Other 13 Flashcards

1
Q

CD34

A

Antigen/marker of HSC cells

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

CD45

A

leukocyte common antigen – found on all white cells

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

Hematopoietic stem cells

A

Pluripotent == can generate all mature blood cells
mature blood elements are terminally differentiated cells with finite life spans
One cell → all lineages
Capacity for self-renewal
Common origin of all formed blood elements
Myeloid: erythrocytes, granulocytes, monocytes, platelets
MMEG == Monocytes, megakaryocytes, erythrocytes, and granulocytes
this is how Putthoff refers to Myeloid derived cells
MPO (peroxidase) positive
Lymphoid: lymphocytes
TdT positive (pre-B and pre-T cell marker

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

HSC location

A

3rd week of development: transient in the yolk sac; definitive hematopoiesis in the mesoderm of the intraembryonic aorta/gonad/mesonephros region
3rd month of development: Migration to liver (also seen in placenta)
liver == chief sit of blood cell formation until shortly before birth
4th month of development: Migration to bone marrow (entire skeleton)
Puberty: Restricted to axial skeleton

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

Bone marrow sinusoids

A

Network of thin-walled sinusoids lined by a single layer of endothelial cells on top of a discontinuous basement membrane and adventitial cells
There are clusters of HSC and fat cells that sendoff differentiated cells via transcellular migration
Megakaryocytes lie next to sinusoids and extend out cytoplasmic processes that bud off into the blood stream to release platelets
RBC precursors surround macrophages (nurse cells) that provide the iron to make the hemoglobin

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

Extramedullary hematopoiesis

A

Extramedullary Hematopoiesis
In stress conditions, HSC migration occurs and tissues other than the bone marrow can provide adequate environment to allow hematopoiesis to occur
Liver and spleen

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

Erythropoietin

A
Regulates RBC progenitor cells
Produced by peritubular capillary lining cells in the kidney
Relative to the PO2
Released constantly at Hb > 10
Released logarithmically at Hb < 20
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

Leukoerythroblastosis

A

processes that distort the marrow architecture, such as deposition of metastatic cancer or granulomatous disorders, can cause the abnormal release of immature precursors into the peripheral blood

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

Marrow aspirate

A

Allows for the best assessment of the morphology of hematopoietic cells
immature precursors (-blasts) forms are identified with lineage-specific antibodies and histochemical markers
Mature marrow precursors are identified based on morphology alone
Normal adults: fat:hematopoietic elements = 1:1
Hypoplastic states: proportion of fat cells is greatly increased
Hyperplastic states: fat cells disappear

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

Normal % of T cells of lymphocytes in peripheral blood

A

80% are T cells

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

WBC disorders

A

Proliferative: expansion of leukocytes
Reactive (infection, inflammation) – fairly common
Neoplastic – much more important to recognize
Leukopenia: deficiency of leukocytes

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

Leukopenia

A

Abnormally low WBC count
Often a result of reduced neutrophils (so leukopenia is usually a neutropenia/granulocytopenia)
Lymphopenia is less common
If present, often due to HIV infection, after glucocorticoid or cytotoxic therapy, with autoimmune disorders, malnutrition, acute viral infections (redistribution of lymphocytes because type I IFNs lead to the sequestration of activated T cells in lymph nodes and increased adherence to endothelial cells

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

Neutropenia

A

Neutropenia: reduction in the number of neutrophils in the blood
Most common cause of agranulocytopenia is drug toxicity

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

Agranulocytosis

A

Agranulocytosis: clinically significant reduction in neutrophils
patients have increased susceptibility to bacterial and fungal infections
Most commonly due to drug toxicity: idiosyncratic and unpredictable due to metabolic polymorphisms or auto-antibodies (chloramphenicol, sulfa, chlorpromazine, thiouracil, phenylbutazone)
chloramphenicol == aplastic anemia and gray baby syndrome

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

Pathogenesis neutrophils

A

Pathogenesis
Inadequate or ineffective granulopoiesis
Suppression of HSC’s (aplastic anemia, infiltrative marrow disorders)
granulocytopenia, anemia, and thrombocytopenia
Suppression of committed granulocytic precursors due to drugs – most common cause
Disease states → ineffective hematopoiesis (megaloblastic anemia, myelodysplastic syndromes)
Congenital conditions (Kostmann syndrome: inherited defects in specific genes impair granulocytic differentiation)
Increased destruction or sequestration of neutrophils in the periphery
Immunologically mediated injury (idiopathic, related to autoimmune disorder, drug-related)
Splenomegaly → splenic enlargement leads to sequestration of neutrophils and modest neutropenia
associated with anemia and thrombocytopenia
Increased peripheral utilization (Overwhelming bacterial, fungal, or rickettsial infection)

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

Agranulocytosis morphology : hypocellularity in the bone marrow

A

Hypo- or hypercellularity in the bone marrow
Hyper: excessive destruction of the cells in the periphery, neutropenias caused by ineffective granulopoiesis (i.e. megaloblastic anemias and myelodysplastic syndromes)
Hypo: agents that suppress or destroy granulocytic precursors

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

Agranulocytosis morphology infections

A

Leads to infections with ulcerating necrotizing lesions of the oral cavity – agranulocytic angina
Deep lesions covered by dark necrotic membranes from which bacteria or fungi can be isolated
Infection can also occur in other locations
Increased risk of infection by Candida and Aspergillus

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

Clinical agranulocytosis

A

Signs and symptoms related to infection: Malaise, chills, fever followed by marked weakness and fatigability
Overwhelming infections can cause death within hours to days
Serious infections occur with a count < 500/mm3

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

Treatment agranulocytosis

A

Broad spectrum antibiotics

GCSF administration to stimulate production of granulocytes from marrow precursors

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

Leukocytosis

A

Increase in the number of WBCs in the blood
Common in inflammatory states
leukemoid reaction == elevated LAP
leukemia == normal LAP
Driven by production of cytokines (TNF, IL1), growth factors and adhesion molecules
May include d, eosinophilia, basophilia, monocytosis, lymphocytosis

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

Peripheral leukocyte count influences

A

Related to size of precursor in marrow pool, circulation and peripheral tissues
Rate of release of cells from the storage pools into the circulation
Proportion of cells adherent to the blood vessel walls (marginal pool)
Rate of extravasation of cells from the blood into tissues

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

Role of infection on leukocytosis

A

TNF and IL-1, if sustained, can cause there to be an egress of mature granulocytes out of the bone marrow and for there to be increased production of growth factors
IL5: Stimulates production of eosinophils
G-CSF: Stimulates production of granulocytes (neutrophils

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

Sepsis of severe inflammatory disease on leukocytosis

A

In sepsis or severe inflammatory diseases, can be accompanied by morphologic changes in neutrophils
Toxic granulations: coarser and darker, abnormal azurophilic (primary) neutrophilic granules
due to strong response to infection
Dohle bodies: patches of dilated endoplasmic reticulum seen as sky-blue cytoplasmic “puddles”
seen in neutrophils
due to strong response to infection

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

Leukomoid rection : many immature granulocytes

A

Leukemoid Reaction: many immature granulocytes appear in the blood due to an infection; looks like myeloid leukemia
LAP will be elevated in leukemoid reaction
LAP will be normal in leukemia

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

Lymphopenia causes

A

Decreased number of circulating lymphocytes
Immunodeficiency (Di George syndrome [chromosome 22q11 deletion])
High cortisol state (Cushing syndrome)
Autoimmune destruction (SLE)
Whole body radiation

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

Neutrophilic leukocyteis causes

A

Increased circulating neutrophils
Bacterial infection or tissue necrosis (left shift) with Decreased CD16 and Fc receptors
High cortisol state

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

Eosinophilic leukocytosis causes

A
Increased circulating eosinophils
Allergic reactions (type I hypersensitivity)
Parasitic infections
Hodgkin lymphoma
Drug reactions
Some vasculitides
*increased IL5 --> eosinophils
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
28
Q

Basophils leukocytosis

A

Increased circulating basophils
CML (chronic myeloid leukemia)
Rare, except in CML ^^

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

Monocytosis causes

A
Increased circulating monocytes
Chronic inflammatory states (autoimmune or infectious)
TB, bacterial endocarditis, malaria
Malignancy
Collagen vascular disorders (SLE)
Inflammatory bowel diseases
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Lymphocytic lymphocytosis causes

A
Increased circulating lymphocytes
Viral infections (HAV, CMV, EBV)
Bordetella pertussis infection
Chronic immunologic stimulation (TB, brucellosis)
Often accompanies monocytosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
31
Q

Germinal centers

A

Develop in lymph nodes within several days of antigenic stimulation
due to enlargement of primary follicles; pale staining
Highly dynamic structures where B cells acquire the capacity to make high-affinity antibodies against specific Antigens
Dark zone of proliferating centroblasts (blast like B cells)
Light zone of centrocytes (B cells with irregular or cleaved nuclear contours

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

Acute nonspecific lymphadenitis : localized

A

Localized: Direct microbiologic drainage (i.e. cervical region due to dental or tonsillar infection; axillary or inguinal regions due to infections in the extremities

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
33
Q

Acute nonspecific lymphadenitis systemic

A

Systemic: Associated with bacteremia and viral infections (particularly in children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
34
Q

Mesenteric acute nonspecific lymphadenitis

A

Mesenteric: An infection that causes mesenteric lymph nodes to enlarge and become tender
Can look like appendicitis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
35
Q

Morphology acute nonspecific lymphadenitis

A

Large germinal centers with numerous mitotic figures
Nodes are swollen, gray-red and engorged
Presence of macrophages with particulate debris
In severe infections the entire lymph node can become necrotic or a bag of pus
Less severe infections have scattered infiltrates of neutrophils that accumulate in the lymphoid sinuses
Endothelial cell hyperplasia (lining the sinus

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
36
Q

Calinical acute nonspecific lymphadenitis

A

Clinical
Affected nodes are enlarged and tender; may become fluctuant (moveable) when abscess formations is extensive; overlying skin is red
suppurative infections that penetrate the lymph node capsule & track to the skin –> draining sinuses –> scar

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
37
Q

Chronic nonspecific lymphadenitis

A

Nontender lymph nodes without acute inflammation or tissue damage
Tender LAD == acute process (infection)
nontender LAD == chronic process (cancer)
Common in inguinal and axillary lymph nodes as they drain large areas of the body and are stimulated by trivial injury/infections of extremities

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
38
Q

Tertiary lymphoid organs

A

chronic immune reactions can promote the appearance of organized collections of immune cells in nonlymphoid tissues
chronic gastritis caused by Helicobacter pylori infection –> peyer’s patch hyperplasia
can lead to MALToma (marginal cell lymphoma)
rheumatoid arthritis –> B-cell follicles appear in the inflamed synovium
can lead to MALToma (marginal cell lymphoma)
lymphotoxin is probably involved

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
39
Q

Follicular hyperplasia

A

due to stimuli activating the humoral immune response
Presence of large oblong germinal centers (2° follicles) surrounded by a collar of small resting naive B cells (mantle zone)
Germinal centers contain centroblasts (dark) and centrocytes (light)
Dendritic cells and tingible-body macrophages are interspersed
tingible-body macrophages == macrophages that have phagocytosed the apoptotic B-cells
May be due to rheumatoid arthritis, toxoplasmosis, or early HIV – B cell responses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
40
Q

Tangible body macrophages

A

Tingible-body macrophages
Contain the nuclear debris of B cells
B cells undergo apoptosis if they fail to produce an antibody with a high affinity for antigen
Seen in ALL and Burkitt Lymphoma
tingible body == apoptotic B cells in a macrophage

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
41
Q

Reactive follicular hyperplasia

A

Preservation of lymph node architecture – neoplastic means things are going awry
Marked variation in follicular shape and size
Frequent mitotic figures, phagocytic macrophages and recognizable light (-cyte) and dark (-blast) zones

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

Paracortical hyperplasia

A

due to stimuli triggering the T-cell mediated immune response (e.g. acute viral infection)
infectious mononucleosis (EBV)
In exuberant reactions, the T-cell zones (containing immunoblasts) may encroach on the B-cell follicles
Hypertrophy of sinusoidal and vascular endothelial cells
May be accompanied by infiltrating macrophages and eosinophils

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
43
Q

Imunoblasts (dark staining)

A

Activated T cells 3-4 times the size of resting lymphocytes
Round nuclei, open chromatin, several prominent nucleoli, moderate amounts of pale cytoplasm
If numerous, exclude lymphoid neoplasm from differential diagnoses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
44
Q

Sinus histiocytosis

A

inus Histiocytes (aka reticular hyperplasia)
Increase in the number and size of cells that line lymphatic sinusoids
Nonspecific, but can be prominent in lymph nodes draining cancers such as carcinoma of the breast
lymphatic endothelial cells are markedly hypertrophied and significant increases in macrophages occurs
Expansion and distension of the sinuses
sinus histiocytosis does not mean metastatic cancer; these lymph nodes are reactive; sinus histiocytosis is a reactive phenomenon in a lymph node draining cancer
histiocytosis X is dendritic cell proliferation – what does the X stand for??

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
45
Q

Hemophagocytic lymphohistiocytosis

A

Definition: macrophage activating syndrome
Reactive condition with cytopenias and signs and symptoms of systemic inflammation due to macrophage activation
Familial (early onset) and sporadic (any age

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
46
Q

Pathogenesis hemophagocytic lymphohistiocytosis

A

Systemic activation of macrophage and CD8+ cytotoxic T-cells
macrophages phagocytose blood cell progenitors in the marrow and formed elements in peripheral tissues
Mediators released from macrophage and lymphocytes suppress hematopoiesis and produce signs and symptoms of systemic inflammation
“Cytokine storm” or SIRS can occur due to the cytopenias
SIRS criteria
Hyper/Hypo-thermia
Tachycardia
Tachypnea
Leukocytosis/leukopenia
Appear shock-like
unbridled HLH is associated with extremely high levels of inflammatory mediators such as IFNγ, TNFα, IL-6, and IL-12, as well as soluble IL-2 receptor
infection is the most common trigger for HLH, especially Epstein-Barr virus (EBV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
47
Q

Clinical hemophagocytic lymphohistiocytosis

A

patients present with acute febrile illness and hepatosplenomegaly
Hemophagocytosis may be seen on bone marrow exam – neither sufficient nor required to make diagnosis
Anemia, thrombocytopenia,
increased plasma ferritin and soluble IL-2 receptor (indicators of severe inflammation)
Elevated LFT’s and TGs (related to hepatitis)
coagulation studies may show evidence of DIC
Untreated = rapid progression to multiorgan failure, shock, death

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
48
Q

Treatment HLH

A

Immunosuppressive drugs and “mild” chemotherapy
Germline mutations or persistent/resistant disease? HSC transplantation
Treated: 50% survive
May have significant sequelae (renal damage in adults, growth and mental retardation in children)
Untreated: grim prognosis, < 2 months survival if familial

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
49
Q

Familial hemophagocytic lymphohistiocytosis

A

Mutations impact the ability of cytotoxic T cells and NK to properly form or deploy cytotoxic granules
Uncontrolled disease
Greatly increased IFNγ, TNFα, IL6, IL12 and soluble IL2 receptor
familial form is more severe

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

Leukocyte alkaline phosphatase (LAP)

A

Elevated in reactive WBC disorders
LAP will be high in reactive states, e.g. infection
Low in myeloproliferative and neoplastic WBC disorders

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

WBC neoplasma

A

WBC Neoplasms
Lymphoid
Myeloid
Histiocytoses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

Lymphoid neoplasma

A

Neoplasm of B-cell, T-cell and NK-cell origin

Phenotype may resemble a particular stage of normal maturation – useful for diagnosis and classification

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
53
Q

Myeloid neoplasma

A

Neoplasm of early hematopoietic progenitors

Tend to evolve over time to more aggressive forms of disease – clinically important

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
54
Q

Acute myeloid leukemia

A

Immature progenitor cells (blasts) accumulate in the bone marrow suppressing normal hematopoiesis
Both myelodysplastic syndromes and myeloproliferative disorders can “transform” to this disease

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
55
Q

Myelodysplastic syndromes

A

Ineffective hematopoiesis due to defective maturation of myeloid progenitors
Resultant peripheral blood cytopenias
more severe than myeloproliferative disorders

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
56
Q

Chronic. Myeloproliferative disease

A

hronic Myeloproliferative Disease
Increased production of one or more terminally differentiated myeloid elements leads to elevated peripheral blood counts
Polycythemia Vera, Essential Thrombocytopenia, Primary Myelofibrosis, and CML

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
57
Q

Histiocytosis

A

Uncommon proliferative lesions of macrophage and dendritic cells
Langerhans cell (special type of immature dendritic cell) gives rise to a spectrum of neoplastic disorders referred to as the Langerhans cell histiocytoses
look for cutaneous involvement; all are CD4 +ve

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
58
Q

Factors influencing WBC neoplasia

A

Chromosomal translocations and other acquired mutations
Inherited genetic factors
acute leukemias: dominant negative oncogenic mutations involving transcription factors are often present that interfere with early stages of lymphoid or myeloid cell differentiation
Viruses
Chronic inflammation (i.e. Helicobacter Pylori)
Iatrogenic factors (radiation and some chemotherapy)
Smoking
oncogenic mutations occur most frequently in germinal center B cells during attempted antibody diversification
class switching == intragenic recombination event in which the IgM heavy-chain constant gene segment is replaced with a different constant gene
somatic hypermutation == point mutations within Ig genes that may increase antibody affinity for antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
59
Q

Translocations in WBC neoplasma

A

Translocations in WBC neoplasms

Most common nonrandom chromosomal abnormality seen in WBC neoplasms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
60
Q

WBC mutations

A

Play crucial roles in the development, growth or survival of the normal counterpart to the malignant cell (gain or loss of function)
Oncoproteins can block normal maturation, activate pro-growth signals or inhibit apoptosis
Protooncogenes may be activated during gene rearrangement and diversification

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
61
Q

Activation induced cytosine deaminase and its role in neoplasia

A

This enzyme is upregulated after Antigen stimulation in germinal center B cells
Allows Ig class switching and somatic hypermutation to increased antibody affinity
May induce MYC/Ig translocations – AID creates lesions in DNA that lead to chromosomal breaks
Can activate protooncogenes (i.e. BCL6

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
62
Q

V(D)J recombinant and its role in neoplasia

A

This enzyme cuts DNA at specific sites within the Ig and T-cell receptor loci
May go awry, leading to the joining of portions of other genes to Ag receptor gene regulatory elements
Unique to B and T cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
63
Q

Leukemia and lymphoma can be influenced by

A
Leukemia &amp; Lymphoma can be influenced by:
Inherited genetic factors: 
Down syndrome (trisomy 21) 
associated with an increased risk of Acute Lymphoblastic Leukemia (ALL) in children < 5 years 
Bloom syndrome
Fanconi anemia
ataxia telangiectasia
Type I neurofibromatosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

HTLV1

A

HTLV1: adult T-cell leukemia/lymphoma (ATLL

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
65
Q

EBV

A

EBV: Burkitt lymphoma, Hodgkin lymphoma, other B-cell Lymphoma
EBV infects B cells in the cortex; response to EBV infection is a paracortical hyperplasia of T-cells

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
66
Q

KSHV/HHV8

A

KSHV/HHV8: B-cell Lymphoma that presents as a malignant effusion in the pleural cavity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
67
Q

HIV

A

HIV: B-cell Lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
68
Q

Chronic inflammation leading to WBC neoplasia

A

Helicobacter pylori: Gastric B-cell Lymphomas
Gluten sensitive enteropathy: intestinal T-cell Lymphomas
Breast implants: T-cell Lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
69
Q

Iatrogenic factors leading to blood neoplasia

A

Iatrogenic factors leading to blood neoplasia

Radiation and chemo can affect the myeloid and lymphoid precursors to cause neoplasia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
70
Q

Smoking on blood neoplasia

A

Acute Myeloid Leukemia increased, especially due to benzene

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
71
Q

Lymphocytic leukemia

A

(Lymphocytic) Leukemia: neoplasms that have wide spread involvement of the bone marrow and often (not always) the peripheral blood
signs and symptoms related to the suppression of normal hematopoiesis by tumor cells in the bone marrow
“acute” leukemia == most common leukemia of childhood (ALL)
infections –> thrombocytosis; leukemia –> thrombocytopenia
generally: acute means kids and are -blasts; chronic means adults and are -cytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
72
Q

Lymphoma

A

Lymphoma: proliferations of lymphoid cells that arise as discrete tissue masses – involvement of the tissue makes it an “-oma” (“-oma” means mass)
2/3 of non-Hodgkin’s lymphomas and virtually all Hodgkin’s lymphomas present as enlarged, nontender lymph nodes; 1/3 of remaining non-Hodgkin’s lymphomas present with symptoms related to tissue/organ involvement

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
73
Q

Leukemia vs lymphoma

A

Leukemia vs. Lymphoma
Lymphomas more commonly exhibit tissue involvement (“-oma” means mass)
leukemia and lymphoma reflect the usual tissue distribution of each disease at presentation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
74
Q

Immature B cells

A

Precursor B cell (-blasts) neoplasms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
75
Q

Mature B cells

A

Peripheral B cells (-cystes) neoplasma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

Immature T cells

A

Precursor T cell neoplasma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
77
Q

Mature T cells and NK cells

A

Peripheral T cell neoplasms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
78
Q

Mature T cells and NK cells

A

Peripheral T cell and NK cell neoplasma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
79
Q

Reed sternberg cells and variants

A

Hodgkin lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
80
Q

Lymphoid neoplasms

A

Histological examination is required for diagnosis
Antigen receptor gene rearrangement precedes transformation of cells – all daughter cells derived from the malignant progenitor share the same antigen receptor gene configuration and sequence
Monoclonal: all daughter cells express the same configuration and sequence, synthesizing identical proteins (immunoglobulins or TCRs) – monoclonal means cancer, malignant
Polyclonal: means that they are immature and are reactive
Most resemble a recognizable stage of B or T cell differentiation (most are B cell neoplasms)
even though 80% of circulating lymphocytes are T-cells
Often associated with immune problems
Behavior resembles normal B and T-cells
most lymphoid tumors are widely disseminated at the time of diagnosis
Hodgkin lymphoma: sometimes restricted to one group of lymph nodes
marginal zone B-cell lymphomas: often restricted to sites of chronic inflammation
MALTomas that arise in the setting of chronic inflammation
t(11;18), t(14;18) or t(1;14)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
81
Q

HOdgkin

A

Hodgkin’s: spreads in an orderly fashion and staging is useful
distinctive pathologic features and is treated in a unique fashion

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
82
Q

Non Hodgkin

A

Non-Hodgkin’s: spreads widely, and less predictably

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
83
Q

Acute lymphoblastic

A

Neoplasm of immature lymphoblasts (most are B-ALL)
There is a lot of clinical overlap between B and T-ALL
Kids with a very low platelet count have ALL until proven otherwise – most common leukemia of childhood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
84
Q

Population ALL

A

Increased risk in Hispanics»whites > African Americans
Slight male predominance
Most common cancer of children (usually patients <15)
*Most common cancer of children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
85
Q

Pathogenesis ALL

A

Dysregulation of expression and function of transcription factors for normal B and T cell development
Disturb differentiation and promote arrest of maturation, inducing self-renewal
Mutations that drive cell growth (i.e. TK) are common
70% of T-ALLs have gain-of-function mutations in NOTCH1
majority of B-ALLs have loss-of-function mutations in PAX5, E2A, and RBF or a balanced t(12;21) involving the genes ETV6 and RUNX1
Numerical or structural chromosomal change: 90% (Translocations, altered ploidy)
Caused by relatively few mutations as compared to solid tumors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
86
Q

ALL morphology

A

Leukemic presentation: marrow is hypercellular and packed with lymphoblasts that replace the normal marrow elements
Scant basophilic cytoplasm with nuclei larger than small lymphocytes
Delicate, finely stippled nucleoli with a rim of condensed chromatin
Subdivided nuclear membrane (convoluted)
High mitotic rate
“Starry sky” due to macrophage ingestion of tumor cells
A bone marrow biopsy is performed and on microscopic examination shows nearly 100% cellularity with replacement by primitive cells that have large nuclei with delicate chromatin and indistinct nucleoli with scanty cytoplasm. These cells mark for CD10 (CALLA) antigen

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
87
Q

Lymphoblasts (different features than myeloblasts

A

must differentiate ALL from AML because they differ in their response to chemotherapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
88
Q

Acute lymphoblastic leukemia/lymphoma

A

More condensed chromatin

Less conspicuous nuclei

Smaller amounts of cytoplasm whihc lacks granules

MPO(-), PAS (+), TdT (+)

Abrupt; stormy onset”, within days to few weeks of first symptoms

Related to depression of marrow function; fatigue due to anemia; fever due to infection from neutropenia; bleeding due to thrombocytopenia;

More common in all; bone pain

More common in ALL ; HA, vomiting, nerve palsies from meningeal spread

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
89
Q

Acute myeloid leukemia

A

MPO (+)

No nerve palsies

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
90
Q

B and T cell antibody stain are required for definitive diagnosis of ALL or AML

A

Clinical
most common translocation == t(4;11) – ??? (Hubbard)
Normal hematopoiesis is suppressed due to physical crowding, growth factor competition, etc.
Abrupt, “stormy” onset in days to weeks of first signs and symptoms
Signs and symptoms related to depression of marrow function
Fatigue (anemia), fever (neutropenia → infection), bleeding (thrombocytopenia)
Mass effects due to neoplastic infiltration
Bone pain from marrow expansion and infiltration, LAD, HSM, testicular enlargement
neurological manifestations due to meningeal spread: headache, vomiting, nerve palsies; diplopia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
91
Q

With aggressive chemotherapy, 95% of children with ALL achieve complete remission, and 75-85% are cured

A

still leading cause of cancer deaths in children (most common cancer)
only 35-40% of adults are cured due to different molecular pathogenesis and inability to tolerate the necessary chemo regimen effective in children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
92
Q

Poor prognosis

A

Age < 2 (strong association of infantile ALL with translocation of MLL gene)
t(9:22) Philadelphia chromosome
Presentation in adolescence or adulthood
Peripheral blood blast count > 100,000 (high tumor burden likely)
Detection of residual disease after therapy

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
93
Q

Favorable prognosis

A
Age between 2 and 10 years old
Low white cell count
Hyper-diploidy
trisomy of chromosome 4, 7, 10 
Presence of t(12:21) -- involves ETV6 and RUNX1 genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

B cell ALL

A

Acute childhood leukemia
Peak incidence at age 3
Extensive marrow and peripheral blood involvement
B-cell tumors present with cyclic tumors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

Morphology B cell ALL

A

Marrow is hypercellular and packed with lymphoblasts

Pancytopenia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
96
Q

Genetics B cell ALL

A

loss-of-function mutations in PAX5, E2A, EBF or *t(12:21) ETV6/RUNX1
Hyperdiploidy and hypodiploidy may be present and are exclusive in B-ALL

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
97
Q

Immunophenotype B cell all

A
TdT (+) (pre-B-cell marker)
CD10, 19, 20 (+)
Very immature neoplasm: CD10 (-)
Mature neoplasm: IgM heavy chain (+) (μ chain)
PAX5 (+)
t(9:22) BCR/ABL protein
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

Treatment B cell all

A

Effectively treatment in children with chemotherapy

Prophylax scrotum and CSF

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
99
Q

T cell all

A

Presents in adolescent male (teenagers) as thymic mass (mediastinal) “lymphomas”; and T-cells (thymocytes) – mnemonic == 3 T’s
May evolve to a leukemia
lymphadenopathy and splenomegaly
Signs and symptoms may include complications related to compression of vessels or airways in the mediastinum

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
100
Q

Genetics T cell all

A

NOTCH1 gain-of-function mutations – T-cells are NOTCH1 GOF

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
101
Q

Immunophenotype T cell ALL

A

TdT (+) – precursor lymphocyte (i.e. lymphoblast) markers
CD1 and CD8 positive
Immature: CD3, CD4, and CD8 negative

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
102
Q

Chronic lymphocytic leukemia (CLL) & small lymphocytic lymphoma

A

only difference between CLL and SLL is the degree of peripheral blood lymphocytosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
103
Q

Follicular lymphoma

A

lar lymphoma == efface the normal architecture of the lymph nodes
follicular hyperplasia does not
t(14;18

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
104
Q

DLBCL

A

BCL6

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

Burkitt lymphoam

A

T(8;14)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
106
Q

Mantle cell lymphoma

A

T(11;14)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
107
Q

Marginal zone lymphoma MALToma

A

t(11;18), t(14;18), or t(1;14

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
108
Q

Hairy cell leukemia

A

TRAP, BRAF

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
109
Q

Chronic lymphocytic leukemia

A

Most common leukemia of adults in the Western world
*Morphologically, phenotypically and genotypically indistinguishable from SLL, differ only in the degree of peripheral blood lymphocytosis (absolute WBC count > 5000/mm3

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
110
Q

Populations at risk CLL

A

Male 2:1
Age: 60yrs
More common in the West than Asian countries

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
111
Q

Chromosomal anomalies CLL

A

Chromosomal translocations are rare
Deletions: 13q14.3(microRNAs 15a & 16-1), 11q, 17p
Trisomy 12q – “tr-elve,” and the mnemonic becomes “11, 12/trelve, 13, 14, 15, 16, and 17

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
112
Q

Genetics CLL

A

Somatic hypermutation of immunoglobulin genes can occur indicating cell of origin may be post-germinal center memory B cell or naive B cell
Unmutated Ig segments = increased aggressive course
NOTCH1 receptor gain of function mutations – this is a poor prognostic factor
RNA splicing mutations

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
113
Q

Tell growth CLL

A

Confined to proliferation centers
NFκB is stimulated by stromal cells = promotion of growth and survival
BCRs produce signals required for growth and survival via BTK (Bruton TK)
*BTK inhibitors have produced sustained response in many patients

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
114
Q

Morphology CLL

A

Proliferation centers (pathognomonic): Larger lymphocytes gathered in loose aggregates that contain mitotically active cells
Smudge cells: Small round lymphocytes in the peripheral blood with scant cytoplasm, disrupted when making the smear
smudge cells are not characteristic of anything (non-specific)
lymph node architecture is effaced by small lymphocytes (6-12um) with round or slightly irregular nuclei, condensed chromatin and scant cytoplasm
Infiltrates seen in bone marrow, splenic pulp (white + red) and hepatic portal tracts

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
115
Q

Immunophenotype CLL

A

CD 5, 19, 20, 23 (+)
B cell proliferation that is positive for CD5, think CLL
Low levels of surface Ig (IgM +/ IgD) are also seen
When lymph node involvement occurs this becomes known as Small Lymphocytic Lymphoma
only difference between CLL and SLL is the degree of peripheral blood lymphocytosis
Prolymphocytes are seen in lymph nodes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
116
Q

Clincial CLL

A

patients are often asymptomatic at diagnosis
Signs and symptoms appear as nonspecific: easy fatigability, weight loss, anorexia, generalized lymphadenopathy, hepatosplenomegaly
Variable leukocyte count
Leukopenia in SLL with marrow involvement
Leukocytosis in CLL with heavy tumor burden
Monoclonal Ig “spike” may be present in the blood – more indicative of multiple myeloma?
patients with monoclonal B cells < # than required for diagnosis can have similar genetic defects but only 1% progress to symptomatic disease
Median survival 4-6 years, 10 years in patients with decreased tumor burden at diagnosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
117
Q

CLL effect on immune function

A

Hypogammaglobulinemia = bacterial infection

Neoplastic B cell auto-antibodies = hemolytic anemia or thrombocytopenia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
118
Q

Poor prognosis cll

A

11q, 17p deletions
Lack of somatic hypermutation
ZAP70 (+): augments immunoglobulin receptor signaling
NOTCH1 mutations
Richter syndrome: tendency of CLL/SLL to transform to a more aggressive tumor in the form of diffuse large B-cell lymphoma; transformation of CLL/SLL to an aggressive B-cell lymphoma
Rapidly enlarging mass within a lymph node or the spleen, likely due to additional acquired mutations
large cell transformation is BAD; survival < 1 yea

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
119
Q

Treatment CLL symptomatic

A

Gentle chemotherapy
Immunotherapy with antibodies to surface proteins (CD20)
hematopoietic stem cell transplant is offered to young patients
BTK inhibitors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
120
Q

Follicular lymphoma

A

Tumor of lymph nodes that causes obliteration of the lymph node and loss of architecture
lymphoma (neoplastic) will efface lymph node architecture; hyperplasia (reactive) won’t
Most common form of indolent non-Hodgkin’s lymphoma in the US

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
121
Q

Who gets follicular lymphoma

A

Less common in Europe

Rare in Asian population

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
122
Q

Pathogenesis follicular lymphoma

A

Arises from germinal center cells
strongly associated with chromosomal translocations involving BCL2
t(14:18) BCL2/Ig heavy chain – seen in >90% of follicular lymphomas (characteristic translocation of follicular lymphoma)
IGH locus on chromosome 14
BCL2 locus on chromosome 18
BCL2 antagonizes apoptosis (promotes cell survival)
Overexpression of BCL2 inhibits apoptosis of B cells that fail somatic hypermutation
Devoid of apoptotic cells because BCL2 is anti-apoptotic
MLL2: histone methyltransferase (epigenetics!); mutated in >90% of follicular lymphomas
Neoplastic cells grown in lymph nodes in a network of reactive follicular dendritic cells, macrophages and T cells and their microenvironment determines treatment response

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
123
Q

Morphology follicular lymphoma

A

Follicular (nodular) and diffuse proliferation of centrocytes (predominant) and centroblasts
Lymphocytosis in 10%, makes it more aggressive
Para-trabecular lymphoid aggregates in bone marrow (also seen in splenic white pulp and hepatic portal triads

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
124
Q

Centrocytes follicular

A

Small cells with irregular or cleaved nuclear contours

Scant cytoplasm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
125
Q

Centroblasts cll

A

Large cells with open nuclear chromatin
Several nucleoli
Modest amounts of cytoplasm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
126
Q

Immunophenotype

A
CD 10, 19, 20 (+)
CD5 (-)
BCL6 (+) -- anti-apoptotic
BCL2 (+) normal follicular cells are normally devoid of this marker
Surface Ig (+)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

Clinical follicular

A

Presents in middle age as painless, generalized lymphadenopathy
Extra-nodal site involvement is uncommon
Incurable with waxing-waning course; survival is 7-9 years not improved by aggressive therapy
palliate the patient with low dose chemotherapy or immunotherapy (anti-CD20 antibody)
Transformation in 30-50% to diffuse large B-cell Lymphoma (usually) or Burkitt Lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
128
Q

Diffuse large B cell lymphoma BAD: patient at risk

A

Most common form of non-Hodgkin’s lymphoma

Median age: 60 years old (may also occur in young adults and children

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
129
Q

Pathogenesis DLBCL

A

3q27 mutation (translocation or acquired) involving BCL6
BCL6 is required for formation of normal germinal centers – dysregulation of BCL6 == DLBCL
Overexpression of BCL6 inhibits germinal center B-cell differentiation, growth arrest and apoptosis
Other potential mutations
t(14;18) involving BCL2 (anti-apoptotic)
tumors with BCL2 rearrangements usually lack BCL6 rearrangements
this translocation is characteristic/pathognomonic for follicular lymphoma
MYC mutation
myc dysregulation == Burkitt Lymphoma
Histone acetyltransferase mutations (p300, CREBP)
large, destructive masses; extranodal involvement
this is a mature B cell neoplasm

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
130
Q

Morphology DLBCL

A

Large cells (4-5X > normal lymphocytes)
Diffuse pattern of growth
Substantial morphologic variation
Commonly has a round-oval nucleus (vesicular in appearance)
Multiple (2-3) nucleoli (or single and centrally placed)
Cytoplasm: moderately abundant and pale or basophilic
More anaplastic tumors may contain multinucleated cells with inclusion-like nucleoli

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

DLBCL immunophenotype

A

CD 19, 20 (+)
Variable expression of CD10 and BCL6
Most have surface immunoglobulin

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
132
Q

DLBCL location

A

Rapidly enlarging mass at nodal or extranodal site
May arise anywhere in the body
commonly Waldeyer ring (oropharyngeal lymphoid tissue including the tonsils and adenoids)
DLBCL == Waldeyer ring
Liver and spleen involvement leads to destructive masses
Extranodal is common: GI, skin, bone, brain
Bone marrow involvement is rare (late course

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
133
Q

DLBCL clincial

A

aggressive tumors that are rapidly fatal without treatment
Treated: 60-80% complete remission, 40-50% cured
Adjuvant treatment with anti-CD20 antibody –> improved initial response and improved outcomes
Limited disease has better outcome than widespread disease or those with bulky tumor masses
Inhibition of NFκB and B cell receptor pathways is beneficial
MYC translocations have worse outcome and may be best treated similar to Burkitt lymphoma
myc dyregulation == Burkitt Lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
134
Q

Immunodeficiency associated large B cell lymphoma

A

Occurs with severe T-cell immunodeficiency (HIV or allogenic bone marrow transplant)
EBV infection of neoplastic B cells
May regress with restoration of T cell immunity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
135
Q

Primary effusion lymphoma DLBCL

A

Malignant pleural or ascitic effusion
Most common in patients with advanced HIV or who are elderly
Tumor cells infected with KSHV/HHV8 which appears to have a causal role – always associated with Kaposi’s
Anaplastic tumor cells that do not express B or T cell markers
Possess clonal immunoglobulin heavy chain gene rearrangements

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
136
Q

Burkitt lymphoma

A

Burkitt Lymphoma == “Burkitt is Burk-EIGHT” [t(8;14)]
Definition
Most commonly affect kids and young adults
Highly associated with MYC translocations on chromosome 8 that leads to high MYC protein levels
MYC increases genes for aerobic glycolysis (Warburg effect) allowing cells to biosynthesize building blocks for growth and cell division, if glucose and glutamine are available – this is important
Burkitt Lymphoma == dysregulation of c-MYC
Three variants: African (endemic), sporadic (nonendemic), HIV associated
histologically identical, but different in some clinical, genotypic, and virologic characteristics
not ALL Burkitt lymphomas are EBV positive; All African (endemic) Burkitt lymphomas are though
aka Diffuse Small Non-Cleaved Lymphoma (Hubbard)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
137
Q

Pathogenesis burkitt

A

t(8:14) MYC/Ig heavy chain
May also be t(2:8) or t(8:22) for the light chains
chromosome 2 == kappa
chromosome 22 == lambda
chromosome 8 == MYC
chromosome 14 == IgH
Induced by AID in germinal center B cells
MYC overexpression, increased activity
Fastest growing human tumor – doubles in size every 36 hours
Infection with EBV precedes transformation

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
138
Q

Morphology burkitt

A

Diffuse infiltrate of intermediate sized lymphoid cells
Round/oval nuclei, coarse chromatin, several nucleoli, moderate amount of cytoplasm
High mitotic index
Numerous apoptotic B cells – tingible body macrophage
“Starry sky” pattern due to phagocytosis of nuclear remnants by macrophages
If bone marrow is involved, tumor cells exhibit clumped nuclear chromatin, 2-5 nucleoli and royal blue cytoplasm with clear cytoplasmic vacuoles

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
139
Q

Immunophenotype burkitt

A
CD 10, 19, 20 (+)
BCL6 (+)
Surface IgM (+)
BCL2 (-)
MYC (+) -- Burkitt Lymphoma is dysregulation of c-MYC
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

Burkitt clincial

A

30% of childhood non-Hodgkin’s lymphoma in the USA
Aggressive lymphoma
Responds well to chemotherapy
Children and young adults are usually cured
Older adults have more guarded outcome

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
141
Q

Burkitt sporadic

A

Burkitt Lymphoma: Sporadic (nonendemic)
t(8:14)
Breakpoint on IgH locus is in the class switch region
15-20% of patients are latently infected with EBV
Mass of ileocecum and peritoneum – just GI

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
142
Q

Burkitt african endemic

A

Burkitt Lymphoma: African (endemic)
t(8:14)
Breakpoint on IgH locus is in the 5’ V(D)J sequence
All patients are latently infected with EBV
Mass of mandible/face and/or abdominal viscera (kidneys, ovaries, adrenals)
Bone marrow or peripheral blood involvement is rare

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
143
Q

Burkitt lymphoma HIV

A

t(8;14)
25% of patients are latently infected with EBV

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
144
Q

Plasma cell neoplasma(dyscrasias)

A

Most common plasma cell dyscrasia == Monoclonal Gammopathy of Uncertain Significance (MGUS)
most common plasma cell malignancy == multiple myeloma
Cells secrete monoclonal Ig (serve as tumor markers)
Will commonly see plasma cell clusters, they don’t normally present in clusters
M component: a monoclonal Ig in the blood
Restricted to plasma due to high molecular weight
not seen in urine unless glomerular damage has occurred
neoplastic plasma cells often synthesize excess light chains along with complete immunoglobulins
light chains are small in size and excreted in the urine (Bence-Jones proteins)
One light chain is increased others are low
15% of deaths due to lymphoid neoplasms

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
145
Q

Monoclonal gammopathy

A

Monocolonal gammopathy
Heavy chain disease == rare monoclonal gammopathy that is seen in association with a diverse group of disorders including lymphoplasmacytic lymphoma and an unusual small bowel marginal zone lymphoma that occurs in malnourished populations (so-called Mediterranean lymphoma)
common feature == synthesis and secretion of free heavy chain fragments

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
146
Q

Multiple myeloma

A

Multiple Myeloma (plasma cell myeloma) == the most common plasma cell neoplasm
Definition
causes bony destruction of the skeleton and often presents with pain due to pathologic fractures
Plasma cell neoplasm that commonly causes tumorous masses throughout the skeletal system
“moth eaten”
lytic bone lesions, hypercalcemia, renal failure, and acquired immune problems

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
147
Q

Multiple myeloma patients

A

Male
African descent
Peak incidence: 65-70 years old – disease of older adults

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
148
Q

Multiple myeloma variants

A

Solitary myeloma: single mass in bone or soft tissue
Smoldering myeloma: lack of signs and symptoms with high plasma M component
does not exhibit lytic bone lesions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
149
Q

Multiple myeloma pathogenesis

A
athogenesis
Frequent IgH (heavy chain) rearrangements
Translocated with cyclin D1 and D3: good prognosis
Poorer prognosis
Deletion of chromosome 13q
Deletion of chromosome 17p (TP53)
t(4;14)
MYC acquisition
NFκB promoting B-cell survival
high levels of IL-6
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

Growth and survival multiple myeloma

A

Growth and survival
IL-6 is important for growth of plasma cells, produced by neoplastic plasma cells and resident marrow stromal cells (higher levels = poor prognosis)
Bone destruction is mediated by factors produced by neoplastic cells
MIP1α upregulates NFκB ligand (RANKL) activating osteoclasts
Osteoblasts are inhibited via Wnt pathway
increased bone resorption → hypercalcemia and pathologic fractures

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
151
Q

Morphology multiple myeloma

A

Destructive plasma cell tumors (plasmacytomas) involving the axial skeleton
Vertebral column, ribs, skull, pelvis, femur, clavicle, scapula
Appear as 1-4cm punched-out defects radiographically
Soft, gelatinous, red tumor masses
Can also present as widespread myelomatous bone disease that causes diffuse demineralization (osteopenia) instead of focal defects (i.e. pathologic fractures

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

Morphology neoplastic plasma cells

A

Increased number of plasma cells in the marrow with perinuclear clearing due to large Golgi apparatus and odd placed nucleus
Rouleaux formation: high level of M proteins causes RBCs in blood smears to stick together linearly
characteristic/indicative but not specific – seen whenever immunoglobulin levels are elevated
Flame cells: intracellular accumulation of degraded protein with fiery red cytoplasm
Mott cells: multiple grapelike cytoplasmic droplets
Cells containing inclusions of fibrils, crystalline rods and globules
Globular inclusions: Russell bodies (cytoplasmic) or Dutcher bodies (nuclear)
Dutch == nuclear families

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
153
Q

Immunophenotype multiple myeloma

A

CD138 (+) (aka syndecan-1)

May be CD56 (+)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
154
Q

Signs ANS symptoms multiple myeloma

A

Effects of plasma cell growth in tissues (bones)
Production of excess immunoglobulin
Suppression of normal humoral immunity

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
155
Q

Clincial sequelae multiple myeloma

A

Pathologic fracture and chronic pain due to resorption
Hypercalcemia –> Neurologic signs and symptoms (confusion, weakness, lethargy)
stones, bones, groans, and psychiatric moans + shortened QT
Renal failure (second biggest cause of death after infections)
myeloma kidney
Acquired immune abnormalities
Recurrent bacterial infection (most common cause of death)
Cellular immunity is unaffected

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
156
Q

Bence jones proteinuria

A

Excreted light chains are toxic to renal tubular epithelial cells –> myeloma kidney –> renal failure/insufficiency (second most common cause of death in multiple myeloma, after infection)
Some (λ3, λ6) may be prone to amyloidosis of the AL type
primary or immunocyte-associated amyloidosis results from a monoclonal proliferation of plasma cells secreting light chains (usually of the λ isotype) that are deposited as amyloid
Increased immunoglobulins (especially IgG) and/or light chains in the plasma & urine
most multiple myelomas are IgG with kappa light chains - ???
Monoclonal immunoglobulins M proteins: IgG (55%), IgA (25%)
Excessive production and aggregation of M proteins, especially IgA or IgG3 –> hyperviscosity
Waldenström macroglobulinemia == a syndrome in which high levels of IgM lead to symptoms related to hyperviscosity of the blood; occurs in older adults; associated with lymphoplasmacytic lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
157
Q

IgA or IgG3->hyperviscosity

A

Waldenström macroglobulinemia == a syndrome in which high levels of IgM lead to symptoms related to hyperviscosity of the blood; occurs in older adults; associated with lymphoplasmacytic lymphoma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
158
Q

Marrow involvement

A

Diagnosis requires bone marrow examination

Marrow involvement often gives rise to a normocytic, normochromic anemia +/- moderate leukopenia and thrombocytopenia

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
159
Q

Treatment

A

survival is 4-7 years; no cure
Proteasome inhibitors: Degrade unwanted proteins (cells are prone to accumulation of misfolded, unpaired immunoglobulin chains). Induce cell death and retard bone resorption (through stromal cells)
also thalidomide
Bisphosphonates inhibit bone resorption; reduce the amount of pathologic fractures but can exacerbate renal complications of disease; ensure patient is adequately hydrated; follow creatinine
HSC transplant prolongs life but is not curative

160
Q

Solitary myeloma

A

Solitary lesion of bone or soft tissue
Bone lesion in same locations as multiple myeloma → multiple myeloma in 10-20 years even if it is resected
solitary osseous plasmacytoma almost inevitably progresses to multiple myeloma
extraosseous lesions in lungs, oronasopharynx or nasal sinuses → cured by local resection
Modest increase in M proteins in blood or urine

161
Q

Smoldering myeloma

A

Middle ground between multiple myeloma and Monoclonal Gammopathy of Uncertain Significance (MGUS)
Plasma cells are 10-30% of marrow cellularity
Elevated serum M protein > 3gm/dl (same level as in multiple myeloma, but no signs and symptoms)
patients are asymptomatic
75% progress (unpredictably) to multiple myeloma in 15 years

162
Q

Monoclonal gammopathy of uncertain significance

A

Most common plasma cell disorder/dyscrasia
most common plasma cell neoplasm/malignancy == multiple myeloma
3% of people > 50
5% of people > 70
Asymptomatic patients with small to moderately large M components in their blood (< 3g/dl)
low but constant rate of transformation to symptomatic monoclonal gammopathies
most often multiple myeloma
1% of patients progress to symptomatic plasma cell neoplasm every year
May be a precursor to multiple myeloma as many of the same chromosomal aberrations are present (unpredictable)
monitor Bence-Jones Proteinuria and serum levels of M component to trend and predict transformation

163
Q

Multiple myeloma

A

Elevated m component

Symptoms

164
Q

Smoldering myeloma

A

Elevated m component

No sym[toms

165
Q

MGUS

A

No m component

No symptoms

166
Q

Lymphoblastic lymphoma

A

Definition
B-cell neoplasm of older adults (6-7th decade)
Substantial fraction of the tumor cells undergo terminal differentiation to plasma cells
how it differs from CLL/SLL
Can cause hyperviscosity (Waldenström macroglobulinemia)
Waldenström macroglobulinemia == a syndrome in which high levels of IgM lead to symptoms related to hyperviscosity of the blood; occurs in older adults; associated with lymphoplasmacytic lymphoma
Rarely does damage from secretion of light chains or bone destruction occur

167
Q

Pathogenesis lymphoplasmacytic lymphoma

A

Acquired MYD88 mutation
Normally codes a protein that activates NFκB and augments signals downstream of the B-cell receptor (Ig) complex (aka it promotes the growth and survival of tumor cells

168
Q

Morphology lymphoplasmacytic lymphoma

A

Marrow infiltrate of lymphocytes, plasma cells and plasmacytoid lymphocytes
Mast cell hyperplasia
Russell bodies: PAS (+) inclusions with Ig in the cytoplasm
Dutcher bodies: PAS (+) inclusions with Ig in the nucleus
Dutch == nuclear families
Dutcher and Russell bodies only seen in plasma cell dyscrasias
At diagnosis: tumor spread to lymph nodes, spleen and liver
Nerve Roots, meninges and brain may be affected with progression

169
Q

Immunophenotype lymphoplasmacytic lymphoma

A

CD20 (+)
Surface Ig (+)
Plasma cells will secrete the same Ig that is expressed on the surface of the lymphoid cells
Usually IgM (may be IgG or IgA)

170
Q

Clinical lymphoplasmacytic lymphoma

A

Nonspecific signs and symptoms: weakness, fatigue, weight loss
lymphadenopathy and hepatosplenomegaly in 50%
10% have autoimmune hemolysis due to cold agglutinins (IgM binding RBCs at T < 37°C)
No bone lesions

171
Q

Waldenstrom macroglobunemia

A

Waldenström macroglobulinemia == a syndrome in which high levels of IgM lead to symptoms related to hyperviscosity of the blood; occurs in older adults; associated with lymphoplasmacytic lymphoma
IgM secreting tumors cause a hyperviscosity syndrome
retinopathy
neuro symptoms: HA to seizures and coma
spontaneous bleeding
Plasmapheresis alleviates signs and symptoms of increased IgM
Visual impairment due to venous congestion
Retinal vein tortuosity and distention +/- retinal hemorrhage and exudates
Neurological problems due to sluggish blood flow: headache, dizziness, deafness, and stupor
Bleeding due to macroglobulin-clotting factor complex formation and effects on platelet function
Cryoglobulinemia due to precipitation of macroglobulins at low Temperature == Raynaud phenomena and cold urticaria
cold agglutinin antibodies (IgM

172
Q

Treatment

A

Incurable, progressive disease that rarely progresses to large cell lymphoma
Low dose chemotherapy and immunotherapy (antiCD20) may control tumor growth for a period of time
Median survival of 4 years

173
Q

Mantle cell lymphoma

A

Uncommon non-Hodgkin’s lymphoma

tumor cells closely resemble the normal mantle zone B cells that surround germinal centers

174
Q

Patients at risk mantle cell lymphoma

A

Presents in 5-6th decade

male predominance

175
Q

Pathogenesis MCL

A

t(11:14) cyclin D1/IgH –> overexpression of cyclin D1 promotes G1 to S phase progression
IgH genes lack somatic hypermutation (== naïve B cell origin

176
Q

MCL tumor location

A

Generalized painless lymphadenopathy, +/- peripheral blood involvement
Extranodal sites: bone marrow, spleen, liver, gut
Lymphomatoid polyposis: if mucosal involvement of the small bowel or colon → polyps (can be misdiagnosed as ulcerative colitis)
mantle cell lymphoma frequently presents in the GIT

177
Q

Morphology MCL

A

Nodular appearance or diffusely efface the lymph node
Homogenous population of small lymphocytes with irregular/clefted (cleaved) nuclei
Condensed nuclear chromatin, inconspicuous nucleoli and scant cytoplasm
No centroblasts or proliferation centers (distinguished from CLL/SLL)
Requires immunophenotyping to distinguish blastoid variants from ALL
Can look like follicular lymphoma
Follicular lymphoma don’t have a germinal center
Mantel cell lymphoma will have a germinal center

178
Q

MCL immunophenotype

A

Immunophenotype
Increased cyclin D1
CD 5, 19, 20 (+)
CD 23 (-) == distinguished from CLL/SLL
Elevated levels of surface Ig (usually IgM and IgD with κ or λ light chain)
IgH lacks somatic hypermutation (origin = naive B cell)

179
Q

MCL clincal

A

Painless lymphadenopathy
Spleen and gut involvement are common – mantle cell lymphoma frequently presents in the GIT
Poor prognosis
Median survival: 3-4 years
COD: organ dysfunction due to lack of effective chemo and tumor infiltration
Blastoid variant and “proliferative” expression –> decreased survival

180
Q

MCL treatment

A

Chemo is not helpful

HSC and proteasome inhibitors are now being used with potential

181
Q

Marginal zone lymphoma

A

Heterogenous group of B-cell tumors in lymph nodes, spleen, or extranodal tissue
Tumor cells show evidence of somatic hypermutation (== memory B-cell origin)
marginal zone lymphoma and hairy cell leukemia are memory B-cell origin
Often referred to as MALTomas

182
Q

Extranodal sites and 3 special characteristics

A

Arise in tissues affected by chronic inflammation (autoimmune or infectious)
i.e. Sjogren’s (salivary glands), Hashimotos (thyroid) and stomach (H. Pylori)
Remain localized for long periods of time, but may spread late in their course
May regress if the inciting agent (i.e. Helicobacter) is eliminated – cure cancer with antibiotics!
Lies on a spectrum of reactive lymphoid hyperplasia and full-blown lymphoma

183
Q

Pathogenesis marginal zone lymphoma

A

Initially: polyclonal immune reaction that acquires mutations → B-cell clone that depends on Ag stimulated Th cells (eventually becomes monoclonal)
Withdrawal at this stage = involution of tumor
Additional mutations acquired over time renders growth and survival to be Ag independent
t(11:18), t(14:18) or t(1:14) which leads to activation of NFκB and promotes cell growth/survival
t(11:18), t(14:18) or t(1:14) are specific for extranodal marginal zone lymphomas (MALTomas)
upregulate the expression and function of BCL10 or MALT1 –> NFκB –> growth and survival
Further evolution = metastasis and transformation to large B-cell lymphoma

184
Q

Hairy cell leukemia

A

Rare B-cell neoplasm that has hair-like projections coming out of the leukemic cells – hence, “hairy cell”
marginal zone lymphoma and hairy cell leukemia are memory B-cell origin
Patients at risk
Middle age (55), white males (males 5:1)
Pathogenesis
BRAF (serine/threonine kinase) activating mutation (valine → glutamate)
BRAF is positioned immediately downstream of RAS in the MAPK signaling cascade

185
Q

Morphology hairy cell leukemia

A

Leukemic cells have fine, hair-like projections
Round, oblong or reniform (kidney shaped) nuclei
Moderate amounts of pale blue cytoplasm with bleb-like extensions
“Dry tap” because tumor cells are trapped in the ECM
Only seen on marrow biopsy
Heavily infiltrated splenic red pulp = obliteration of white pulp with a beefy red gross appearance
Hepatic portal triads may be involved

186
Q

Hairy cell leukemia immunophenotype

A

CD 11c, 19, 20, 25, 103 (+)
annexin A1 (+)
Surface Ig (IgG) +ve
tartrate-resistant acid phosphatase (TRAP) +ve

187
Q

Clincial hairy cell leukemia

A

Massive splenomegaly (hepatomegaly and lymphadenopathy are rare)
many patients will present with splenomegaly (dragging sensation in the LUQ)
Pancytopenia (from marrow involvement and splenic sequestration (hypersplenism)
1/3 have infection (often atypical mycobacterial)
increased susceptibility to mycobacterium kansasii
Exceptionally sensitive to gentle chemotherapy = long lasting remission
Relapse in 5 years but respond well to re-treatment with the same agents
BRAF inhibitors = excellent tumor response in patients who fail chemotherapy
Excellent prognosis

188
Q

A 49-year-old man has had increasing fatigue for the past 4 months. On physical examination he has massive splenomegaly but no lymphadenopathy. Laboratory studies show a Hgb of 10.1 g/dL, Hct 30.3%, MCV 90 fL, WBC count 1600/microliter, and platelet count 48,000/microliter. Examination of his peripheral blood smear shows increased numbers of peripheral blood lymphocytes containing tartrate-resistant acid phosphatase (TRAP). Which of the following is the most likely diagnosis

A

Hairy cell leukemia is trap positive

189
Q

Peripheral T cell lymphoma, unspecified

A

Derived from mature T cells so it has T-cell markers

diagnosis of exclusion

190
Q

Immunophenotype peripheral T cell lymphoma

A

CD2+, CD3+, and CD5+
some may be CD4+ or CD8+
either αβ or γδ TCRs
requires immunophenotyping

191
Q

Morphology peripheral T cell lymphoma

A

Tumors efface lymph nodes diffusely
Pleomorphic mix of variably sized malignant T-cells
Prominent infiltrate of reactive cells (eosinophils, macrophages) attracted by tumor cytokines
Neoangiogenesis may be seen

192
Q

Clincial peripheral T cell lymphoma, unspecified

A

Present with general lymphadenopathy +/- pruritus, eosinophilia, fever, weight loss
Cure is rare, worse prognosis than comparably aggressive B cell neoplasms

193
Q

Anaplastic large cell lymphoma

A

T-cell lymphoma with good prognosis

194
Q

Pathogenesis ALK+ anaplastic large cell lymphoma

A

ALK rearrangement on chromosome 2p23 –> break the ALK locus and lead to formation of chimeric genes encoding ALK fusion proteins, constitutive active tyrosine kinases that trigger RAS and JAK/STAT pathways
ALK is not expressed in normal lymphocytes therefore the detection of ALK in tumor cells is a reliable indicator of an ALK gene rearrangement – presence of ALK is pathognomonic

195
Q

Immunophenotype anaplastic large cell lymphoma alk+

A

CD30 positive

CD8 cell is anaplastic

196
Q

Morphology anaplastic large cell lymphoma

A

Large anaplastic cells with horseshoe nuclei and voluminous cytoplasm (“hallmark cells”)
Tumor cells surround venules, infiltrating lymphoid sinuses (mimic metastatic carcinoma

197
Q

Clinical anaplastic large cell lymphoma alk+

A

More common in children and young adults
Involve soft tissue == very good prognosis
Chemotherapeutic cure = 75-80%
ALK (-) tumors in older adults have a worse prognosis
Recombinant anti CD30 antibodies have produced promising responses in patients

198
Q

Adult T cell leukemia/lymphoma atll

A

Neoplasm of CD4+ T-cells
patients are infected with HTLV-1
Common in Japan, West Africa and the Caribbean

199
Q

Pathogenesis adult T cell leukemia/lymphoma atll

A

Virus produces Tax protein that activates NFκB

200
Q

Morphology atll

A

Morphology

Cells have multi-lobulated nuclei (“cloverleaf” or “flower” cells) and contain clonal HTLV1 proteins

201
Q

Clincial atll

A

Skin lesions, general lymphadenopathy, hepatosplenomegaly, peripheral blood lymphocytosis, hypercalcemia
HTLV-1 can also cause a progressive demyelinating disease of the CNS and spinal cord

202
Q

Prognosis atll

A

Rapidly progressive disease, fatal in months to 1 year despite aggressive chemotherapy
If only skin is involved (rare), disease has a more indolent course

203
Q

My oasis fungoides

A

Tumor of CD4+ helper T-cells that homes to the skin
Cutaneous lesions progress through three phases: premycotic, plaque, and tumor
Epidermis and upper dermis are infiltrated by neoplastic T cells with a cerebriform appearance
due to nuclear membrane infolding
Late disease progresses extracutaneously, commonly to lymph nodes and bone marrow
Mycosis fungoides (with no relationship to fungi) is one form of cutaneous T-cell lymphoma. The abnormal CD4 cell proliferation is infiltrating the skin and producing erythroderma. If these neoplastic cells circulate, it is known as Sezary syndrome

204
Q

Sezary syndrome

A

Tumor of CD4+ helper T-cells that homes to the skin
Rarely progresses to tumefaction
Generalized exfoliative erythroderma
Associated leukemia of cells with characteristic cerebriform nuclei

205
Q

Mycosis fungoides and sezary syndrome==tumor of cd4 helper T cells that homes to the skin

A

CLA (+) *cutaneous leukocyte antigen
CCR4 and CCR10 (+) – chemokine receptors
Homes to skin
Tumor cells circulate through the blood, bone marrow, and lymph nodes
Indolent tumors
Median survival of 8-9 years
Occasionally transforms to aggressive T-cell lymphoma (terminal event)

206
Q

Large granular lymphocytic leukemia variants and immunophenotype

A

T-cells variant (indolent): CD3 (+)

NK cells variant (subtler, common, aggressive): CD56 (

207
Q

Morphology large granular lymphocytic leukemia

A

STAT3 mutation
Large lymphocytes with abundant blue cytoplasm and azurophilic granules
Sparse lymphocytic infiltrates in the marrow, spleen and liver

208
Q

Clincial large granular lymphocytic leukemia

A

Clinical
Neutropenia and anemia dominate the clinical picture (despite low marrow involvement)
neutropenia –> striking decrease in late myeloid forms in the marrow
Mild-moderate lymphocytosis and splenomegaly of adults
No lymphadenopathy or hepatomegaly
Increased incidence of rheumatoid disorders – autoimmunity provoked by the tumor seems likely
Variable course based on cytopenias and their response to low dose chemo or steroids

209
Q

Felty syndrome

A

Felty Syndrome == rheumatoid arthritis + Splenomegaly + Neutropenia
*associated with large granular lymphocytic leukemia as an underlying cause

210
Q

Extranodal nk/t cell lymphoma

A

Extranodal NK/T-cell Lymphoma (nee lethal midline granuloma disorder)
Definition
Destructive nasopharyngeal mass
extra-nodal == extra-nosal
May also be seen in the testis and skin
Surrounds and invades small vessels = ischemic necrosis
More common in Asia
Tumor originates from a single *EBV-infected cell

211
Q

Morphology extranodal nk/t cell lymphoma

A

Large azurophilic granules in tumor cell cytoplasm

small to large pyknotic cells cuffing and invading small vessels with ischemic necrosis

212
Q

Immunophenotype extranodal nk/t cell

A
No CD21 (B-cell EBV receptor)
implication is that EBV has other receptors associated with it that are not CD21
No CD3
No TCR rearrangements
Express NK cell markers
213
Q

Clincal extranodal nk/t cell lymphoma

A

Highly aggressive; respond well to radiation; resistant to chemotherapy
Poor prognosis in patients with advanced disease

214
Q

Hodgkin lymphoma

A

Often localized to a single, axial group of lymph nodes (cervical, mediastinal, para-aortic, axillary, groin)
Hodgkin Lymphoma arises in a single node or chain of nodes and spreads first to anatomically contiguous lymphoid tissues
Mesenteric lymph nodes and Waldeyer ring rarely involved
Extranodal presentation is rare
Presence of Reed-Sternberg cells: transformed B cells
fever!

215
Q

Non Hodgkin lymphoma

A

non-Hodgkin Lymphoma vs Hodgkin Lymphoma
non-Hodgkin Lymphoma has:
Frequent involvement of multiple peripheral lymph nodes
Noncontiguous spread (not predictable)
Waldeyer ring and mesenteric lymph nodes commonly involved
Extranodal presentation common

216
Q

Hodgkin

A

efinition
average age: 32
treated with radiation and chemotherapy, curable in most cases
Need to see Reed-Sternberg cells on a background of non-neoplastic inflammatory cells

217
Q

5 subtypes of Hodgkin

A
Nodular sclerosis (classic)
Mixed cellularity (classic)
Lymphocyte rich (classic)
Lymphocyte depleted (classic)
Lymphocyte predominance: Reed-Sternberg cells have distinctive B-cell immunophenotype
218
Q

Classical Hodgkin

A

Classical: frequently associated with acquired mutations that activate NFκB and with EBV infection
Lymphocyte Predominant: express B-cell markers and is not associated with EBV infection

219
Q

Hodgkin lymphoma diagnosis

A

Di agnosis:

depends on the identification of Reed-Sternberg cells in a background of non-neoplastic inflammatory cells

220
Q

Pathogenesis s

A

Pathogenesis
Ig genes have undergone V(D)J recombination and somatic hypermutation, but fail to be expressed
NFκB activation

221
Q

NFKB activation hodgkin

A
EBV infection (or other mechanism promoting lymphocyte survival and proliferation)
EBV+ cells have LMP1 = upregulation of NFκB
Acquired loss-of-function mutations in IκB or A20 (both negative regulators of NFκB)
Rescue of "crippled" germinal centers by EBV creates an environment of acquisition of other mutations --> Reed-Sternberg cells
222
Q

Reed sternberg cells

A

Reed-Sternberg Cells == “Owl-Eyed Nuclei”
Definition
Derived from germinal center or post-germinal center B-cells
Reed-Sternberg cells are transformed B cells
despite having the genetic signature of a B-cell, the Reed-Sternberg cells of classical Hodgkin Lymphoma fail to express most B-cell specific genes, including the immunoglobulin genes
Release factors (cytokines, chemokines, etc.) inducing accumulation of reactive lymphocytes, macrophages, and granulocytes (> 90% of tumor cells)

223
Q

Morphology reed sternberg cells

A

Large, aneuploid B cells with multiple nuclei or a single nucleus with multiple lobes and inclusion-like nucleolus == Owl-Eyed Nuclei
Clearing around the nucleoli; abundant cytoplasm is seen
background lymphocytes will be reactive
Immunophenotype
REL protooncogene (chromosome 2p) often gained that increase NFκB
CD15, CD30, and CD40 positivity (activated via eosinophils and T-cells)
Immunoglobulin (-)

224
Q

Mononuclear variants

A

Contain a single round or oblong nucleus with large inclusion like-nucleolus

225
Q

Lacunar variants

A

Seen in nodular sclerosis subtype
Delicate, folded or multilobate nuclei
Abundant pale cytoplasm that retracts during tissue processing = nucleus in an empty hole (a lacuna)
Seen in the nodular sclerosis type of Hodgkin Lymphoma

226
Q

Mummification

A

Mummification: the process by which the Reed-Sternberg cells die
The cells shrink and become pyknotic

227
Q

Lymphohistiocytosic variants

A
Lymphohistiocytic Variants (L&amp;H cells)
Polypoid (multilobed) nuclei
Inconspicuous nucleoli
Moderate cytoplasm
"Popcorn cells"
Seen in the lymphocyte predominant subtype of Hodgkin Lymphoma
Express CD20, BCL6
No CD15 or CD30
228
Q

HODGKIN LYMPHOMA CLASSIC immunophenotype

A

Classic Immunophenotype
PAX5 positive
CD15 and CD30 positive
except lymphocyte predominant
No other B-cell, T-cell markers, or CD45 (leukocyte common antigen)
Hodgkin Lymphoma does not present in extranodal locations

229
Q

Hodgkin lymphoma nodular sclerosis subtype

A

Most common form of Hodgkin Lymphoma (65-70%)
Adolescents and young adults
Excellent prognosis

230
Q

Morphology nodular sclerosis subtype

A

Lacunar variant Reed-Sternberg cells that are uncommonly found == lacunar cells
Deposition of collagen bands that divide lymph nodes into circumscribed nodules
Often involves cervical, supraclavicular and mediastinal lymph nodes
No EBV
Classic immunophenotype
PAX5 positive
CD15 and CD30 positive
No other B-cell, T-cell markers, or CD45 (leukocyte common antigen

231
Q

Mixed cellularity subtype

A
Second most common subtype
Male predominance
Biphasic age distribution
Signs and symptoms include night sweats and weight loss
Commonly present in advanced tumor stage
Prognosis is very good
232
Q

Morphology hodgkin

A

Diagnostic Reed-Sternberg cells and mononuclear variants are plentiful
lymph nodes are diffusely effaced
70% of Reed-Sternberg cells are EBV+
Classic immunophenotype
PAX5 positive
CD15 and CD30 positive
No other B-cell, T-cell markers, or CD45 (leukocyte common antigen

233
Q

Hodgkin lymphoma lymphocyte rich rubtype

A

Reactive lymphocytes make up the vast majority of the cellular infiltrate
Uncommon
lymph nodes are diffusely effaced with vague nodularity due to occasional residual B-cell follicles
Very good to excellent prognosis

234
Q

Morphology lymphocyte rich subtype

A
Frequent mononuclear variants
Diagnostic Reed-Sternberg cells
40% of cases are EBV+
Classic immunophenotype
PAX5 positive
CD15 and CD30 positive 
No other B-cell, T-cell markers, or CD45 (leukocyte common antigen
235
Q

Definition lymphocyte depletion subtype

A

Least common form of Hodgkin Lymphoma
Elderly, HIV+ individuals, non-industrialized countries
Advanced stage and systemic signs and symptoms are frequent
Prognosis is less favorable than other subtypes

236
Q

Morphology lymphocyte depletion subtype

A

Scarce lymphocytes
Abundant Reed-Sternberg cells or pleomorphic variants
EBV+ in 90% of cases
Classic immunophenotype
PAX5 positive
CD15 and CD30 positive
No other B-cell, T-cell markers, or CD45 (leukocyte common antigen)
Essential to do immunophenotype, as most tumors with lymphocyte depletion are actually large cell non-Hodgkin Lymphoma

237
Q

Lymphocyte predominance subtype

A

Nonclassical Hodgkin Lymphoma subtype
Male < 35 years have cervical or axillary lymphadenopathy
Excellent prognosis, even though it is more likely to recur as compared to the other subtypes
lymph nodes are effaced with a nodular infiltrate of small lymphocytes and macrophages

238
Q

Immunophenotype lymphocyte predominance subtype

A

munophenotype: L&H variants express B-cell markers in contrast to Reed-Sternberg cells from classical forms of Hodgkin Lymphoma
Express CD20, BCL6 (unique)
CD15 and CD30 negative

239
Q

Morphology lymphocyte predominance subtypes

A

Not associated with EBV
IgH genes have ongoing somatic hypermutation – a feature found only in germinal centers
35% transform to diffuse large B-cell lymphoma due to expanded B-cell follicles
Classic Reed-Sternberg cells are difficult to find
L&H variant cells “popcorn cells” are seen
Eosinophils and plasma cells are scant or absent

240
Q

Nodular sclerosis or lymphocyte predominant subtypes clincial

A

Typically stage I-II = free of systemic manifestations

90% cure rate

241
Q

Mixed cellularity or lymphocyte depletion or disseminated disease clincial

A

Stage III-IV → constitutional signs and symptoms
5 year survival 60-70%
Anergy (cutaneous immune unresponsive)
TH1 suppression due to factors released by Reed-Sternberg cells

242
Q

Hodgkin lymphoma progression

A

Generally presents as painless lymphadenopathy

spread is stereotyped: nodal disease → splenic disease → hepatic disease → marrow and other tissues

243
Q

Prognosis hodgkin lymphoma

A

Tumor stage (not histology) is most important prognostic factor
Low stage 1st cured with involved field radiotherapy: increased risk of radiotherapy induced malignancies (lung cancer, melanoma, breast cancer)
2° tumors reduced by using minimal radiation and less genotoxic chemo without loss of therapeutic efficacy
AntiCD30 antibodies produce excellent response in patients failing conventional methods
wouldn’t work in Lymphocyte Predominant subtype as this is CD30 negative

244
Q

Stage I

A

Hodgkin Lymphoma: Stage I
Involvement of a single lymph node region (I)
Involvement of a single extra-lymphatic organ or site (IE)

245
Q

Hodgkin II

A

Hodgkin Lymphoma: Stage II
Involvement of 2+ lymph node regions on the same side of the diaphragm alone (II)
Localized involvement of an extra-lymphatic organ or site (IIE)

246
Q

Hodgkin III

A

Hodgkin Lymphoma: Stage III
Involvement of lymph nodes on both sides of the diaphragm without (III) or with (IIIE) localized involvement of an extra-lymphatic organ or site

247
Q

Hodgkin IV

A

Hodgkin Lymphoma: Stage IV

Diffuse involvement of one or more extra-lymphatic organs or sites +/- lymphatic involvement

248
Q

Acute myeloid leukemia

A

a: accumulation of immature myeloid forms (blasts) in the bone marrow suppresses normal hematopoiesis

249
Q

Myelodysplastic syndromes

A

D efective maturation of myeloid progenitors gives rise to ineffective hematopoiesis, leading to cytopenias; group of clonal stem cell disorders characterized by maturation defects that are associated with ineffective hematopoiesis and a high risk of transformation to AML
generally have a poor prognosis

250
Q

Myeloproliferative disorders

A

Myeloproliferative disorders: there is usually increased production of one or more types of blood cells

251
Q

AML

A

Definition
Accumulation of at least 20% or > immature myeloid blasts in the bone marrow due to acquired oncogenic mutations impeding differentiation
Causes marrow failure
Complications: anemia thrombocytopenia, neutropenia
Increased incidence throughout life, peak after 60 years old
Diagnosis is confirmed with myeloid specific antigen stains (i.e. MPO)
diagnosis of AML is based on the presence of at least 20% myeloid blasts in the bone marrow
Two most common chromosomal rearrangements == t(8:21) and inv(16)
t(8;21) == RUNX1 disrupted
inv(16) == CBFB disrupted
RUNX1 and CBFB normally bind one another to form a RUNX1/CBF1β transcription factor that is required for normal hematopoiesis

252
Q

Class I

A

Genetic aberrations with favorable prognosis
t(8:21) == RUNX1 disrupted
inv(16) == CBFB disrupted
Normal cytogenetics, mutated NPM
Genetic aberrations with intermediate prognosis: t(15:17) – acute promyelocytic leukemia (subtype)
Genetic aberrations with poor prognosis: t(11:q23:v)

253
Q

Class II

A
AML with MDS-like features (myelodysplastic syndrome)
Prior MDS
Multi-lineage dysplasia
MDS-like cytogenic aberrations
All have poor prognosis
254
Q

Class III

A

AML that is therapy related
Post alkylating agent or radiation therapy = 2-8 year latency, MDS-like cytogenic aberrations
Post topoisomerase II inhibitor = 1-3 year latency, MLL translocations
Very poor prognosis

255
Q

Class IV

A

ass IV
All AML’s that lack features of the other classes (NOS)
Classified based on degree of differentiation and lineage of leukemic blasts
Intermediate prognosis

256
Q

AML FAB class MO

A

Minimally differentiated AML
2-3% of cases
Blasts lack cytologic and cytochemical markers of myeloblasts
Express myeloid lineage antigens and ultra-structurally resemble myeloblasts

257
Q

AML FAB class M1

A
AML without maturation
20% of cases
Very immature cells
≥ 3% are MPO (+)
Few granules or Auer rods
Little maturation past myeloblast stage
258
Q

AML FAB class M2

A
AML with myelocytic maturation
M2 == AML
M3 == APL (P and 3)
M4 = AMML (has four letters)
30-40% of cases
Full range of myeloid maturation throughout granulocytes
Auer rods present in most cases
t(8:21
259
Q

AML FAB class M3

A
Acute promyelocytic leukemia (APL)
M2 == AML
M3 == APL (P and 3)
M4 = AMML (has four letters)
5-10% of cases
Most cells are hypergranular promyelocytes
Many Auer rods /cell
Patients are younger (35-40 years old)
increased incidence of DIC
t(15:17) with Flt3
260
Q

AML FAB class M4

A

AML: FAB Class M4
AML with MyeloMonocytic maturation (Acute MyeloMonocytic Leukemia (AMML)
M2 == AML
M3 == APL (P and 3)
M4 = AMML (has four letters)
15-20% of cases
Myelocytic and monocytic differentiation is evident
Myeloid elements with range of maturation
MPO positive
Monoblasts are positive for non-specific esterase
non-specific esterase is non-specific; positivity only tells you that there are monocytes present
diagnostic in the setting of AML; non-specific anywhere else
Inv(16

261
Q

AML FAB class M5 a/b

A

AML with monocytic maturation (acute monocytic leukemia)
10% of cases
Monoblasts: MPO (-), non-specific esterase positive
Promonocytes
Older patients
High incidence of organomegaly, lymphadenopathy and tissue infiltration

262
Q

AML FAB class M6 a/b

A

AML with erythroid maturation (acute erythroleukemia)
Dysplastic erythroid precursors (megaloblastoid or multiple nuclei) dominate
Nonerythroid cells = >30% myeloblasts
Elderly patients
*20% of therapy related AML

263
Q

AML FAB class M7

A

AML with megakaryoblastic maturation (acute megakaryocytic leukemia)
Blasts of megakaryocytic lineage predominate
Blasts react with platelet specific antibodies (GPiib/iiia or vWF)
Myelofibrosis or increased reticulin

264
Q

Pathogenesis

A

Genetic aberrations disrupt genes encoding transcription factors required for normal myeloid differentiation
t(8:21) RUNX1, inv(16) CBFB which lead to blocking of myeloid cell maturation
Mutations activating GF signaling pathways
t(15:17) → PML-RARα fusion protein interferes with terminal differentiation of granulocytes
treatment: all-trans retinoic acid or arsenic trioxide (chapter 7)
t(15;17) == acute promyelocytic leukemia subtype of AML
may exhibit early, significant, life-threatening bleeding
DIC, once established, is often fatal
also have frequent activating mutations of FLT3 mutation (receptor tyrosine kinase)
Epigenetic alterations: DNA methylation and posttranslational modification of histones is altered

265
Q

Morphology

A

Diagnosis is made by > 20% myeloid blasts in the bone marrow
Myeloblasts
Monoblasts
Megakaryoblast
Erythroblast
*blasts may be entirely absent from the blood (aleukemic leukemia), hence a bone marrow examination is essential (especially in pancytopenic patients

266
Q

Myeloblast morphology

A

Delicate nuclear chromatin
2-4 nucleoli
Voluminous cytoplasm (more than lymphoblasts)
Peroxidase (+) azurophilic granules
Auer rods: distinctive needle like azurophilic granules
particularly numerous in AML with the t(15:17) (acute promyelocytic leukemia)
Auer rods are formed of the cytoplasmic granules of the myeloid blasts of acute myelogenous leukemia (AML) and are a typical finding with AML. Myelodysplastic syndromes can precede development of AML, as can some cases of myeloproliferative disorders, paroxysmal nocturnal hemoglobinuria, and chemotherapeutic regimens

267
Q

Monoblast morphology

A
onoblast morphology
Folded or lobulated nuclei
Lack Auer rods
Nonspecific esterase (+)
Peroxidase (-)
268
Q

Cytogenetics based on clincial scenario

A

Younger adults: Balanced translocations t(8:21), inv(16), and t(15:17)
Following MDS or chemo or radiation: Deletions or monosomy of chromosome 5 or 7 with a lack of translocations
Following topoisomerase II inhibitors: MLL translocation chromosome 11q23
Older adults: Worse aberrations (deletion of 5q, 7q)

269
Q

Clincal presentation

A

Signs and symptoms very similar to ALL
Anemia, fatigue, neutropenia.
Fever due to opportunistic infections of oral cavity, skin, lungs, kidneys, bladder, colon (fungi, Pseudomonas, commensals)
Thrombocytopenia = spontaneous mucosal and cutaneous bleeding (gingiva, urinary tract); cutaneous petechiae and ecchymoses; exacerbated by procoagulants and fibrinolytics released by leukemic cells (especially in AML with the t(15;17

270
Q

Tumor location

A

Signs and symptoms due to involvement of tissues other than bone marrow is less striking than in ALL
CNS involvement is less common
Often infiltrate the skin (leukemic cutis) and gingiva
May present as a localized soft-tissue mass (myeloblastoma, granulocytic sarcoma/chloroma)
Without systemic treatment these will progress to full-blown disease

271
Q

Prognosis

A

60% remission with chemotherapy
5 years: 70-85% relapse
Molecular subtype targeted therapy improves outcomes
80% of t(15:17) are cured with all-trans retinoic acid and arsenic salts
t(8:21), inv(16) without KIT mutation = good prognosis with chemotherapy
AMLs that follow MDS or genotoxic therapy, in older adults, or returned AML = dismal prognosis
HSC transplant is performed with these “high-risk” disease

272
Q

Myelodysplastic syndromes are more severe than myeloproliferative

A

MDS generally have a poor prognosis

273
Q

Myelodysplastic syndromes

A

Clonal stem cell disorders with maturation defects associated with ineffective hematopoiesis
Increased risk of transformation to AML
Bone marrow is partly or wholly replaced by clonal progeny of a neoplastic multipotent stem cell with the capacity to differentiate (but does so ineffectively)
Peripheral blood *pancytopenias occur
primary MDS == idiopathic
t-MDS == secondary to previous genotoxic drug or radiation therapy (iatrogenic?)
More severe
Typically, older adults
myelodysplasias are characterized by thrombocytopenia

274
Q

Cause MDS

A

May be primary or secondary to genotoxic drug or radiation (t-MDS) and appears 2-8 years after exposure
Transformation to AML occurs most rapidly and most frequently in t-MDS

275
Q

Pathogenesis MDS

A

Mutations of epigenetic factors regulating DNA methylation and histone modifications
Mutations of RNA splicing factors involving the 3’ end of the RNA splicing machinery
Mutation of transcription factors for normal myelopoiesis
10% have TP53 loss-of-function mutations (poor outcome)
Monosomies 5 and 7; deletions of 5q, 7q, 20q; trisomy 8
MYC gene is on chromosome 8

276
Q

Morphology

A

Hypercellular marrow is common, but can also be normal or hypocellular
Disordered differentiation of erythroid, granulocytic, monocytic and megakaryocytic lineages to varying degrees is the most characteristic finding

277
Q

Erythroid lineage effects

A

Ring sideroblasts: erythroblasts with iron-laden mitochondria visible as perinuclear granules
seen with Prussian blue stain of aspirates or biopsies
Megaloblastoid maturation similar to B12 and folate deficiencies
Nuclear budding problems = misshapen nuclei (often with polypoid outlines

278
Q

Granulocytes lineage effects

A

Neutrophils with decreased numbers of secondary granules, toxic granulations or Dohle bodies
Pseudo-Pelger-Hüet cells: Neutrophils with only two nuclear lobes
Hüet == Duet
Neutrophils lacking nuclear segmentation
Myeloblasts may be increased but account for < 20% of the overall marrow cellularity
recall: AML is when myeloblasts account for at least 20% of the marrow aspirate

279
Q

Megakaryocytic lineage effects

A

Cells with single nuclear lobes or multiple separate nuclei (‘pawn ball’) are characteristic

280
Q

Primary MDS clincial

A

Average age: 70 years
50% discovered incidentally on routine blood tests
Survive 9-29 months (up to 5+ years)
Signs and symptoms of weakness, infection, hemorrhage (due to pancytopenia)
Worse outcomes: higher blast counts, more severe cytopenias, multiple clonal chromosomal abnormalities
10-40% progress to AML (high rate of transformation to AML is in the definition)
cause of death: thrombocytopenia (bleeding) and neutropenia (infection

281
Q

T-MDS clincial

A

Appears 2-8 years after treatment with genotoxic agents
Median survival of 4.8 months – terrible prognosis
Cytopenias are severe
Progression to AML is rapid

282
Q

Treatment

A

HSC transplant in young patients
Older patients receive antibiotics and transfusions
Thalidomide-like drugs and DNA methylation inhibitors
Improve effectiveness of hematopoiesis and peripheral blood counts in some patients

283
Q

FOUR Myeloproliferative Disorders == CML, polycythemia vera, essential thrombocytosis, and primary myelofibrosis

A

Mutated, constitutively active tyrosine kinases or other aberrations in signaling pathways → growth factor independence so there are lots of cells
tyrosine kinase inhibitors are a big deal here (e.g. Gleevec!)
Do not impair differentiation
Increased production of one or more mature blood elements – high counts of everything (pancytosis)
Originate in multipotent myeloid progenitors
Paradoxically, there is bleeding because the platelets are functionally abnormal, even though there are lots of them

284
Q

Clinical

A

Increased proliferative drive in the bone marrow – tend to have thrombocytosis
Displace normal bone and suppress hematopoiesis
Homing of neoplastic stem cells to secondary hematopoietic organs → extramedullary hematopoiesis (liver)
Variable transformation to a spent phase with marrow fibrosis and peripheral blood cytopenias
Variable transformation to acute leukemia

285
Q

Complications

A

Can either turn into AML (from the common myeloid progenitor) or go into a spent phase
A few will turn into an ALL and they have a terrible prognosis, even worse than if they turn into an AML

286
Q

CML

A

Chimeric BCR-ABL gene driven by t(9:22), Philadelphia chromosome
Constitutive BCR-ABL tyrosine kinase activation driving growth factor independent proliferation and survival of bone marrow progenitors
Does not interfere with differentiation
Originates from pluripotent hematopoietic stem cell
Preferentially drives proliferation of granulocytic and megakaryocytic progenitors
Most common of the myeloproliferative disorders

287
Q

Morphology CML

A

Hypercellular marrow from increased maturing granulocytic precursors
**Basophilia + eosinophilia
Increased megakaryocytes (small, dysplastic), increased platelets
Erythroid precursors are found at normal levels
Sea-blue histiocytes are a characteristic finding
Increased reticulin deposition

288
Q

Sea blue histiocytes

A

Scattered macrophages with abundant wrinkled, green-blue cytoplasm
Seen in chronic myelogenous leukemia (CML)

289
Q

Clincial CML

A

Primarily a disease of adults (5-6th decade) with insidious onset
Putthoff: male predominance
Huge spleen (extramedullary hematopoiesis) +/- infarcts, potential hepatomegaly, and lymphadenopathy
patients may present with fatigue, weight loss, anemia, anorexia and hyper-metabolism (increased cell turnover) or LUQ pain due to splenomegaly (dragging sensation) or splenic infarction
Much worse prognosis if they don’t have the Philadelphia chromosome
Gleevec treats the Philadelphia chromosome

290
Q

Diagnose CML

A

BCR-ABL gene is detected via PCR or chromosome analysis
Leukocyte alkaline phosphatase (LAP) is low
LAP will be high in reactive states, e.g. infection
Blasts are < 10% of circulating cells (has to be less than 20% or else it’s an AML

291
Q

Prognosis cml

A

Without treatment patients survive 3 years
“accelerated phase” == increased anemia and thrombocytopenia +/- basophilia
within 6-12 months, the accelerated phase terminates in a picture resembling acute leukemia (blast crisis)
70% are myeloid in origin, 30% are lymphoid in origin indicating the disease originates from a pluripotent stem cell with both myeloid and lymphoid potential

292
Q

Treatment CML

A

90% of patients achieve remission with BCR-ABL inhibitors (Gleevec!)
Resistance has emerged due to mutations and are treated with 2nd and 3rd generation kinase inhibitors
hematopoietic stem cell transplant may be offered to young patients (75% cured)
Prognosis is less favorable when accelerated phase or blast crisis supervenes

293
Q

Polycythemia Vera

A

Polycythemia Vera (PCV) == markedly increased red cell mass
Definition
Activating point mutation in JAK2 (tyrosine kinase)
Greatly increased RBCs (main source of signs and symptoms), slight increase in granulocytes and platelets

294
Q

Pathogenesis PCV

A

Valine → phenylalanine at residue 617 – feels like a Dobson question
JAK2 then acts independently of growth factor
two mutated copies: higher WBC counts, more significant splenomegaly, pruritus and greater progression rate of the spent phase
Constitutively active JAK2 affects the JAK/STAT signaling pathway, downstream of the hematopoietic growth factor receptors
Decreased requirements for EPO (often suppressed

295
Q

PCV morphology

A

Subtle increase in RBC progenitors with an associated increase in granulocytic precursors and megakaryocytes
Hypercellular marrow with residual fat
Increased reticulin fibers (10%)
Early phase: organomegaly due to congestion, and peripheral blood shows basophilia and large platelets

296
Q

Spent phase pcv

A

Late in the course
Extensive marrow fibrosis that displaces hematopoietic cells, increased extramedullary hematopoiesis (hepatosplenomegaly)
1% transform to AML

297
Q

Clincial pcv

A

Insidious, uncommon disease of adults
Signs and symptoms related to increased RBC mass and hematocrit (hyperviscosity) → abnormal blood flow
elevated hematocrit –> increased blood viscosity, + thrombocytosis and abnormal platelet function –> –> patients with PVC are prone to both thrombosis and bleeding
Venous circulation becomes congested and distended (patients are “plethoric” in the face (flushed))
pruritus after taking a hot shower
Deoxygenated blood remains in peripheral vessels longer → HA, dizziness, HTN, GI signs and symptoms

298
Q

Sequelae pcv

A

Increased risk of thrombosis: DVT, hepatic vein (Budd-Chiari), portal vein, dural sinus thrombi
Pruritus due to basophil release of histamine, can also cause peptic ulcers
Hyperuricemia due to high cell turnover (gout

299
Q

Lab findings pcv

A

Increased hemoglobin
Greatly increased hematocrit (>60%)
Iron deficiency due to chronic bleeding can suppress EPO, which decreases hematocrit to the normal range
Increased WBC (leukocytosis)
increased platelet count with morphologic abnormalities (giant with defective aggregation

300
Q

Prognosis pcv

A

Untreated = death within months – not good
Phlebotomy, maintaining normal RBC mass extends survival to 10+ years
Evolves to a spent phase after this
2% transform to AML lacking JAK2 mutations
Disease is likely due to a progenitor mutation committed to myeloid differentiation
ALL transformation is rare (different from CML)

301
Q

Spent phase pcv

A

15-20% of patients transition around 10 years into their disease (less common than other MPD)
Obliterative fibrosis of marrow (myelofibrosis)
Extensive medullary hematopoiesis (*spleen)
Massive splenomegaly

302
Q

Essential thrombocytosis

A

Elevated platelet counts without polycythemia or marrow fibrosis
Increased megakaryocytes
Mild leukocytosis
Diagnosis of exclusion

303
Q

Mutations essential thrombocytosis

A

mportant mutations
Activating JAK2 mutation (50%)
MPL (5-10%) [receptor tyrosine kinase normally activated by thrombopoietin]
Calreticulin

304
Q

Clincial esssential thrombocytosis

A

Adults > 60 (signs and symptoms may resemble PCV)
Extramedullary hematopoiesis → organomegaly
Thrombosis and hemorrhage due to platelet dysfunction and increased number
DVT, portal and hepatic vein thrombosis, myocardial infarct
Thrombosis in patients with increased platelet counts and homozygous JAK2 mutations
Erythromelalgia: occlusion of small arterioles by platelet aggregates → burning and throbbing of hands and feet (may also be seen in PVC, peripheral neuropathy, peripheral vascular deficiency)
this is a non-specific finding

305
Q

Prognosis et

A

Insidious: long asymptomatic phase with occasional thrombotic or hemorrhagic crises
Survival: 12-15 years

306
Q

Treatment et

A

Treatment: gentle chemotherapy to suppress thrombopoiesis

307
Q

Primary myelofibrosis

A

Development of obliterative marrow fibrosis due to non-neoplastic fibroblasts
Decreased bone marrow hematopoiesis → cytopenias and extramedullary hematopoiesis
Appears identical to the spent phase of other myeloproliferative disorders
Least common of the MPDs
chief pathologic feature == extensive deposition of collagen in the marrow by non-neoplastic fibroblasts

308
Q

Mutations primary myelofibrosis

A

Activating JAK2 or MPL mutations (also calreticulin

309
Q

Pathogenesis PM

A

Extensive deposition of collagen in the marrow by non-neoplastic fibroblasts
Displaces normal cells = marrow failure
Inappropriately released megakaryocyte factors: PDGF and TGFβ – both cause fibrosis and scarring
^^ bother are fibroblast mitogens
Extramedullary hematopoiesis takes over in spleen, liver and lymph node
Red cell production is impaired –> anemia

310
Q

Morphology primary myelofibrosis

A

Marrow is hypercellular with large, dysplastic megakaryocytes that are abnormally clustered
Minimal fibrosis with leukocytosis and thrombocytosis

311
Q

Myelofibrosis morphology late

A

Marrow becomes hypocellular and diffusely fibrotic
“Cloudlike” clusters of megakaryocytes
(spleen?) Sinusoids are markedly dilated
Osteosclerosis (fibrotic marrow → bone

312
Q

Morphology spleen myelofibrosis

A

Marked splenomegaly, appearing red-gray and firm
Subcapsular infarcts occur
Initially hematopoiesis occurs in the sinusoids, but extends to the cords later
Liver and lymph nodes may also be involved in hematopoiesis (lymphadenopathy is uncommon)

313
Q

Morphology blood myelofibrosis

A

Leukoerythroblastosis: Premature release of nucleated erythroid and early granulocyte precursors
DaCRYocytes: Tear drop shaped cells damaged during release from the fibrotic marrow
seen in pulmonary myelofibrosis, myelophthisic anemia, thalassemia, s/p splenomegaly
Abnormally large platelets
Basophilia
*these are also seen in other infiltrative disorders of the marrow (granulomatous disease and metastatic tumors

314
Q

Clinical myelofibrosis

A

patients > 60 years old
Present with progressive anemia and LUQ “fullness”
Night sweats, fatigue and weight loss due to increased metabolism
Hyperuricemia and secondary gout due to high cell turnover
Diagnosis: bone marrow biopsy

315
Q

Lab finding myelofibrosis

A

Normocytic, normochromic anemia with leukoerythroblastosis
WBC count is variable
Platelet count is variable, but thrombocytopenia will eventually show

316
Q

Prognosis myelofibrosis

A

Prognosis
Survival: 3-5 years
Infection, thrombotic episodes, bleeding, transformation to AML (5-20%)

317
Q

Treat myelofibrosis

A

JAK2 inhibitors for splenomegaly and signs and symptoms and HSC in young patients

318
Q

Langerhans cell histiocytosis

A

Umbrella designation for a variety of proliferative disorders of dendritic cells or macrophages
considered neoplasms

319
Q

Morphology langerhans histiocytosis

A

Abundant, vacuolated cytoplasm
Vesicular nuclei with linear grooves or folds
*Birbeck granules in the cytoplasm is characteristic: pantalaminar tubules with a dilated terminal end (“tennis racket” appearance) *contain langerin
Immunophenoty

320
Q

Immunophenotype langerhans cell histiocytiosis

A

HLA-DR, S100, and CD1a positive

321
Q

Mutations langerhans cell histiocytosis

A

BRAF activating mutation (600: valine → glutamate) as seen in hairy cell leukemia
Other mutations in tp53, RAS and MET (a tyrosine kinase

322
Q

A 2-year-old boy has had a seborrheic eruption over the scalp and trunk over the past month. He then develops a right ear ache. On physical examination the right tympanic membrane is erythematous and bulging. He has hepatosplenomegaly and generalized lymphadenopathy. Laboratory studies show Hgb 9.5 g/dL, Hct 28.7%, MCV 90 fL, platelet count 58,000/microliter, and WBC count 3540/microliter. A bone marrow biopsy is performed and on microscopic examination shows 100% cellularity with extensive infiltration by cells resembling macrophages that express CD1a antigen and, by electron microscopy, have prominent HX bodies (Birbeck granules). Which of the following conditions is most likely to produce this boy’s findings

A

(D) CORRECT. The Langerhans cell histiocytoses include Letterer-Siwe disease (as in this case, it is typically a disseminated disease of children), and localized eosinophilic granuloma (often involving bone).

323
Q

Multifocal Multisystem Langerhans Cell Histiocytosis (Letterer-Siwe Disease

A

Malignant proliferation of Langerhans cells

Mostly seen in kids <2 years old, but can be seen in adults

324
Q

Clincial multifocal multisystem langerhans celll histiocytosis

A
Seborrheic eruption (rash) over the trunk and scalp
HSM, LAD, pulmonary lesions and destructive osteolytic bone lesions
Anemia, thrombocytopenia, recurrent infections (otitis media, mastoiditis) due to marrow involvement
Anaplastic = Langerhans cell sarcoma
325
Q

Prognosis treatment multifocal multisystem langerhans cell histiocytosis

A

Untreated = rapidly fatal
Treatment: intensive chemotherapy
5 year survival: 50%

326
Q

Unifocal Unisystem Langerhans Cell Histiocytosis (eosinophilic granuloma

A

Benign proliferation of Langerhans cells in medullary cavity of bone typically in the calvarium, ribs, femur
Adolescents

327
Q

Unifocal Unisystem Langerhans Cell Histiocytosis (eosinophilic granuloma) clincila

A

Pain, tenderness, pathologic fracture
Heal spontaneously, excised or radiated
Skin, lungs or stomach are uncommonly involved
Biopsy: Langerhans cells with mixed inflammatory cells (numerous eosinophils

328
Q

Multifocal unisystem langerhans cell histiocytosis

A

Definition
Benign proliferation of Langerhans cells
Affects young children

329
Q

Clincial multifocal unisystem langerhans cell histiocytosis

A

Multiple erosive bone masses
May extend into soft tissue
May involve posterior pituitary stalk = diabetes insipidus (50%)
Spontaneous regression or treatment with chemotherapy
Hand-Schuller-Christian Triad: Calvarial bone defects, diabetes insipidus, and exophthalmos – “probably should know that

330
Q

Pulmonary langerhans cell histiocytosis

A

Reactive proliferation of Langerhans cells
Nodules and cysts in the middle and upper lobes
40% are associated with BRAF mutations and may be neoplastic in origin – as seen in hairy cell leukemia
Adult smokers (disease may regress with smoking cessation)
pulmonary disease associated with smoking, shocking

331
Q

Immunophenotype of langerhans cell histiocytosis

A

Aberrant expression of chemokine receptors on Langerhans cells allows homing
Normally express CCR6
Neoplastic: CCR6 + CCR7 allowing cells to enter tissues in skin, bone, and lymphoid organs

332
Q

Periarteriolar lymphatic sheath

A

Periarteriolar lymphatic sheath: an artery with an eccentric collar of T lymphocytes; at intervals this sheath expands to form lymphoid nodules composed mainly of B lymphocytes

333
Q

Open circulation

A

open circulation: blood flows through capillaries into the cords and then squeezes through gaps in the discontinuous basement membrane of the endothelial linings to reach the sinusoids
cells are deformed when passing from cords to sinusoids

334
Q

Closed circulation

A

closed circulation: blood passes rapidly and directly from the capillaries to the splenic veins

335
Q

Spleen path

A

primary lesions to the spleen are rare; if they happen they are benign
metastatic disease to the spleen is rare

336
Q

Splenic phagocytosis

A

phagocytosis of blood cells and particulate matter
conditions in which red cell deformability is decreased, more RBCs become trapped in the cords and phagocytosed by macrophages
macrophage are responsible for RBC “pitting:” process by which inclusions of Heinz bodies and Howell-jolly bodies are excised
Also remove particles (encapsulated bacteria

337
Q

Splenic antibody production

A

Plasma cells within the sinuses of the red pulp
dendritic cells in periarteriolarl lymphatic sheaths present Antigens to T-lymphocytes
T & B cells interact at the edge of white pulp follicles –> plasma cells within the sinuses of the red pulp
Important production site against microbial polysaccharides and auto-antibodies to self-antigens

338
Q

Splenic function hematopoiesis

A

Major site during fetal development, but disappears at birth
In chronic anemia, thalassemia, and patients with myeloproliferative disorders (CML, primary myelofibrosis) becomes an extramedullary site for production

339
Q

Splenic sequesteration

A

Splenomegaly significantly increases the capacity from 30ml of RBCs
Normally contains 30-40% of the body’s platelets but can trap 80-90% of total platelet mass (in red pulp)
Thrombocytopenia and leukopenia can result when cells remain trapped in the spleen

340
Q

Major manifestation of spleen

A

infections outside of the US

alcohol and disease in the US

341
Q

Asplenia

A

Removal or dysfunction of this organ leads to increased susceptibility to sepsis caused by encapsulated bacteria
encapsulated bacteria == blood borne
Sickle Cell Patients –> increased autoinfarction of the spleen –> functionally asplenic (splenic insufficiency)
Decreased phagocytic capacity
Decreased antibody production
patients should be vaccinated against encapsulated bacteria: pneumococci, meningococci, and haemophilus influenzae

342
Q

Dragging sensationin LUQ

A

Due to splenomegaly
Discomfort after eating due to pressure on the stomach

splenomegaly can cause hypersplenism: anemia, leukopenia, and thrombocytopenia either alone or in combination

343
Q

Nonspecific acute splenitis

A

Reactive enlargement of the spleen that occurs due to blood borne infections (microbial agents and cytokines released due to immune response

344
Q

Morphology nonspecific acute splenitis

A

Enlarged and soft spleen
Major feature: acute congestion of the red pulp that may encroach on and efface lymphoid follicles
White pulp follicles may undergo necrosis especially due to hemolytic streptococcus
Abscess formation rarely occurs

345
Q

Congestive splenomeglay

A

Definition
Due to chronic venous outflow obstruction
Obstruction due to intrahepatic disorder affecting portal venous drainage

346
Q

Causes congestive splenomegaly

A

Cirrhosis (ETOH, parasitic schistosomiasis, pigment)
Extrahepatic disorders impinging on portal or splenic veins
Spontaneous portal vein thrombosis, pyelophlebitis (inflammation of the portal vein), infiltrating tumors
Causes portal or splenic vein hypertension
Systemic congestion (CHF) may produce a moderately enlarged spleen

347
Q

Morphology splenomegaly

A

Cut surface is gray-red to deep red
Firm
Thick, fibrous capsule
Red pulp: congested then fibrotic
Deposition of collagen in basement membrane of sinusoids that appear dilated (rigid walls) due to increased portal venous pressure
Hypersplenism (excess destruction) due to slow blood flow and increased exposure to macrophages

348
Q

Splenic infarct

A

Common lesions; due to occlusion of major splenic artery or branches
Lack of collateral blood supply
Common in markedly enlarged spleens due to tenuous blood supply that is easily compromised
Common site of emboli (from the heart

349
Q

Morphology splenic infarct

A

Bland: pale, wedge shaped, subcapsular, fibrous capsule
more common; splenic infarcts are usually bland
Septic: suppurative necrosis that leads to depressed scars
common in the setting of individuals with infective endocarditis of the mitral or aortic valves

350
Q

Spleen neoplasms

A

Primary neoplastic involvement is rare; when it does happen it is benign
Lymphoid and myeloid tumors often cause splenomegaly
Benign fibromas, osteomas, chondromas, lymphangiomas, hemangiomas may arise; often cavernous in type

351
Q

Spleen congenital anomalies

A

Complete absence is rare and associated with other anomalies (e.g. situs inversus and cardiac malformations)
Hypoplasia is more common
Accessory spleen (spleniculi): common; small, spherical structures histologically and functionally identical to the normal structure found anywhere in the abdominal cavity
very important to recognize in diseases with splenectomy as a treatment (i.e. hereditary spherocytosis and immune thrombocytopenia purpura

352
Q

Splenic rupture

A

more commonly precipitated by blunt trauma
Spontaneous: never involves a truly normal organ, but likely involves an underlying condition
most common predisposing conditions: infectious mononucleosis, malaria, typhoid fever, lymphoid neoplasm
Splenomegaly with a thin, tense capsule susceptible to rupture
rupture causes intraperitoneal hemorrhage that must be treated with splenectomy
Chronically: unlikely to rupture due to reactive fibrosis of the capsule

353
Q

Thymus

A

Derived from the 3rd and 4th pharyngeal pouches
Grows until puberty then involutes
Lobular structures contain
Thymic epithelial cells → Hassall corpuscles with keratinized cores
Immature T lymphocytes
Location of maturation of T cells
myoid cells (muscle-like cells) found in the thymus are thought to play some role in the development of myasthenia gravis (Myasthenia Gravisis an autoimmune disease which results in musclefatigability and weakness throughout the day. Symptoms improve with rest. Its main symptoms, which the ophthalmologist may encounter, areptosis, diplopia, variable extra-ocular muscle palsies or incomitant strabismus, and external ophthalmoplegia.)
fatigable ptosis
MOA: auto-antibodies against cholinesterase

354
Q

Thymic hypoplasia/asplasia

A

Thymic Hypoplasia/Aplasia (DiGeorge Syndrome)
problem with the development of 3rd and 4th pharyngeal pouches
Severe defects in cell-mediated immunity
Variable anomalies of parathyroid development (hypoparathyroidism) –> hypocalcemia
Chromosome 22q11 deletion == CATCH-22
Cardiac defects, Abnormal facies, Thymic hypoplasia, Cleft palate, and Hypocalcemia (from hypoparathyroidism) resulting from 22q11 deletions

355
Q

Thymic cysts

A

Uncommon, incidental finding
< 4 cm in diameter
Spherical or arborizing
Lined with stratified squamous to columnar epithelium
Fluid may be serous or mucinous
neoplastic thymic masses compress and distort adjacent normal thymus and sometimes cause cysts to form
presence of a thymic cyst in a symptomatic patient should invoke a search for adjacent neoplasm (lymphoma or thymoma

356
Q

Thymic hyperplasia

A

Thymic (Follicular) Hyperplasia == B cell proliferation in the thymus
Appearance of B-cell germinal centers in the thymus (thymic follicular hyperplasia)
thymic hyperplasia is a B-cell lesion! (think about it)
Most common in myasthenia gravis 65-75% (or Graves disease, systemic lupus erythematous, scleroderma, rheumatoid arthritis, and other autoimmune disorders)
May be naturally large for a patient’s age and can be mistaken for a thymoma
True enlargement of the thymus is rare

357
Q

Thymoma

A

hymoma == epithelial malignancy of squamous type cells
Definition
Tumor of thymic epithelial cells – these are squamous cells
thymoma is of squamous cell origin; this is an epithelial cancer; will not have abnormal peripheral blood smears (i.e. no blast count with a thymoma)
Typically contain benign immature T cells (thymocytes)
Adults > 40 years old; thymomas are rare in children
defined as either benign (proliferation of squamous cell component) or malignant (thymic epithelium)
look for cellular atypia to differentiate between benign and malignant

358
Q

Three histologic subtypes of thymoma

A

Benign
Cytologically benign and noninvasive == thymoma
Malignant
Cytologically benign but invasive or metastatic == invasive thymoma
no cellular atypia
Cytologically malignant (thymic carcinoma) == malignant thymoma
cellular atypia

359
Q

Location thymoma

A

Common in the anterior superior mediastinum (also a common location of some lymphomas)
May cause impingement of structures

360
Q

Morphology thymoma

A

Morphology
Lobulated, firm, gray-white masses
+/- cystic necrosis and calcification
Most are encapsulated
20-25% of the tumors penetrate the capsule and infiltrate peri-thymic tissues
unremarkable thymic cortical and medullary regions; no atypia; whorled

361
Q

Noninvasive Thomas

A

50% of all thymomas are mixed medullary type and cortical type epithelial cells
Tumors with substantial medullary epithelial cells are usually noninvasive
Medullary type epithelial cells: elongated/spindle shaped
Sparse infiltrate of thymocytes
Cortical type epithelial cells: polygonal

362
Q

Malignant thymoma type I invasive

A

Invasive Thymoma (Type 1)
20-25% of all thymomas
Tumor that is cytologically bland but locally invasive
by definition, invasive thymomas penetrate through the capsule into surrounding structures
Cells are usually cortical with abundant cytoplasm and round, vesicular nuclei
The prognosis depends on how much the tumor has invaded surrounding structures
minimal invasion + complete excision –> 90% 5 year survival – good prognosis
extensive invasion –> 50% 5 year survival – not great

363
Q

Thymic carcinoma type II malignant thymoma

A

5% of thymomas
Fleshy, invasive masses that metastasize (often to lungs)
Most are squamous cell carcinomas
second most common are lymphoepithelioma-like carcinoma
Sheets of cells with indistinct borders
Resemble nasopharyngeal carcinoma
EBV genomes are found in 50

364
Q

Thymoma clincila

A

40% of patients present with signs and symptoms related to mediastinum impingement
30-45% of patients present with myasthenia gravis
other associated autoimmune disorders: hypogammaglobulinemia, pure red cell aplasia, Graves disease, pernicious anemia, dermatomyositis-polymyositis, and Cushing syndrome
thymocytes that arise within thymocytes give rise to long-lived CD4+ and CD8+ T cells, and cortical thymomas rich in thymocytes are more likely to be associated with autoimmune disease (statistically

365
Q

Spleen

A

Periarteriolar lymphatic sheath: an artery with an eccentric collar of T lymphocytes; at intervals this sheath expands to form lymphoid nodules composed mainly of B lymphocytes
open circulation: blood flows through capillaries into the cords and then squeezes through gaps in the discontinuous basement membrane of the endothelial linings to reach the sinusoids
cells are deformed when passing from cords to sinusoids
closed circulation: blood passes rapidly and directly from the capillaries to the splenic veins

primary lesions to the spleen are rare; if they happen they are benign
metastatic disease to the spleen is rare

366
Q

Splenic function : phagocytosis

A

phagocytosis of blood cells and particulate matter
conditions in which red cell deformability is decreased, more RBCs become trapped in the cords and phagocytosed by macrophages
macrophage are responsible for RBC “pitting:” process by which inclusions of Heinz bodies and Howell-jolly bodies are excised
Also remove particles (encapsulated bacteria*)

367
Q

Splenic function: antibodies

A

Plasma cells within the sinuses of the red pulp
dendritic cells in periarteriolarl lymphatic sheaths present Antigens to T-lymphocytes
T & B cells interact at the edge of white pulp follicles –> plasma cells within the sinuses of the red pulp
Important production site against microbial polysaccharides and auto-antibodies to self-antigens

368
Q

Splenic function hematopoiesis

A

Major site during fetal development, but disappears at birth
In chronic anemia, thalassemia, and patients with myeloproliferative disorders (CML, primary myelofibrosis) becomes an extramedullary site for production

369
Q

Splenic function sequesteration

A

Splenomegaly significantly increases the capacity from 30ml of RBCs
Normally contains 30-40% of the body’s platelets but can trap 80-90% of total platelet mass (in red pulp)
Thrombocytopenia and leukopenia can result when cells remain trapped in the spleen

370
Q

Splenomegaly

A

splenomegaly is the major manifestation of disorders of the spleen
infections outside of the US
alcohol and disease in the US

371
Q

Asplenia

A

Removal or dysfunction of this organ leads to increased susceptibility to sepsis caused by encapsulated bacteria
encapsulated bacteria == blood borne
Sickle Cell Patients –> increased autoinfarction of the spleen –> functionally asplenic (splenic insufficiency)
Decreased phagocytic capacity
Decreased antibody production
patients should be vaccinated against encapsulated bacteria: pneumococci, meningococci, and haemophilus influenzae

372
Q

Dragging sensation LUQ

A

Due to splenomegaly
Discomfort after eating due to pressure on the stomach

splenomegaly can cause hypersplenism: anemia, leukopenia, and thrombocytopenia either alone or in combination

373
Q

Nonspecific acute splenitis

A

Reactive enlargement of the spleen that occurs due to blood borne infections (microbial agents and cytokines released due to immune response

374
Q

Morphology acute splenitis

A

Enlarged and soft spleen
Major feature: acute congestion of the red pulp that may encroach on and efface lymphoid follicles
White pulp follicles may undergo necrosis especially due to hemolytic streptococcus
Abscess formation rarely occurs

375
Q

Congestive splenomegaly

A

Due to chronic venous outflow obstruction

Obstruction due to intrahepatic disorder affecting portal venous drainage

376
Q

Causes congestive splenomegaly

A

Cirrhosis (ETOH, parasitic schistosomiasis, pigment)
Extrahepatic disorders impinging on portal or splenic veins
Spontaneous portal vein thrombosis, pyelophlebitis (inflammation of the portal vein), infiltrating tumors
Causes portal or splenic vein hypertension
Systemic congestion (CHF) may produce a moderately enlarged

377
Q

Morphology congestive splenomegaly

A

Cut surface is gray-red to deep red
Firm
Thick, fibrous capsule
Red pulp: congested then fibrotic
Deposition of collagen in basement membrane of sinusoids that appear dilated (rigid walls) due to increased portal venous pressure
Hypersplenism (excess destruction) due to slow blood flow and increased exposure to macrophages

378
Q

Splenic infarct

A

Common lesions; due to occlusion of major splenic artery or branches
Lack of collateral blood supply
Common in markedly enlarged spleens due to tenuous blood supply that is easily compromised
Common site of emboli (from the heart

379
Q

Morphology splenic infarct

A

Bland: pale, wedge shaped, subcapsular, fibrous capsule
more common; splenic infarcts are usually bland
Septic: suppurative necrosis that leads to depressed scars
common in the setting of individuals with infective endocarditis of the mitral or aortic valves

380
Q

Spleen neoplasma

A

Primary neoplastic involvement is rare; when it does happen it is benign
Lymphoid and myeloid tumors often cause splenomegaly
Benign fibromas, osteomas, chondromas, lymphangiomas, hemangiomas may arise; often cavernous in type

381
Q

Spleen congenital anomalies

A

n: Congenital Anomalies
Complete absence is rare and associated with other anomalies (e.g. situs inversus and cardiac malformations)
Hypoplasia is more common
Accessory spleen (spleniculi): common; small, spherical structures histologically and functionally identical to the normal structure found anywhere in the abdominal cavity
very important to recognize in diseases with splenectomy as a treatment (i.e. hereditary spherocytosis and immune thrombocytopenia purpura

382
Q

Splenic rupture

A

more commonly precipitated by blunt trauma
Spontaneous: never involves a truly normal organ, but likely involves an underlying condition
most common predisposing conditions: infectious mononucleosis, malaria, typhoid fever, lymphoid neoplasm
Splenomegaly with a thin, tense capsule susceptible to rupture
rupture causes intraperitoneal hemorrhage that must be treated with splenectomy
Chronically: unlikely to rupture due to reactive fibrosis of the capsule

383
Q

Thymus

A

Derived from the 3rd and 4th pharyngeal pouches
Grows until puberty then involutes
Lobular structures contain
Thymic epithelial cells → Hassall corpuscles with keratinized cores
Immature T lymphocytes
Location of maturation of T cells
myoid cells (muscle-like cells) found in the thymus are thought to play some role in the development of myasthenia gravis (Myasthenia Gravisis an autoimmune disease which results in musclefatigability and weakness throughout the day. Symptoms improve with rest. Its main symptoms, which the ophthalmologist may encounter, areptosis, diplopia, variable extra-ocular muscle palsies or incomitant strabismus, and external ophthalmoplegia.)
fatigable ptosis
MOA: auto-antibodies against cholinesterase

384
Q

Digeorge

A

Thymic Hypoplasia/Aplasia (DiGeorge Syndrome)
problem with the development of 3rd and 4th pharyngeal pouches
Severe defects in cell-mediated immunity
Variable anomalies of parathyroid development (hypoparathyroidism) –> hypocalcemia
Chromosome 22q11 deletion == CATCH-22
Cardiac defects, Abnormal facies, Thymic hypoplasia, Cleft palate, and Hypocalcemia (from hypoparathyroidism) resulting from 22q11 deletions

385
Q

Thymic Systems

A

Uncommon, incidental finding
< 4 cm in diameter
Spherical or arborizing
Lined with stratified squamous to columnar epithelium
Fluid may be serous or mucinous
neoplastic thymic masses compress and distort adjacent normal thymus and sometimes cause cysts to form
presence of a thymic cyst in a symptomatic patient should invoke a search for adjacent neoplasm (lymphoma or thymoma

386
Q

Thymic follicular hyperplasia

A

Thymic (Follicular) Hyperplasia == B cell proliferation in the thymus
Appearance of B-cell germinal centers in the thymus (thymic follicular hyperplasia)
thymic hyperplasia is a B-cell lesion! (think about it)
Most common in myasthenia gravis 65-75% (or Graves disease, systemic lupus erythematous, scleroderma, rheumatoid arthritis, and other autoimmune disorders)
May be naturally large for a patient’s age and can be mistaken for a thymoma
True enlargement of the thymus is rare

387
Q

Thymoma

A

Tumor of thymic epithelial cells – these are squamous cells
thymoma is of squamous cell origin; this is an epithelial cancer; will not have abnormal peripheral blood smears (i.e. no blast count with a thymoma)
Typically contain benign immature T cells (thymocytes)
Adults > 40 years old; thymomas are rare in children
defined as either benign (proliferation of squamous cell component) or malignant (thymic epithelium)
look for cellular atypia to differentiate between benign and malignant

388
Q

Benign thymoma

A

Cytologically benign and noninvasive == thymoma

389
Q

Malignant thymoma

A

Cytologically benign but invasive or metastatic == invasive thymoma
no cellular atypia
Cytologically malignant (thymic carcinoma) == malignant thymoma
cellular atypia

390
Q

Location thymus tumor

A

Common in the anterior superior mediastinum (also a common location of some lymphomas)
May cause impingement of structures

391
Q

Morphology thymic tumor

A

ocation of the tumor
Common in the anterior superior mediastinum (also a common location of some lymphomas)
May cause impingement of structures
Morphology
Lobulated, firm, gray-white masses
+/- cystic necrosis and calcification
Most are encapsulated
20-25% of the tumors penetrate the capsule and infiltrate peri-thymic tissues
unremarkable thymic cortical and medullary regions; no atypia; whorled

392
Q

Noninvasive thymoma

A

50% of all thymomas are mixed medullary type and cortical type epithelial cells
Tumors with substantial medullary epithelial cells are usually noninvasive
Medullary type epithelial cells: elongated/spindle shaped
Sparse infiltrate of thymocytes
Cortical type epithelial cells: polygonal

393
Q

Malignan thymoma : type 1

A

Invasive Thymoma (Type 1)
20-25% of all thymomas
Tumor that is cytologically bland but locally invasive
by definition, invasive thymomas penetrate through the capsule into surrounding structures
Cells are usually cortical with abundant cytoplasm and round, vesicular nuclei
The prognosis depends on how much the tumor has invaded surrounding structures
minimal invasion + complete excision –> 90% 5 year survival – good prognosis
extensive invasion –> 50% 5 year survival – not great

394
Q

Malignant thymoma type 2

A

5% of thymomas
Fleshy, invasive masses that metastasize (often to lungs)
Most are squamous cell carcinomas
second most common are lymphoepithelioma-like carcinoma
Sheets of cells with indistinct borders
Resemble nasopharyngeal carcinoma
EBV genomes are found in 50%

395
Q

Thymoma clinical

A

40% of patients present with signs and symptoms related to mediastinum impingement
30-45% of patients present with myasthenia gravis
other associated autoimmune disorders: hypogammaglobulinemia, pure red cell aplasia, Graves disease, pernicious anemia, dermatomyositis-polymyositis, and Cushing syndrome
thymocytes that arise within thymocytes give rise to long-lived CD4+ and CD8+ T cells, and cortical thymomas rich in thymocytes are more likely to be associated with autoimmune disease (statistically