiency
Doses: 81-325 mg PO daily
Used for post-heart attack and stroke
Aspirin
MOA: inhibits platelet activation and aggregation but mechanism is different from any other antiplatelet drug
Onset of action is 24-48 hours
Side effects: diarrhea, rash, reversible neutropenia (2%)
Drug interactions: Substrate of CYP3A4 inhibits CYP2C19 strong
Dose: 250 mg twice daily with food
Monitoring: CBC with diff q 2 weeks for 1st 3 months
Ticlopidine (Ticlid)
MOA: blocks ADP receptors  inhibits platelet activation and aggregation
Prevents activation of GPIIb/IIIa
Platelets that are affected last the rest of the platelet life
Steady state: 3-7 days
Drug interactions: minor CYP3A4
Dose: load 300 mg and then 75 mg daily with or without food
MP: routine CBC monitoring not necessary
Clopidogrel (Plavix)