Midterm Flashcards

1
Q

Where do blood cell progenitors derive from embryologically

A

Yolk sac

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

Where do definitive hematopoietic stem cells arise from

A

Mesoderm of the intraembryonic aorta/gonad/mesonephros region; they then migrate to liver which becomes the chief site of blood cell formation until shortly before birth

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

What are the formed elements of the blood

A

Red cells, granulocytes, monocytes, platelets, lymphocytes

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

What are the progenitors of neutrophils,eosinophils basophils, platelet, and erythrocytes

A
  • neutrophils: myeloblasts
  • Monocytes: monoblast
  • eosinophil: eosinophiloblast
  • basophils: basophiloblast
  • platelets: megakaryoblast
  • erythrocytes: erythroblast
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q

What are the characteristis of HSCs

A

Pluripotent and self renewal capability

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

What factors are expressed on every early committed progenitor

A

Receptors for KIT ligand, FLT3 Ligand; EPO, CS and thrombopoietin act on committed progenitor cells with more restricted differentiation potentials

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

What is the morphology of bone marrow

A

Megakaryocytes lie next to sinusoids and extend cytoplasmic processes that busy off into bloodstream to produce platelets; red cell precursors surround macrophages (nurse cells) that provide iron for hemoglobin; if marrow architecture distorted can lead to leukoerythroblastosis (release of immature precursors into peripheral blood); in normal adults, fat:hematopoietic elements is 1:1; in plastic anemia, fat cells increase; in hemolytic anemia’s, fat is lost

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

What is the most common cause of lymphopenia

A

HIV, after therapy with glucocorticoids or cytotoxic drugs, autoimmune disorders, malnutrition, acute viral infections (lymphocyte redistribution in last case)

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

What can cause neutropenia

A
  • Inadequate or ineffective granulopoiesis: suppression o HSC (aplastic anemia, tumors, granulomatous dz - accompanied by anemia and thrombocytopenia), suppression of committed granulocytic precursors via drugs, megaloblastic anemia and myelodysplastic syndromes, congenital syndromes (kostmann)
  • increased destruction or sequestration of neutrophils in periphery: immune mediated injury (SLE), splenomegaly, increased peripheral utilization (overwhelming bacterial, fungal, or rickettsial infections)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

What is the most common cause of agranulocytosis

A

Drug toxicity: alkylating agents and antimetabolites in cancer treatment, aminopyrine, chloramphenicol, sulfonamides (ab mediated destruction of mature neutrophils), chlorpromazine (toxic to precursors), thiouracil, phenylbuazone

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

What is LGL leukemia

A

Can cause severe neutropenia; monoclonal proliferation’s of large granular lymphocytes

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

What is the morphology of the bone marrow in neutropenia

A

Vary with cause; with destruction of neutrophils in periphery, marrow is hypercellular (also with ineffective granlopoiesis - megaloblastic anemia’s and myelodysplasic syndromes); if destroy precursors - marrow hypocellularity

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

What infections are a result of agranulocytosis

A

Ulceration necrotizing lesions of gingiva, floor of mouth, Bucal mucosa, pharynx (agranulocytosis angina); covered by gray to green-black necrotic membranes; *high is for Candida and aspergillus; bacteria grow in colonies (botryomycosis)

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

What is the peripheral blood leukocyte count influenced by

A
  • size of myeloid and lymphoid precursor and storage cell poools in BM, thymus, circulation, and periphery
  • rate of release of cells from storage pools into circulation
  • proportion of cells that are adherent to bv walls (marginal pool)
  • rate of extravasation of cells from blood into tissue
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q

What are the mechanisms and causes of leukocytosis

A
  • increased production in marrow: chronic infection or inflammation (GF dependent), paraneoplastic (Hodgkin, GF dependent), myeloproliferative disorders(chronic myeloid leukemia, GF independent)
  • increased release from marrow stores: endotoxemia, infection, hypoxia
  • decreased margination: exercise, catecholamines
  • decreased extravasation into tissues: glucocorticoids
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q

What mediates the release of mature granulocytoes from BM during infection

A

TNF and IL-1; if prolonged, will stimulate macrophages, BM stromal cells, and T cells tp prude increased mounts of hematopoietic growth factors

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

What happens in sepsis or severe inflammatory disorders (Kawasaki)

A

Leukocytosis is accompanied by morphological changes in neutrophils ie: toxic granulation (axurophilic granules), Dohle bodies (patches of dilated ER that appear as sky-blue cytoplasmic puddles), cytoplasmic vacuoles

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

What are the causes of leukocytosis

A
  • neutrophilic leukocytosis: acute bacterial infections (pyogenic), sterile inflammation caused by tissue necrosis (burns, MI)
  • eosinophilic: allergic disorders, asthma, hay fever, parasite infections, drug reactions, malignancies (Hodgkin), AI (pemphigus, dermatitis herpetiformis), Vasculitis, atheroembolic dz
  • basophils: rare indicative of myeloproliferative dz (CML)
  • monocytosis: chronic infections (TB), bacterial endocarditis, rickettsiosis, malaria, AI (SLE), IBD
  • lymphocytosis: chronic immune stimulation (TB, brucellosis), viral infections (Hep A, CMV, EPV), bordatella pertussis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

What does acute nonspecific lymphadentitis of specific sites indicate

A
  • cervical: teeth or tonsils
  • axillary or inguinal: extremities
  • mesenteric: appendicitis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

What is follicular hyperplasia

A

Caused by stimuli that activate humoral immune responses; large ablong germinal centers surrounded by naive B cells (mantle zone); tingible-body macrophages ; causes: rheumatoid arthritis, toxoplasmosis, early stages of infection with HIV *features favoring reactive vs neoplastic hyperplasia: preservation of LN architecture (interfolliciular T cell zone and sinusoids), marked variation in shape and size of follicles, presence of frequent mitotic figures, phagocytic macrophages and recognizable light and dark zones

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

What is paracortical hyperplasia

A

Caused by a stimuli that triggers T cell mediated immune responses (acute viral infection) hypertrophy of sinusoidal and vascular endothelial cells

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

What are sinus histiocytosis

A

Aka reticular hyperplasia; increase in number and size of cells that line lymphatic sinusoids; prominent in LN draining cancers such as carcinoma of breast; but nonspecific

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

Are LN in chronic reactions tender

A

No; only acute; common in inguinal an axillary nodes

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

What is hemophagocytic lymphohistiocytosis

A

Reactive condition marked by cytopenias and signs and sx of systemic inflammation related to macrophage activation; referred to as macrophage activation syndrome; some forms are familial

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

What is the pathogensis of hemophagoytic lymphohistiocytosis

A

*systemic activation of macrophages and CD8 cytotoxic T cells; phagocytosis blood cell progenitors and formed elements in peripheral tissues; sx of systemic inflammation; shock-like picture and cytopenia

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

What are familial forms of HLH associated with

A

Several mutations; all impact ability of cytotoxic T cells and NK cells to properly form or deploy cytotoxic granules; high levels of IFN-gamma, TNFalpha, IL-6, IL-12, IL-2R

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

What is the most common trigger for HLH

A

Infection, most EBV

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

What are the clincial features of HLH

A

ACute febrile illness assoc with HSM; hemophagocytosis seen on BM exam but not required to make diagnosis nor is it sufficient; anemia, thrombocytopenia, very high levels of plasma ferritin and soluble IL-2R; elevated liver function test and triglyceride levels; can progress to death if untreated; treat its immunosuppressive drugs and mild chemo; even with treatment, those who survive have significant sequelae such as renal damage and growth aNd mental retardation in children

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

What are the types of malignancies of white cells

A
  • lymphoid neoplasms: B cell, Tcell, NK cell origin
  • myeloid neoplasms: arise from early HSC progenitors; acute myeloid leukemia (accumulate in BM), myelodysplastic syndromes (ineffective hematopoietic aNd peripheral blood cytopenia), chronic myeloproliferative disorders (increased production of terminally diff myeloid elements)
  • histiocytoses: uncommon; proliferative lesions of macrophages and dendritic cells; langerhans cell histiocytosis
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
30
Q

Where are dominant negative mutations often seen

A

Acute Leukemias; causes arrest in differentiation

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

What is the function of AID (activation induced cytosine delaminase)

A

Causes class switching and somatic hypermutation (increases ab affinity for antigen); sufficient to induce MYC/Ig translocations in Normal germinal center B cells

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

What individuals are at increased risk for acute leukemia

A

Bloom syndrome, fanconi anemia, ataxia telangiectasia, Down syndrome, type I NF

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

What are the lymphotrophic viruses

A

HTLV-1: adult T cell leukemia/lymphoma
EBV: Burkett, Hodgkin lymphoma, B cell lymphomas arising insetting of T cell immunodeficiency and NK cell lymphomas
HHV-8: B cell lymphoma that presents as malignant effusion, often in pleural cavity

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

What are examples of chronic inflammation that have risk of lymphoid neoplasia

A

H pylori (B cell lymphoma), gluten sensitive enteropathy (T cell lymphoma), breast implants (T cell lymphoma), HIV (B cell)

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

What iatrogenic factors put people at risk or lymphoid neoplasia

A

Radiation and certain forms of chemo

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

What is the effect of smoking on risk of lymphoid neoplasia

A

Increased risk of acute myeloid leukemia

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

What are features of plasma cell neoplasms

A

Most often arise in BM and only infrequently involve LN or peripheral blood

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

How do Hodgkin and NH lymphomas present

A

Nontener LN; some NHL present its sx related to involvement of exranodal sites

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

How do lymphocytic Leukemias often present

A

Signs and sx of suppression of normal hematopoiesis

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

How does multiple myeloma (plasma cell neoplasm) present

A

Bony destruction of skeleton; presents with pain

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

What are the categories of lymphoid neoplasms

A
  • precursor B cell: neoplasmas of immature B cells
  • peripheral B cell: neoplasms of mature B cells
  • precursor T cell
  • peripheral T cell and NK cell
  • Hodgkin lymphoma: neoplasms of reed-stern berg cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
42
Q

What are the precursor B cell neoplasms

A

B cell acute lymphoblastic leukemia/lymphoma

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

What are the peripheral B cell neoplasms

A

Chronic lymphocytic leukemia/small lymphocytic lymphoma, B cell prolymphocytic leukemia, lymphoplasmacytic lymphoma, splenic and nodal marginal zone lymphomas, extranodal marginal zone lymphoma, mantle cell lymphoma, follicular lymphoma, marginal zone lymphoma, hairy cell leukemia, plasacytoma/plasma cell myeloma, diffuse large B cell lymphoma, Burkitt lymphoma

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

What are the precursor T cell neoplasms

A

T cell acute lymphoblastic leukemia/lymphoma

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

What are the peripheral T cell and NK cell neoplasms

A

T cell prolymphocytic leukemia, large granular lymphocytic leukemia, MYC os is fungoides/sezary syndrome, peripheral T cell lymphoma, anaplastic large cell lymphoma, angioimmunoblastic T cell lymphoma, enteropathy associated T cell lymphoma, paniculitis-like T cell lymphoma, hepatosplenic T cell lymphoma, adult T cell leukemia/lymphoma, extranodal NK/T cel lymphoma, NK cell leukemia

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

What are the Hodgkin lymphoma subtypes

A

Nodular sclerosis, mixed cellularity, lymphocyte rich, lymphocyte depletion

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

What is significant about the antigen receptor of neoplasms

A

They will all be the same

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

Which lymphoid neoplasias are not always disseminated at time of diagnosis

A

Hodgkin and marginal zone B cell lymphomas

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

What are the the CD markers associated with primarily T cell associated lymphoid neoplasia

A
  • CD1: thymocytes and langherans
  • CD3: thymoytes and mature T cells
  • CD4: helper T cells, subset of thymocytes
  • CD5: T cells and small subset of B cells
  • CD8: cytotoxic T cells, subset of thymocytes nad some NK cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
50
Q

What are the CD markers that re primarily B cell associated

A
  • CD10: pre-Bcells and germinal center B cells
  • CD19: pre-B cells and mature B cells but not plasma cells
  • CD20: pre B cells after CD19 and mature B cells but not plasma cells
  • CD21: EBV receptor; mature B cells and follicular dendritic cells
  • CD23: activated mature B cells
  • CD79a: marrow pre-Bcells and mature bcells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
51
Q

What are the primarily monocyte or macrophage associated CD markers

A
  • CD11c: granulocytes, monocytes, macrophages; hairy cell Leukemias
  • CD13: immature and mature monocytes and granulocytes
  • CD14: monocytes
  • CD15: granulocytes, reed-sternberg cells
  • CD33: myeloid progenitors and monocytes
  • CD64: mature myeloid cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
52
Q

What are the primarily NK cell associated CD markers

A
  • CD16: NK cells and granulocytes

- CD56: NK cells and subset of T cells

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

What are the primarily stem cell and progenitor cell associated CD markers

A

CD34: pluripotent HSC and progenitor cells of many lineages

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

What re the activation CD markers

A

CD30: activated B, T cells and monocytes; Reed-Sternberg cells

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

What is the CD present on all leukocytes

A

CD45

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

What is the diff in Hodgkin and NHL in terms of spread

A

Hodgkin spread in orderly fashion; NHL spreads widely early in course and less predictable

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

What is acute lymphoblastic leukemia/lymphoma

A

Neoplasms composed of immature B or T cells (lymphoblasts); B-ALLs manifest in childhood as leukemia; T-ALLs manifest in adolescent males as thymic lymphomas; Hispanics have highest incidence; peaks in incidence at 3 for B-ALL; T-ALL peaks in adolescence

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

What is the pathogensis of ALL

A

Chrom ab that dysregulate the expression and function of TF required for normal B and T cell development; T-ALLs have GOF in NOTCH1; B-ALLs have LOF in PAX5, E2A, EBF or balanced 12;21 translocation involving ETV6-RUNX1; promote arrest and increased self renewal; hyperdiploidy and hypo diploids only seen in B-ALL

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

What is the cell of origin for burkitt lymphoma

A

Germinal center B cell; translocations involving MYC-Ig 8;14 - subset is EBV related; clin presentation: adolescents or young adults with extranodal masses; aggressive

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

What is the cell of origin of diffuse large B cell lymphoma

A

Germinal center or postgerminal center B cell; rearragnemts o BCL6, BCL or MYC; presents at all ages but more common in adults; appears as rapidly growing mass; aggressive

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

What is the cell of origin of extranodal marginal zone lymphoma

A

Memory B cell; translocations: 11;18, 1;14, 14;18 - MALT1-IAP2, BCL10-IgH, MALT-IgH union genes; arises at extranodal sites in adults with chronic inflammatory dz; indolent

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

What is the cell of origin for follicular lymphoma

A

Germinal center B cell; translocation: 14;18 - BCL2-IgH; presents in older adults with generalized LAD and marrow involvement; indolent

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

What is the cell of origin of hairy cell leukemia

A

Memory B cell; activating BRAF mutation; seen in older males with pancytopenia and splenomegaly; indolent

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
64
Q

What is the cell of origin of mantle cell lymphoma

A

Naive B cell; 11;14 translocation - cylinD-IgH fusion; older males with disseminated dz; moderately aggressive

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

What is the cell of origin of multiple myeloma/solitary plasmacytoma

A

Post germinal colter bone marrow homing plasma cell; diverse rearragnemtns involving IgH; 13q deletions; myeloma seen in older adults with lytic bone lesions, hypercalcemia, and renal failure; moderately aggresive; plasmacytoma - see isolated plasma cell masses in bone or soft tissue; indolent

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

What is the cell of origin for small lymphocytic lymphoma/ chronic lymphocytic leukemia

A

Naive B cell or memory B cell; trisomy 21, deletions of 11q, 13 and 17p; seen in older adults with BM, LN, spleen and liver dz; AI hemolysis and thrombocytopenia; indolent

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

What is the cell of origin of adult T cell leukemia/lyphoma

A

Helper T cell; HTLV-1 povirus present in tumor cells; presents in adults with cutaneous lesions, marrow involvement and hypercalcmia; mainly in Japan, west Africa and Caribbean; aggressive

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

What is the cell of origin of peripheral T cell lymphoma

A

Helper or cytotoxic T cell; no specific chrom ab; seen in older adults with LAD; aggresive

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

What is the cell of origin of anaplastic large cell lymphoma

A

Cytotoxic T cell; rearrangements o ALK (anaplastic large cell lymphoma kinase); seen in children and young adults with LN and soft tissue dz; aggresive

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

What is the cell of origin of extranodal NK/Tcell lymphoma

A

NK cell (common) or cytotoxic T cell (rare); EBV associated; seen in adults with destructive extranodal masses; most commonly sinonasal; aggressive

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

What is the cell of origin of mycosis fungoides/sezary syndrome

A

Helper Tcell; no chrom ab; seen in adult patients with cutaneous patches, plaques, nodules or generalized erythema; indolent

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

What is the cell of origin of large granular lymphocytic leukemia

A

2 types: cytotoxic T cell and NK cell; point mutations in STAT3; seen in adult patients with splenomegaly, neutropenia and anemia;

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

What is the morphology of ALL

A

In leukemia presentation: marrow is hypercellular and packed with lymphoblasts; mediastinal thymic masses occur in most T-ALLs and more likely to be assoc with LAD and splenomegaly; scant basophils cytoplasm; starry sky appearance; compared to myeloblasts, lymphoblasts have more condensed chromatin, less conspicuous nucleoli and smaller amounts of cytoplasm that usually lacks granules *also lymphoblasts are myeloperoxisdase neagtive

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

What staining is positive in ALL

A

Immunostaining for terminal deoxynucleotidyl transferase (TdT)
B-ALL: CD19 and PAX5; CD10; in very mature B-ALL, CD10 is negative; IgM in late pre-B ALLs
T-ALL: CD1, CD2, CD5, CD7; more mature CD3, CD4, CD8

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

What are the clinical features of ALL

A
  • abrupt stormy onset within days to weeks of first sx
  • sx related to depression of BM function; fatigue due to anemia ,fever (neutropenia), bleeding (thrombocytopenia)
  • mass effects: bone pain resulting from marrow expansion and infiltration of subostium, generalized LAD, HSM, testicular enlargement, in T-ALL - compression of vessels and airways in mediastinum
  • CNS manifestations such as HA, vomiting and nerve palsies resulting from meningeal spread (more common in ALL than AML)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
76
Q

What factors are assoc with a worse prognosis in ALL

A

Age <2 yrs (b/c involve translocations involving MLL gene), presentation in adolescence or adulthood, peripheral blood blast counts >100,000 (high tumor burden)

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

What are good prognostic factors in ALL

A

Age btw 2-10, low white cell count, hyperdiploidy, trisomy of chromosomes 4,7,10, presents of 12;21

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

What dos the 9;22 translocation form of ALL do

A

Creates fusion gene that encodes an active BCR-ABL tyrosine kinase; treatment with BCR-ABL kinase inhibitors in combo with chemo is effective

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

What is the most common leukemia in adults in the western world

A

chronic lymphocytic leukemia; median age: 60; more common in males

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

What is the pathogensis of CLL/SLL

A

Most common anomaly is deletion of 13q14.3, 11q, 17p, and trisomy 12q; tumors with unmutated Ig segments (naive B cell origin) have a more aggresive course;GOF in notch1 receptor; tour cells rely on signals enervated b B ell receptor transducer by BTK (defective in x linked BTK)

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

What is the morphology of CLL/SLL

A

LN diffusely effaced by infiltrated of small lymphocytes; proliferation centers - pathognomonic for CLL/SLL; blood contains large numbers of small round lymphocytes with scant cytoplasm; smudge cells; BM almost always involved by interstitial infiltrates or aggregates of tumor; also seen in splenic white and red plump and hepatic portal tracts

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

What is the immunophenotype of SLL/CLL

A

Pan Bcell markers; CD19 and CD20; CD23, CD5

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

What is the clinical presentation of CLL/SLL

A

Asymptomatic at diagnosis; when sx, nonspecific - fatiguability, weight loss, anorexia; generalized LAD, HSM; leukopenia or Leukocytosis; disrupts normal immune function hypogammaglobulinemia hemolytic anemia or thrombocytopenia due to abs produced by NON-neoplastic B cells

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

What correlates with a worse outcome of CLL/SLL

A

Presence of deletions of 11q and 17p; lack of somatic hypermutation, expression of ZAP-70, presence of NOTCH1 mutation

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

How is CLL/SLL treated

A

Gentle chemo and immunotherapy with abs against CD20

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

What factors impacts patient survival in CLL/SLL

A

It can transform to more aggresive tumor; most commonly in form of diffuse large B cell lymphoma (richter syndrome) - rapidly enlarging mass in LN or spleen; not good survival

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

What is the most common form of indolent NHL in the US

A

Follicular lymphoma; presents in middle age and affects both genders equally; strongly assoc with chrom translocations involving BCL2 14;18; devoid of apoptosis cells; mutations in MLL2 (encodes histone methyltranserase that regulations gene expression - epigenetics impt) response to therapy influenced by surrounding microenvironment

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

What is the morphology of follicular lymphoma

A

Predominantly nodular or diffuse pattern in involved LN; 2 cell types present: small cells with irregular or cleaved nuclear contours (centroytes) and larger cells with open nuclear chromatin, several nucleoli (centroblasts); BM involved in most cases - paratrabecular lymphoid aggregates* splenic white pulp and hepatic portal triads frequently involved

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

What is the immunophenotype of follicular lymphoma

A

CD19, CD20, CD10, surface Ig, BCl6; CD5 NOT expressed; BCL2 in most cases

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

What are the clinical features of follicular lymphoma

A

Painless generalized LAD; involvement of extranodal sites is uncommon; incurable, but follows indolent waxing and waning course; survival not improved by aggressive therapy; palliative care with low dose chemo or immunotherapy when become sx; histo transformation occurs in half of cases most commonly to diffuse B cell lymphoma; can also transform to burkitt lymphoma; not long survival after transformation

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

What is the most common form of NHL

A

Diffuse large B cell lyphoma; slight male predominance; median age 60 but can occur in young adults and kids

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

What is the pathogenesis of DLBCL

A

Dysregulation of BCL6 (required for formation of normal germinal centers) - product of somatic hypermutation that results in overexpression of BCL6; BCL6 normally represses expression of factors t hat promote germinal center B cell diff, growth arrest, and apoptosis; some assoc with 14;18 translocation; mutations in p300 (histone acetylase) and CREBP

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

What is the immunophenotype of DLBCL

A

CD19 CD20; variable expression of CD10 and BCL6

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
94
Q

What are the subtypes of DLBCL

A
  • immunodeficiency associated: occurs in setting of severe T cell immunodeficiency (HIV and allogeneic BM transplant); usually infected with EBV restoration of T cell immunity may lead to regression
  • primary effusion lymphoma: malignant pleural or ascitic effusion mostly in patients with advanced HIV or older adults; tumor cells are anaplastic and fail to express surface B or T cell markers but have long as IgH gene arrangements; in all cases* tumor cells are infected with HHV-8
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
95
Q

What are the clinical features of DLBCL

A

Presents as rapidly enlarging mass at nodal or extranodal site; can arise anywhere; waldeyer ring (oropharyngeal lymphoid tissue) commonly involved; extranodal sites include GI, skin, bone, brain; BM involvement uncommon; aggressive and rapidly fatal without treatment; with chemo good prognosis; anti-CD20 improves outcome; if have a MYC translocation, worse prognosis

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

What are the categories of burkitt lymphoma

A

African (endemic) burkitt lymphoma; sporadic (nonendemic) burkitt lymphoma, and subset of aggresive lymphomas occuring in individuals with HIV; histologically identical but differ in clinical, genotype, and virologic characteristics

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

What is the pathogenesis of burkitt lymphoma

A

All forms assoc with translocations of MYC on chrom 8 that leads to increased MYC protein levels; Warburg effect - increases genes that are required for aerobic glycolysis *fastest growing human tumor; 8;14 translocation (IgH locus) or 2;8 (Igk) or 8;22 (gamma light chain); breakpoint in sporadic in class switch regions; in endemic in VDJ sequences

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
98
Q

What are all endemic burkitt lymphomas infected with

A

latent EBV; also present in 1/4 of HIV assoc tumors and some sporadic cases

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

what is the morphology of burkitt lymphoma

A

Diffuse infiltrate of intermediate size lymphoid cells; coarse chromatin, several nucleoli; high mitotic index and numerous apoptotic cells - phagocytosed by macrophages which creates starry sky appearance; in BM, royal blue cytoplasm containing clear cytoplasmic vacuoles

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

What is the immunophenotype of burkitt

A

IgM, CD19, CD20, CD10, BCL6; does not express BCL2 (unlike other tumors of germinal center origin)

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

What are the clinical features of burkitt lymphoma

A

Both endemic and sporadic found mainly in children; most manifest as tumors in extranodal sites; endemic usually presents as a mass involving mandible and abdominal viscera (kidneys, ovaries, and adrenal glands); sporadic appears as mass involving ileocecum and peritoneum; involvement of BM and peripheral blood uncommon; very aggressive but responds well to intense chemo

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

What are the plasma cell neoplasms referred to as

A

Dyscrasias; most common and deadly is multiple myeloma; identified by monoclonal Ig in blood (M component); neoplastic plasma cells can synthesize excess light chains and complete Igs; light chains can be excreted in urine (bence-jones protein)

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

what are the terms used to describe abnormal Igs in plasma cell neoplasms

A

Monoclonal gammapathy, dysproteinemia, paraproteinemia

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

What pathologies are associated with abnormal igs

A
  • multiple myeloma (plasma cell myeloma): presents as tumorous masses scattered throughout skeletal system; solitary myeloma (plasmacytoma) is variant that presents as single mass in bone or soft tissue; smoldering myeloma is variant with lack of sx and a high plasma M component
  • waldenstrom macroglobulinemia: high levels of IgM lead tot sx related to hyperviscosity of blood; older adults in association with lymphoplasmacytic lymphoma
  • heavy chain dz: monoclonal gammopaty seen in assoc with lymphoplasmacytic lymphoma and small bowel marginal zone lymphoma (malnourished - Mediterranean lyymphoma); synthesis of free heavy chain
  • primary or immunocyte assoc amyloidosis: monoclonal proliferation of plasma cells secreting light chains (usually gamma) that are deposited as amyloid
  • monoclonal gammopathy of undetermined significance: patients w/o sx who have small to moderately large M components in blood; common in older adults and low, but constant rate of transformation to sx monoclonal gammopathies, mostly multiple myeloma
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
105
Q

What is multiple myeloma

A

Plasma cell neoplasm assoc with lytic bone lesions, hypercalcemia, renal failure, and acquired immune abnormalities; can spread in late course to LN and extranodal sites; higher in men and Africans; peak incidence age 65

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

What is the pathogenesis of multiple myeloma

A

Assoc with frequent rearrangements involving IgH locus with cyclinD1 and D3; deletions on chrom 17p that involves TP53 indicate poor outcome; frequent mutationis of NFkB; survival of tumor dependent on IL-6* - high levels seen in active dz and assoc with poor prognosis; MIP1alpha up regulates express ion of receptor activator of NFkB (RANKL) which activates osteoclasts (increased bone resorption) - hypercalcemia and breakdown of bone

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

what is the morphology of multiple myeloma

A

Destructive plasma cell tumors involving axial skeleton; most effected: vertebral column, ribs, skull, pelvis, femur, clavicle, scapula; lesions begin in medullary cavity and erode callous bone, destroy the bony cortex; bone lesions appear as punched out defects on radiograph and consist of soft gelatinous red tumor masses; some variants can have flame cells with fiery red cytoplasm, Mott cells with multiple grasp like cytoplasmic droplets; globular inclusions (Russell bodies - if cytoplasmic) (duthcer bodies if nuclear) *M protein causes rouleaux formation

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

what is the immunophenotype of multiple myeloma

A

CD138 (adhesion molecule aka syndecan-1), often express CD56

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

what are the clinical features of multiple myeloma

A

Pathologic fractures and chronic pain; hypercalcemia - neuro sx, weakness, lethargy, constipation, polyuria; recurrent bacterial infxn; renal insufficiency - trails infections as COD; can cause amyloidosis if AL type; most common monoclonal lgis IgG

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

What does definitive diagnosis of multiple myeloma rely on

A

Bone marrow exam; often gives rise to normochomic anemia

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

What is assoc with a good outcome of multiple myeloma

A

Translocations involving cyclin D1; deletions of 13q, 17p, ad 4;14 translocation have more aggressive course

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

What is being explored s a treatment for multiple myeloma

A

Proteasome inhibitors; thalidomide also has activity against myeloma

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

What is solitary myeloma (plasmacytoma)

A

Solitary lesion of bone o soft tissue; extraosseous lesions found in lungs, oronasopharynx or nasal sinuses; progresses to multiple myeloma but can take 10 years; extraosseus can be cured by local resection

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

What is smoldering myeloma

A

Middle ground btw multiple myeloma and monoclonal gammopathy; asymptomatic; about 75% progress to multiple myeloma in 15 years

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

What is monoclonal gammopathy of uncertain significance

A

Most common plasma cell dyscrasia; asymptomatic Nd serum M protein <3gm/dL; 1% envelop symptomatic plasma cell neoplasm (multiple myeloma)

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

What is lymphoplasmacytic lymphoma

A

B cell neoplasm of older adults that usually presents in 6th or 7th decade; substantial reaction of tumor cells undergo. Terminal differentiation to plasma cells; secretes IgM -> hyperviscosity syndrome known as waldenstrom macroglobulinemia; does not have complications of renal and amyloidosis like multiple myeloma

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

What is the pathogensis of lymphoplasmacytic lymphoma

A

All have mutations in MYD88 (encodes adaptor protein for signaling evens that activate NFkB and augment signals downstream of Bcell receptor

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

What is the morphology of lymphoplasmacytic lymphoma

A

Marrow contains infiltrate of lymphocytes, plasma cells, and plasmacytoma lymphomcytes accompanied by mast cell hyperplasia; Russell bodies or dutcher bodies; at diagnosis usually disseminated to LN, spleen, and liver; infiltration of n roots, meninges and brain can also occur

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

What is the immuophenotype of lymphoplasmacytic lymphoma

A

CD20 and surface Ig; secrets IgM or IgG or IgA

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

What are the clinical features of lymphoplasmacytic lymphoma

A

Nonspecific complaints; half have LAD, HSM; anemia is common; autoimmune hemolysis caused by gold glutinous (IgM binds to red cells at temp <37 degrees)

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

What symptoms do patients with IgM secreting tumors

A

Hyperviscosity syndrome: visual impairment (venous congestion, totuosity and distention of retinal veins, retinal hemorrhages and exudates), neuro problems (HA, dizziness, deafness, stupor from sluggish blood flow), bleeding (formation of complexes btw macroglobulinemia and clotting factors), cryoglobulinemia (precipitation of macroglobulinemia at low temps - raynauds and cold urticaria)

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

Is lymphoplasmacytic lymphoma curable

A

No; symptoms alleviated by plasmapharesis; median survival is 4 years

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

What is extramedullary plasmacytoma

A

Solitary mass usually in aero digestive tract; rarely progresses to systemic dz

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

What is mantle cell lymphoma

A

Uncommon; presents in 5ht-6th decade and shows male predominance

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

What is the pathogenesis of mantle cell lymphoma

A

11;14 translocation involving IgH locus and cylin D1

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

What is the morphology of mantle cell lymphoma

A

Generalized LAD; 40% have peripheral blood involvement; extranodal involvement: bon marrow, spleen liver and gut; can produce polyp like lesions - most likely to spread in this fashion of all NHL; nodular appearance; proflieration of small lymphocytes with irregular to deeply clefted nuclear contours

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
127
Q

What is the immunophenotype of mantle cell lymphoma

A

cyclin D1; CD19, CD20 and high levels of IgM and D; usually CD5+ and CD23 - (distinguishes it from CLL/SLL); lacks somatic hypermutation

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

What are the clinical features of mantle cell lymphoma

A

Painless LAD; sx related to gut and spleen common; prognosis is poor* blastoid variant and proliferative expression assoc with even shorter survival

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

What are marginal zone lymphomas

A

Arise within LN, spleen or extranodal tissues; often referred to as MALTomas; somatic hypermutation - memory B cell origin

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

What are the 3 exceptional characteristics of marginal zone lymphomas

A
  • often arise within tissue involved by chronic inflammatory disorders; ie: sjogren, hashimotos, h pylori
  • remain localized for prolonged periods spreading only late in course
  • may regress if inciting agent is eradicated
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
131
Q

What translocations are specific for mantle zone lymphomas

A

11;18 14;18 1;14 - all unregulated function of BCL10 or MALT1 which activate NFkB; with further Clonal evolution, spread to distant sites and transformation to DLBCL can occur

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

What is hairy cell leukemia

A

Disease of middle aged white males; median age of 55

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

What is the pathogensis of hairy cell leukemia

A

Assoc with activating point mutations in BRAF (serine/threonine kinase) - downstream of RAS in MAPK cascade; mutation is valine to glutamate substitution (also sen in melanoma and langerhans cell histiocytosis)

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

What is the morphology of hairy cell leukemia

A

Fine hairlike projections; on blood smear has round, oblong nuclei and pale blue cytoplasm with bleblike projection; cannot be aspirated - “dry tap” and are only seen in marrow biopsies; splenic red pulp usually heavily infiltrated leading to obliteration of white pulp and beefy red. Gross appearance; hepatic portal triad involved frequently

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

What is the immunonphenotype of hair cell leukemia

A

CD19 CD20, surface Ig (IgG), CD11c, CD25, CD103, annexin A1

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

What are the clinical features of hairy cell leukemia

A

Result from infiltration of BM, liver, spleen; massive splenomegly*, hepatomegaly not as common; LAD is rare; pancytopenia, infections - risk of atypical mycobacterial infections; indolent course; sensitive to gentle chemo; relapse in 5 or more years; BRAF inhibitors produce excellent response; excellent prognosis

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

What is peripheral T cell lymphoma unspecified

A

Face LN diffusely and are typically composed of pleomorphic mixture of malignant T cells; prominent infiltrate of reactive cells; express CD2, CD3, CD5, and either alphabeta or deltagamma T cell receptor; most present with LAD, eosinophilia, pruritis ever and wight loss; worse prognosis than mature B cell neoplasms

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

What is anaplastic large cell lymphoma (ALK positive)

A

Uncommon; presence of rearrangements of ALK gene on chrom 2p23; break ALK locus and lead to ALK fusion proteins - trigger JAK/STAT signaling; horseshoe shaped nuclei and voluminous cytoplasm (hallmark cells) clusters around venules and infiltrates lymphoid sinuses mimicking met carcinoma *ALK is not present in normal lymphocytes; occurs in children or young cults and involves soft tissue and have good prognosis; if no ALK rearrangement (adults) poor prognosis; both express CD30

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

What is adult T cell leukemia/lymphoma

A

Neoplasm of CD4 cells only observed in adults infected with HTLV-1; skin lesions, generalized LAD, HSM, peripheral blood lymphocytosis and hypercalcemia; cloverleaf or flower cells; HTLV-1 encodes Tax that is an activator of NFkB; most p resent with rapidly progressive dz that is fatal within months to 1 year

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
140
Q

What else can HTLV-1 infection lead to

A

Progressive Demyelinating dz of CNS and SC

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

What is mycosis fungoides

A

Different manifestation of a tumor of CD4 helper T cells that home to the skin; progresses through premycotic phase, plaque phase and tumor phase; epidermis and dermis infiltrated by neoplastic T cells which have cerebriform appearance

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

what is sezary syndrome

A

Variant in which skin involvement is manifested as generalized exfoliative erythroderma; in contrast to mycosis fungoides, skin lesions rarely proceed to tumefaction; associated leukemia of sezary cells with charactestic cerebriform nuclei

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

What is the immunophenotype of s early syndrome and mycosis fungoides

A

adhesion molecule cutaneous leukocyte antigen (CLA) and chemokine receptors CCR4 and CCR10; indolent tumors

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

What is large granular lymphocytic leukemia

A

T and NK cel variants; adults; in T cell dz (CD3 positive) -> lymphocytosis and splenomegaly; NK dz (CD3 negative, CD56) more subtle; mutations in STAT3; neutropenia and anemia ; T cell indolent; NK more aggressive

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

What is felty syndrome

A

Complication of large granular lymphocytic leukemia; triad of RA, splenomegaly, and neutropenia

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

What is extranodal NK/T cell lymphoma

A

Rare in US; Asia; presents as destructive nasopharyngeal mass; surrounds and invades small vessels leading to extensive ischemic necrosis; large azurophilic granules seen in cytoplasm; assoc with EBV but tumor cells do not express CD21; CD3 -; highly aggresive but respond well to radiation, not hemo; prognosis is poor

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

What is the presentation of Hodgkin lymphoma

A

Arises in single node or chain of nodes and spreads first to anatomically continuous lymphoid tissues; reed stern berg cells (neoplastic giant cells) derived from germ center or post germ center B cells

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

What are the subtypes of Hodgkin lyphoma

A

Nodular sclerosis, mixed cellularity, lymphocyte rich, lymphocyte depletion, lymphocyte predominance

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

What are the classical forms of Hodgkin

A

Nodular sclerosis, mixed cellularity, lymphocyte rich and lymphocyte depletion; have similar immunophenyote.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
150
Q

What is the pathogenesis of Hodgkin

A

Ig genes of reed sternberg cells have undergone VDJ recombination and somatic hypermutation; don’t express B cell specific genes; activation of NFkB

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

How can NFkB b activated in Hodgkin

A
  • EBV infection
  • EBV +tumor cells express latent membrane proline 1 (LMP-1) encoded by EBV genome that upregulates NFkB
  • result of acquired LOF in IkB or A20 (TNF alpha induced protein 3 or TNFAIP3)
  • rescues crippled germ center B cells that cannot express Igs from apoptosis
  • gains in REL Proto-oncogene
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
152
Q

What is the morphology of Hodgkin

A

Reed-Sternberg cells are large with multiple nuclei or single nucleus with multiple lobes; mononuclear variants; lacunae cells seen in nodular sclerosis type have folded or multilobate nuclei

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

What conditions can cells that resemble reed sternberg cells be seen

A

Mono, solid tissue cancers, and large cell NHL

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

What does the diagnosis of Hodgkin lymphoma depend on

A

Reed sternberg cells in background of non-neoplastic inflammatory cells

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

What is the morphology of the nodular sclerosis type of Hodgkin

A

Most common* lacunae variant Reed-Sternberg cells; deposition of collagen in bands that divide involved LN into circumscribed nodules *PAX5, CD15 and CD30 positive, negative for other B cell markers, T cell markers, and CD45; not assoc with EBV; equal in males and females; *involves lower cervical, supraclavicular and mediastinal LN; excellent prognosis

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

What is the morphology of mixed cellularity type Hodgkin

A

LN usually diffusely effaced by infiltrate; *diagnostic reed sternberg cells usually plentiful - most are infected with EBV; immunophenotype same as nodular sclerosis; more common in males, older age, and systemic sx; very good prognosis

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

What is the morphology lymphocyte-rich type Hodgkin

A

Uncommon; reactive lymphocytes make up majority of infiltrate; LN diffusely effaced; *presence of mononuclear variants and diagnostic Reed-Sternberg cells; excellent prognosis

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

What is the morphology of the lymphocyte depletion type of Hodgkin

A

Least common form*; abundance of reed sternberg cells; *most infected with EBV; occurs in old adults, in HIV+ ppl of any age and in non industrialized countries; less favorable outcome

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

What is the morphology of the lymphocyte predominance type of Hodgkin

A

Uncommon; effaced by nodular infilatred of small lymphocytes with variable number of macrophages; reed-sternberg hard to find contains lymphocytic and histioctic variants which have multilobed nucleus resembling popcorn kernel* - express B cell markers typical of germinal center B cells (CD20, BCL6, neg for CD15 and CD30); can transform to DLBCL; not assoc with EBV; males <35, cervical or axillary LAD; excellent prognosisi

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
160
Q

What are the clinical features of Hodgkin

A

Presents as painless LAD; patients with nodular sclerosis or lymphocyte predominance tend to have stage I-II dz and free of systemic sx; cutaneous immune unresponsiveness (anergy) also seen; factors released from Reed-Sternberg cells suppress Th1 immune responses

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
161
Q

How does Hodgkin spread

A

Nodal dz first -> splenic dz, hepatic dz and then marrow and other tissues; staging involves PE, radiologic imaging of ab, pelvis, chest and bx of BM; tumor stage rather than histo subtype is most impt for prognosis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
162
Q

How can you cure low stage localized Hodgkin

A

Field radiotherapy; but risks lung cancer, melanoma, an breast cancer; patients treated with early chemo regimens containing alkylating agents had high incidence of secondary tumors (AML)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
163
Q

What factors do Reed-Sternberg cells release

A

TNF, bFGF (leads to fibrosis); IL-5, galectin-1 (enhances Treg response), inhibits Th1 and CTL with IL-10; M-CSF, IL-13 which binds to a receptor on its membrane

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
164
Q

What is the common feature among myeloid neoplasms

A

Origin from hemtopoietic progenitor cells; present with sx related to altered hematopoiesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
165
Q

What are the categories of myeloid neoplasia

A
  • acute myeloid Leukemias: accumulation of immature myeloid forms (blasts) in BM and suppresses normal hematopoiesis
  • myelodysplastic syndromes: defective maturation of myeloid progenitors gives rise to ineffective hematopoiesis leading to cytopenias
  • myeloproliferative disorders: increased production of. One or more types of blood cells
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
166
Q

What are the manifestations of myeloid neoplasms influenced by

A
  • position of transformed cell within the hierarchy of progenitors
  • effect of transforming events on differentiation
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
167
Q

What is acute myeloid leukemia

A

Tumor of hematopoietic progenitors caused by acquired oncogene culture mutations that impede differentiation leading to accumulation of immature myeloid blasts in marrow; leads to marrow failure and anemia, thrombocytopenia and neutropenia; occurs at all ages but rises throughout life (peak in 60s)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
168
Q

What is the classification of AML

A

Heterogenous; 4 categories: AML with genetic aberrations, AML with MDS-like features, AML therapy related and AML NOS

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
169
Q

What are the AML with genetic aberrations

A
  • AML w/ (8;21)(q22;q22) RUNX1/ETO fusion: favorable prognosis; full range of myelocytic maturation; auger rods easily found; abnormal cytoplasmic granules
  • AML w/ inv(16)(p13;q22); CBFB/MYH1 fusion: favorable prognosis; myelocytic and monocytes diff; abnormal eosinophilic precursors with ab basophilic granules
  • AML w/ t(15;17)(q22;11-12); RARA/PML fusion: favorable; numerous Auer rods in bundles primary granules prominent; high incidence of DIC*
  • AML w/ t(11q23;v) diverse MLL fusion: poor, monocyte diff
  • AML w normal cytogenetic and mutated NPM: favorable; detected by immuno histo staining for NPM
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
170
Q

What are the AML with MDS like features

A
  • with prior MDS: poor prognosis; diagnosis based on clin history
  • AML with multilineage dysplasia: poor; maturing cells with dysplastic features typical of MDS
  • AML with MDS-like cytogenetic aberrations: poor assoc with 5q, 7q, 20q aberrations
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
171
Q

What is AML therapy related

A

Very poor prognosis; if following alkylation or radiation therapy, 2-8 year latency period; MDS like cytogenic aberrations; if following topoisomerase II inhibitor (etoposide) 1-3 year latency, translocations involving MLL

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
172
Q

What is AML NOS

A
  • AML, min diff: intermediated prognosis; neg for myeloperoxidase, myeloid antigens detected on blasts by flow cytomegalovirus
  • AML without maturation: intermediate; >3% of blasts pos for myeloperoxidase
  • AML with myelocytic maturation: intermediate; full range of myelocytic maturation
  • AML with myelomonocytic maturation: intermediate; myelocytic and monocyte diff
  • AML with monocytes maturation: intermediate; nonspecific esterase positive monoblasts and pro monocytes predominate in marrow
  • AML with erythroid maturation:: intermediate; erythroid/myeloid subtype >50% dysplastic maturing erythroid precursors and >20% myeloblasts; pure erythroid subtype defined by >80% erythroid precursors without myeloblasts
  • AML with megakarytocyte maturation: intermediate; blasts of megakaryocytic lineage; detected with abs against megakaryocyte markers (GPIIb/IIIa or vWF); assoc with marrow fibrosis; *most common AML in Down syndrome
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
173
Q

What are the most common rearrangements seen in AML

A

Genetic aberrations disrupt genes encoding TF required for normal myeloid diff; *2 most common are t(8;21) and inv(16) - disrupte RUNX and CBFB genes respectively which encode polypeptide that bind each other to form RUNX1-CBF1beta required for normal hematopoiesis

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
174
Q

What growth factor signaling pathways are mutated in AML

A

Ex: AML with t(15;17) - acute promyelocytic leukemia; creates retinoids acid receptor fusion with PML which interferes with terminal diff of granulocytes - can be overcome with all-trans retinoids acid or arsenic trioxide; but this is not enough to cause dz; also have mutations in FLT3 (RTK)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
175
Q

What epigenomic mutations have been seen in AML

A

Genes that influence DNA methylation or histone modifications; mutations of cohesion complex (proteins that regulate 3D structure of chromatin)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
176
Q

What is the diagnosis of AML based on

A

Presence of at least 20% myeloid blasts in the BM; myeloblasts have delicate nuclear chromatin, 2-4 nucleoli and lots of cytoplasm containing peroxidase pos granules; auer rods - distinct needle like azurophilic granules (acute promyelocytic leukemia is most prominent), monoblast: folded or lobulated nuclei, lack serum rods

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
177
Q

What is aleukemic leukemia

A

When blasts are entirely absent from blood in AML

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
178
Q

What marker do myeloid blasts express

A

CD34 (marker of pluripotency) and CD33 (marker of immature cells) but not CD64

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
179
Q

What are AMLs arising de novo in younger adults assoc with

A

Balanced chromosomal translocations particularly t(8;21), inv(16) and t(15;17)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
180
Q

What are AMLs following myelodysplastic syndromes or exposure to DNA damaging agents assoc with

A

Deletions or monosomies invovling chrom 5 and 7; lack translocations *exception is those that arise after treatment with topoisomerase II inhibitors - assoc with translocations involving MLL gene on chrom 11q23

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
181
Q

What are the clinical features of AML

A

Present within weeks or months of onset of sx with complaints related to anemia, neutropenia and thrombocytopenia (fatigue, fear and spontaneous mucosal and cutaneous bleeding); infections in oral cavity, skin, bladder, kidneys, colon usually caused by fungi, pseudomonas and commensals; tumors with monocytes diff infiltrate skin(leukemia cutis) and gingiva; CNS spread less common than ALL; can present as localized soft tissue mass known as myeloblastoma, granuloccytic sarcoma or chloroma

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
182
Q

What is the prognosis of AML

A

Difficult to treat; *AML with 15;17 treated with all-trans retinoids acid and arsenic salts has best prognosis; AMLs with 8;21 or inv(16) have good prognosis with chemo especially in absence of KIT mutations; prognosis is dismal for AMLs that follow MDS or genotoxic therapy to that occur in older adults (treat with hematopoietic stem cell transplant)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
183
Q

What are myelodysplastic syndromes

A

Group of Clonal stem cell disorders characterized by maturation defects assoc with ineffective hematopoiesis and high risk of transformation to AML; BM is replaced by Clonal progeny of neoplastic multipotent stem cell that can differentiate but does so in an ineffective and disordered fashion; all forms can transform to AML, but the ones after therapy (t-MDS) occur more frequently and rapidly

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
184
Q

What is the pathogenesis of MDS

A
  • epigenetics: mutations in DNA methylation and histone modifications
  • RNA splicing factors: mutations involving 3’ end of RNA splicing machinery
  • TF
  • 10% have LOF in TP53 high correlates with complex karyotype and poor clinical outcom
  • assoc with chrom ab including monosomies 5 and 7 deletions of 5q (RPS14) 7q 20q Nd trisomy 8 (MYC located here)
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
185
Q

What is the morphology of MDS

A

Hypercellular at diagnosis; most characteristic finding is disordered diff affecting erythroid, granulocytic, monocytes and megakaryocytic lineages; *ring siderobblasts, erythroblast with iron-laden macrophages, megaloblastoid maturation (as seen in B12 and folate def) and nuclear budding abnormalities (misshapen nuclei); pseudo-pelger-Huet cells (neutrophils with only 2 nuclear lobes); pawn ball megakaryocytes (single nuclear lobes or multiple separate nuclei); myeloid blasts increased but make up less than 20% of marrow; blood contains pelger-huet, giant platelets, macrocytes and poikilocytes

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
186
Q

What are the clinical features of primary MDS

A

Older adults (mean age 70); half discovered incidentally on blood testing; when sx present with weakness, infections and hemorrhages; worse outcomes predicted by higher blast counts and more severe cytopenias as wel las presence of multiple Clonal chrom ab

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
187
Q

What are the tyrosine kinase mutations in myeloproliferative disorders

A
  • chronic myelogenous leukemia: BCR-ABL fusion
  • polycythemia Vera: JAK2 point mut
  • essential thrombocythemia: JAK2 point mut; MPL point mut
  • primary myelofibrosis: JAK2 and MPL point mut
  • systemic mastocytosis: KIT point mut
  • chronic eosinophilic leukemia: FIP1L1-PDGFRA fusion (common)and PDE4DIP-PDGFRB fusion (rare - responds to imatinib therapy) assoc with Loeffler endocarditis
  • stem cell leukemia:: FGFR1 fusion genes
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
188
Q

What is the median survival in primary MDS

A

9 - 24 month; in tMDS only 4-8 months

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
189
Q

What are the treatment options for MDS

A

Allogeneic hematopoietic stem cell transplant in younger patients; older patients treated with supportive abx Nd transfusions

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
190
Q

What is the common pathogenic features of myeloproliferative disorders

A

Mutated constitutively active tyrosine kinase or other abberations in pathways that lead to growth factor independence

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
191
Q

What are the common clinical features of myeloproliferative disorders

A
  • increased proliferative drive in BM
  • Homing of neoplastic stem cells to secondary hematopoietic organs producing extramedullary hematopoiesis
  • variable transformation to spent phase characterized by marrow fibrosis and peripheral blood cytopenias
  • variable transformation to acute leukemia
How well did you know this?
1
Not at all
2
3
4
5
Perfectly
192
Q

What is CML

A

BCR-ABL fusion - created by a reciprocal (9;22)(q34;q11) translocation (philly chrom); cell of o robin is pluripotent hematopoietic stem cell

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
193
Q

What is the pathogenesis of CML

A

Activation of RAS and JAK/STAT; preferentially drive proliferation of granulocytic and megakaryocytic progenitors and causes ab release of immature granulocytic forms from marrow into blood *does not effect diff so will have mature elements in peripheral blood

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
194
Q

What is the morphology of CML

A

Hypercellular marrow *scattered macrophages with abundant wrinkled green-blue cytoplasm called sea-blue histiocytes ; increased deposition of reticulum is typical; blood reveals leukocytosis (often >100,000); platelets increased; spleen is enlarged (increased red pulp) and contains infarcts of varying age

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
195
Q

What are the clinical features of CML

A

Adults primarily but can occur in children; peak indigene 5th-6th decade; mild anemia and hyper metabolism bc increased cell turnover leading to fatigue, weakness, weight loss, anorexia; dragging sensation in ab (splenomegaly) or LUQ pain due to splenic infarct; slow progression even without treatment - median survival 3 years

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
196
Q

What is the accelerated phase of CML

A

Half of patients experience it; increasing anemia and thrombocytopenia; additional ab (trisomy 8, isochromosome 17q or duplication of Ph chrom seen); within 6-12 months terminates in a picture resembling acute leukemia (blast crisis)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
197
Q

What mutation has been linked to lymphoid blast crisis in CML

A

Mutations that interfere with activity of Ikaros (transcription factor that regulates diff of hematopoietic progenitors) - also seen in BCR-ABL positive B-ALL

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
198
Q

How is CML treated

A

BCR-ABL inhibitors

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
199
Q

What is polycythemia Vera

A

Characterized by increased marrow production of red cells, granulocytes, and platelets (panmyelosis)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
200
Q

What are the serum erythropoietin levels in polycythemia Vera

A

Low; (secondary forms have high); increased hematocrit leads to increased viscosity and sliding; thrombocytopenia and ab platelet function (prone to thrombosis and bleeding)

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
201
Q

What is the pathogenesis of polycythemia Vera

A

Valine to phenylalanine sub at 617; mutation in JAK2; if 2 mutated copies, higher white cell count, more sig splenomegaly, sx prutrits and greater rate of progression to spent phase

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
202
Q

What is the morphology of polycythemia Vera

A

Marrow is hypercellular; increase in red cell progenitors is subtle and accompanied by increase in granulocytic precursors and megakaryocytes; blood contains increased number of basophils and abnormally large platelets; late in course, PCV progresses to spent phase characterized by extensive marrow fibrosis accompanied by increased extramedullary hematopoiesis in splen and liver

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
203
Q

What are the clinical features of polycythemia Vera

A

Uncommon; adults; abnormal blood flow particularly on low-pressure venous side; patients are plethora can and cyanotic due to stagnation and deoxygenation of blood; HA, dizziness, HTN, and GI sx common; intense pruritis and peptic ulcer can occur (release of histamine from basophils); high cell turnover gives rise to hyperuricemia sx gout seen sometimes; DVT, MI, stroke; can cause budd chiari if thrombosis in hepatic vein or bowel infarction; minor hemorrhages common

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
204
Q

What is the prognosis for PCV without treatment

A

Die in months of diagnosis; treatment is maintaining red cell mass levels by phlebotomy - median survival is 10 years

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
205
Q

What is a spent phase of PCV

A

Clinical and anatomic features of primary myelofibrosis develop; undergoes this transition in 15-20% of patients after an average period of 10 years; marked by appearance of obliterative fibrosis in BM and extensive extramedullary hematopoiesis (spleen)

206
Q

What can PCV transform to

A

AML; but often lacks JAK2 mutations; transformation to ALL is rarely seen

207
Q

What is essential thrombocytosis

A

Assoc with point mut in JAK2 (more common) or MPL - RTKs normally activated by thrombopoietin; remaining cases have mut in calreticulin

208
Q

What are the clinical manifestations of essential thrombocytosis

A

Elevated platelet counts; separated from PCV and primary myelofibrosis based on absence of polycythemia and marrow fibrosis; iron deficiency is common; megakaryocytes markedly increased

209
Q

What is a characteristic symptom of essential thrombocytosis

A

Erythromelalgia - throbbing and burning of hands and feet caused by occlusion of small arterioles by platelet aggregates

210
Q

What is the median survival for essential thrombocytosis

A

Indolent; 12-15 years; thrombotic complications most likely in patients with very high platelet counts and homozygous JAK2 mutations; therapy is gentle chemo to suppress thrombopoiesis

211
Q

What is the hallmark of primary myelofibrosis

A

Development of obliterative marrow fibrosis; leads to cytopenias and extramedullary hematopoiesis; histo is identical to spent phase of myeloproliferative disorders

212
Q

What is the chief pathologic features of primary myelofibrosis

A

Extensive deposition of collagen in marrow by non-neoplastic fibroblasts; caused by 2 factors released by megakaryocytes: platelet derived growth factor and TGFbeta

213
Q

What is the morphology of primary myelofibrosis

A

Early marrow is hypercellular; megakaryocytes are large, dysplastic and abnormally clustered - have unusual nuclear shapes (cloud like); with progression marrow becomes hypocellular and fibrotic; veery late, marrow can turn to bone (osteosclerosis)

214
Q

What are the blood findings of primary myelofibrosis

A

Premature release of uncleared erythroid and early granulocytes progenitors (leukoerythroblastosis); tear-drop shaped red cells (dacryocytes) seen

215
Q

What are the clinical features of primary myelofibrosis

A

Less common than PCV and ET; >60 yo; comes to attention bc of normochromic normocytic anemia and splenomegaly; can by complicated by hyperuricemia and secondary gout; white count normal or mildly reduced usually but can be elevated early on; more difficult to treat than PCV and ET; median survival is 3-5 years; use JAK2 inhibitors

216
Q

What is histiocytosis

A

Proliferative disorders of dendritic cells or macrophages

217
Q

What are the langerhans cell histiocytoses

A

Spectrum of proliferation’s of special type of immature dendritic cell (langerhan cell)

218
Q

What is the most common mutation in langerhans cell histiocytosis

A

Valine to glutamate substitution at residue 600 in BRAF (seen in hairy cell leukemia also)

219
Q

What do langerhans cells look like

A

Abundant vacuolated cytoplasm and vesicular nuclei containing linear grooves or. Folds; presence of birbeck granules in cytoplasm is characteristic* - pentalaminar tubules with dilated terminal end producing tennis racket like appearance which contain langerin; express HLA-DR S-100 and CD1a

220
Q

What is multifocal multisystem langerhans cell histiocytosis (letterer-siwe dz)

A

Occurs before 2 years of age; development of cutaneous lesions resembling seborrheic eruption caused by infiltrates of langerhans cells ver font and back of trunk and scalp; most have HSM, LAD, pulm lesions and destructive osteolytic bone lesions; anemia, thrombocytopenia and predisposition to recurrent infections (otitis media and mastoiditis); in some instances can be anaplastic and referred to as langerhans cell sarcoma; *if untreated rapidly fatal; with chemo, 5 year survival

221
Q

What is unifocal and multifocal unisystem langerhans cell histiocytosis (eosinophilic granuloma)

A

Proliferation of langerhans admixed with eosinophils, lymphocytes, plasma cells and neutrophils; arises within medullary cavities of bones (most commonly calvarium, rib and femur); less commonly, unisystem lesions of identical histo arise in skin, lungs or stomach; unifocal lesions most commonly affect skeletal system in older children or adults and is indolent (spontaneous healing to local excision); multifocal unisystem dz usually affects young kids - present with mult erosive bony masses, involvement of post pit stalk leads to DI *Hand-Schiller-Christian triad (DI, calvarial bone defects and exopthalamos); treated successfully with chemo or spontaneous recovery

222
Q

What is pulmonary langerhans cell histiocytosis

A

Adult smokers; regress spontaneously when quit smoking; reactive proliferation’s of langerhans cells; some associated with BRAF mutations

223
Q

What do langerhan’s cells express

A

Normal epidermal: CCR6;; their neoplastic counterparts express CCR6 and CCR7 which allows them to migrate into tissues that express CCL20 (ligand for CCR6 in skin and bone) and CCL19 and 21 (ligands for CCR7 in lymphoid organs)

224
Q

What are the 4 functions of the spleen

A
  • phagocytosis of blood cells and particulate matter: RBCs undergo deformation during passage from cords into sinusoids; if deformability is decreased, become trapped in cords and phagocytosed by macrophages; also responsible for pitting of RBCs (inclusions - Heinz bodies and Howell-jolly bodies are excised)
  • ab production: dendritic cells present to T cells; T and B cell interaction at edges of white pulp lead to gen of ab secreting plasma cells found in red pulp
  • hematopoiesis: compensatory
  • sequestration of formed blood elements: platelets sequestered in red pulp producing thrombocytopenia
225
Q

What are asplenic patient susceptible do

A

Pneumococci, meningococcal, haemophilus influenzae

226
Q

What sx does splenomegaly cause

A

Dragging sensation; discomfort after eating; hypersplenism - anemia, leukopenia, thrombocytopenia via sequestration of elements

227
Q

What is nonspecific acute splenitis

A

Enlargement of spleen as a result of blood-borne infections; caused by microbiologic agents themselves and cytokines released; spleen enlarged and soft; major feature is acute congestion of red pulp which can face lymphoid follicles; white pulp can undergo necrosis *especially when causative agent is hemolytic strep

228
Q

What are the disorders assoc with splenomegaly

A
  • infections: mono, TB, typhoid, brucellosis, CMV, syphilis, malaria, histoplasmosis, toxoplasmosis, kala-azar, trypanosomiasis, schisto, leishmaniasis, echinococcosis
  • congestive states related to portal HTN: cirrhosis, portal or splenic v thrombosis, cardiac failure
  • lyphohematogenous disorders: Hodgkin, NHL, lymphocytic leukemia, multiple myeloma, myeloproliferative disorders, hemolytic anemia’s
  • immune inflamm conditions: RA, SLE
  • storage dz: gaucher, Niemann-pick dz, mucopolysaccharidoses
  • amyloidosis, primary neoplasms and cysts, secondary neoplasms
229
Q

What causes congestive splenomegaly

A

Chronic venous outflow obstruction may be caused by intrahepatic disorder or extrahepatic that directly impinge upon portal or splenic veins; lead to splenic or portal v HTN; right heart failure

230
Q

What is the morphology of congestive splenomegaly

A

Marked enlargement; organ is firm; capsule is thickened and fibrous; red pulp is congested early but becomes fibrotic with time; deposits of collagen in BM of sinusoids; results in excessive destruction of blood cells because slow blood flow so longer exposure

231
Q

What are the most common neoplasms of the spleen

A

Lymphangiomas and hemangiomas

232
Q

What does complete absence of the spleen usually occur with

A

Other congenital abnormalities such as situs inversus and cardiac malformations; hypoplasia is more common

233
Q

What is an accessory spleen (spleniculi)

A

Common; small spherical structures; can ve found anywhere in ab cavity; impt in hereditary sphere Tyson’s and immune thrombocytopenia purpura where splenectomy is used as treatment

234
Q

What are the most common predisposing conditions to splenic rupture

A

Mono, malaria, typhoid, lymphoid neoplasms; chronically enlarged spleens unlikely to rupture because toughening of extensive reactive fibrosis

235
Q

What can cause the thymus to involute

A

Exposure to severe illness and HIV

236
Q

What is thymic hyperplasia

A

Appearance of B ell germinal centers within the thymus which is referred to as thymic follicular hyperplasia; *most frequently encountered in myasthenia graves; sometimes seen in graves, SLE, scleroderma, RA, and other AI dz

237
Q

What is a thymoma

A

Tumors of thymic epithelial cells; also contain benign immature T cells (thymocytes); 3 subtypes: tumors that are cytologically benign and noninvasive, tumors that are cytologically benign but invasive or metastatic, tumors that are cytologically malignant (thymic carcinoma); in all c ategories occur in adults >40 yo males and females equally; most in anterior superior mediastinum

238
Q

What is the morphology of noninvasive thymoma

A

Composed of medullary-type epithelial cells or mix of medullary and cortical; elongated or spindle shaped

239
Q

What is the morph of invasive thymoma

A

Cytologically benign but locally invasive; more likely to met; cortical cells; *penetrate through capsule into surrounding structures; extensive invasion poor outcome

240
Q

What is the morph of thymic carcinoma

A

Fleshy invaisve masses; can met to lungs; micro: squamous cell carcinomas; next most common is lymphoepithelioma like carcinoma (sheets of cells with indistinct borders that looks like nasopharyngeal carcinoma; EBV related)

241
Q

What are the clinical features of thymomas

A

Impingement of mediastinal structures; myasthenia gravis; also assoc with hypogammaglobulinemia, pure red cell aplasia, graves, pernicious anemia, dermatomysotis-polymyositis and cushing; cortical thymoma rich in thymocytes more likely to be assoc with AI dz

242
Q

How is anemia usually diagnosed

A

Reduction in hematocrit (ratio of packed red cells to total blood volume) and hemoglobin concentration of the blood

243
Q

What are microcytic hypochromic anemia’s caused by

A

Disorders of hemoglobin synthesis (mostly iron deficiency)

244
Q

What are macrocytic anemia’s caused by

A

Abnormalities that impair maturation of erythroid precursors in bone marrow

245
Q

What are the most useful red cell indices

A

Mean cell volume, mean cell hemoglobin, mean cell hemoglobin concentration, red cell distribution width

246
Q

What can anemia cause

A

Dyspnea on mild exertion, hypoxia can cause fatty change in the liver, myocardium and kidney

247
Q

What are the classifications of anemia according to underlying mechanisms

A

Blood loss
Increased red cell destruction
Decreased red cell production

248
Q

What are the inherited genetic defects that can lead to hemolysis

A
  • red cell membrane disorders: hereditary spherocytosis; hereditary elliptocytosis
  • hexose monophosophate shunt enzyme def (G6PD def and glutathione synthetase def)
  • glycolytic enzymes def (Pyruvate kinase and hexokinase def)
249
Q

What acquired genetic defects can cause increased hemolysis

A

Def of phosphatidylinositol-liked glycoproteins (paroxysmal nocturnal hemoglobulinuria)

250
Q

What inherited genetic defects an lead to decreased red cell production

A
  • stem cell depletion: fanconi anemia; telomerase defects

- defects affecting erythroblast maturation: thalassemia syndromes

251
Q

What infection can cause decreased red cell production

A

Parvovirus B19

252
Q

What happens to white cells during significant bleeding

A

The decrease in BP stimulates adrenergic hormone which mobilizes granulocytes and results in leukocytosis; initially red cells are normochromic and normocytic but as marrow production increases there is an increase in reticulocyte count (reticulocytosis - macrocytes)

253
Q

What features do all hemolytic anemia’s share

A

Short life of RBC, elevated EPO, accumulation of hemoglobin degredation products

254
Q

What causes extravascular hemolysis

A

Alterations that make the red cell less deformable; some hemolobin will escape phagocytes which leads to decreases in plasma haptoglobin (alpha globulin that binds free hemoglobin); Benefit from splenectomy

255
Q

What can cause intravascular hemolysis

A

Trauma (cardiac valves , thrombotic narrowing, repetitive physical trauma), complement, parasites; toxic injury via clostridial sepsis (digest red cell membrane)

256
Q

What is the manifestation of intravascular hemolysis

A

Anemia, hemoglobinemia, hemoglobinuria, hemosiderinuria, aNd jaundice; as serum haptoglobin is depleted, free hemoglobin oxidizes to methemoglobin which is brown; renal prox tubules reabsorb most of fisted hemoglobin and methemoglobin but some passes into urine (red-brown color); iron can accumulate in tubule cells (renal hemosiderosis)*NO splenomegaly

257
Q

What is the serum bilirubin in all hemolytic anemia’s

A

Unconjugated

258
Q

What is the morphology of hemolytic anemia

A

Increased number of erythroid precursors (normoblasts) in marrow; prominent retioculocytosis; accumulation of hemosiderin in spleen, liver and BM; elevated biliary excretion of bilirubin promotes formation of pigment gallstones (cholelithiasis)

259
Q

What is hereditary spherocytosis

A

Inherited disorder caused by intrinsic defects in red cell membrane skeleton that renders them spheroid, less deformable and vulnerable to splenic sequestration and destruction; AD

260
Q

What is the pathogenesis of hereditary spherocytosis

A

Insufficiency of membrane skeletal components; usually ankyrin and band 4.2 binds section to ion transporter (band 3); mutations most commonly affect ankyrin, band 3, spectrin, or band 4.2 (most are frameshift or nonsense); *splenectomy - stay in spleen and eaten by macrophages; also depletes them of lactose, decreased pH

261
Q

What is the morphology of hereditary spherocytosis

A

Small dark staining red cells on smear that lack central zone of pallor; not pathognomonic; reticulocytosis, marrow erythroid hyperplasia, hemosiderosis and mild jaundice; cholethiasis; moderate splenomegly

262
Q

What is the diagnosis of hereditary spherocytosis based on

A

Family hx, hematologic findings and laboratory evidence; in 2/3 of patients red cells are abnormally sensitive to osmotic lysis when incubated in hypotonic salt solutions; also have increased mean cell hemoglobin concentration (due to dehydration by loss of K and water)

263
Q

What are the clinical features of hereditary spherocytosis

A

Anemia, splenomegaly(only hemolytic anemia with splenomegaly) and jaundice; in compound heterozygotes can present as jaundice at birth and requires transfusion; chronic hemolytic anemia; can have aplastic crises usually triggered by parvovirus infection(kills red cell progenitors so cant compensate); hemolytic crises cause by mono (less sig than aplastic crises); splenectomy decreases sx but risk of sepsis

264
Q

What is glucose 6 phosphate dehydrogenase deficiency

A

G6PD reduces NADP to NADPH which then provides reducing equivalents needed for glutathione to reduced glutathione; X linked recessive; G6PD- (black) and Mediterranean cause most severe dz;

265
Q

What kind of hemolysis is characteristic of G6PD def

A

Episodic; cause by exposures that generate oxidant stress (most commonly infections - viral hep, pneumonia and typhoid most common); drugs (antimalarials, sulfonamindes, nitrofurantoin)and food(fava bean); extra and intravascular hemolysis

266
Q

How do oxidants cause hemolysis in G6PD def

A

Exposure causes cross linking of reactive sulfhyryl groups on globin chains which become denatured and form membrane bound precipitates known as Heinz bodies (seen as dark inclusions within red cells stained with crystal violent); Heinz bodies can damage membrane to cause intravascular hemolysis; as they pass through spleen, Heinz bodies plucked out (bite cells and spherocytosis)

267
Q

Which cells are at risk for hemolysis in G6PD def

A

Only older red cells; hence self limited; because only limited, no splenomegaly or gallstones

268
Q

What is sickle cell

A

Point mutation in beta globin that promotes polymeriztion of deoxygenated hemoglobin leading to red cell distortion, hemolytic anemia, microvascular obstruction and ischemic tissue damage

269
Q

What is the normal hemoglobin in adults

A

HbA (alpha2beta2) and small amounts of HbA2 (alpha2delta2) and fetal HbF (alpha2gamma2)

270
Q

What hemoglobin is seen in sickle cell

A

HbS

271
Q

Why does sickle cell provide protection against malaria

A
  • parasites consumes O2 and decrease intracellular pH which promote sickling; cleared more rapidly by phagocytes in spleen and liver
  • sickling impairs formation of membrane knobs containing a protein made by parasite called PfEMP-1 (implicated in adhesion of infected cells to endothelium - causes cerebral malaria)
272
Q

What is the pathogenesis of sickle CLL

A

HbS polymers stack into polymers when deoxygenated which converts red cell cytosol into viscous gel; with continued deoxygenation, HbS assemble into long needle-like. Fibers within red cells producing distorted sickle or holly leaf shape

273
Q

How does interaction of Hb change the rate of sickling

A

interaction of HbS with other types of hemoglobin: in heterozygotes HbA prevents HbS polymerization; HbF does so even more (reason infants aren’t sx till 5 months); HbC - HbSC (HbS HbC heterozygotes) lose solute and water and become dehydrated which increases intracellular concentration of HbS - sx sickling disorder but milder than sickle cel

274
Q

How does the mean cell hemoglobin concentration affect the rate of sickling

A

Higher HbS concentrations increase the probability that aggregation and polymerization will occur; intracellular dehydration which increases mean cell hemoglobin concentration facilitates sickling; conditions that decrease MCHC will reduce dz severity (ex: homozygous for HbS but also have alpha-thalassemia which reduces Hb synthesis)

275
Q

How does intracellular pH affect sickle cell disease severity

A

Decrease in pH reduces oxygen affinity of hemoglobin which increase the fraction of deoxygenated HbS and will increase sickling

276
Q

How does the transit time of red cells through microvascular beds affect sickle cell

A

Slow transit times increases aggregation (ie: spleen and BM and vascular beds that are inflamed)

277
Q

How does sickling cause damage to red cells

A

HbS polymers herniate through membrane and project from the cell ensheathed by lipid bilayer -> influx of Ca2+ which induces cross linking of membrane proteins and activates efflux of K+ and H2O - become dehydrated and rigid and become irreversibly sickled

278
Q

Are microvascular conclusions related to the number of irreversibly sickled cells

A

No; more dependent on inflammation; reduction in NO plays a part

279
Q

What is the morphology of sickle cell

A

In blood, variable numbers of irreversibly sickled cells; Howell-jolly bodies (nuclear remnants) present in some red cells due to asplenia; BM is hyperplastic; bone resorption results in prominent cheekbones an change in the skull that resembles a crew cut on X-ray; pigment gallstones and hyperbilirubinemia; first have splenomegaly then have splenic infarction (autosplenectomy); vascular occlusions anywhere (if pulm a can cause cor pulmonale); in adults, leg ulcers

280
Q

What are the clinical features of sickle cell

A

Moderately severe hemolytic anemia assoc with reticulocytosis, hyperbilirubinemia; vaso-occlusive crises (pain crises): episodes of hypoxia injury and infarction that cause severe pain most commonly bone, lungs, liver, brain, spleen, and penis; in kids: hand-foot syndrome or dactylitis of bones; acute chest syndrome: vaso-occlusive crisis involving lung - presents with fever, cough, chest pain, pulm infiltrates

281
Q

What affect can sickle cell have specific to males

A

Priapism; hypoxia damage and ED

282
Q

What is the most common cause of mortality in sickle cell

A

Vaso-occlusive crises

283
Q

What is a sequestration crisis

A

Occur in children with intact spleens; rapid splenic enlargement, hypovolemia and sometimes shock; can be fatal; needs exchange transfusion

284
Q

What is aplastic crises

A

Infection of red cell progenitors by parvovirus B19 which causes transient cessation of erythropoietin and sudden worsening of anemia

285
Q

What can chronic hypoxia cause in sickle cell

A

Impaired growth; organ damage; sickling in renal medulla causes hyposhenuria (inability to concentrate urine)

286
Q

What are common infections in ppl with sickle cell.

A

Pneumococcus pneumoniae aNd haemophilus infelunzae septicemia and meningitis

287
Q

What is a treatment for sickle cell

A

Hydroxyurea: increases red cell HbF, has antiinflammatory effect

288
Q

What are the thalassemia syndromes

A

Inherited mutations that decrease the synthesis of either alpha or beta globin gains of hemoglobin A leading to anemia, tissue hypoxia and red cell hemolysis; alpha encoded on chrom 16 and beta encoded on chrom 11; endemic in Mediterranean; decrease red cell production and lifespan

289
Q

What mutations caus beta thalassemia

A
  • beta0 mutations: assoc with absent beta globin synthesis; chain terminator mutations most common - mostly caused. By new stop codon within an exon
  • beta+ mutations: reduced beta globin synthesis; splicing mutations most common cause and most lie within introns; promoter region mutations also assoc with this kind
290
Q

How does impaired beta globin synthesis result in anemia

A
  • deficit in HbA synthesis produces underhemoglobinized hypochromic microcytic red cells with subnormal oxygen transport capacity
  • diminishes survival of red cells resulting from imbalance in alpha and beta globin; red cell precursors form insoluble unpaired alpha chains which damages the membrane and undergo apoptosis - thus have impaired erythropoiesis - those that do leave the marrow undergo extravascular hemolysis
291
Q

What effects does severe beta thalassemia have on the marrow

A

Erythroid hyperplasia and extramedullary hematopoeisis; expanding mass of red cell precursors erodes bony cortex, impairs bone growth and produces skeletal abnormalities; can cause cachexia in severe cases

292
Q

What complication does ineffective erythropoeisis in beta thalassemia cause

A

Excessive absorption of dietary iron; ineffective erythropoeisis suppresses hepcidin - secondary hemochromatosis

293
Q

What are the clinical symptoms of beta thalassemia

A

Anemia; those with 2 beta thalassemia alleles (B+/B+, B0B0 or B+B0) have transfusion dependent anemia called Beta thalassemia major; those with one allele have microcytic anemia referred to as minor or trait

294
Q

What is beta thalassemia intermedia

A

2 defective beta globin genes and an alpha thalassemia defect which improves effectiveness of erythropoiesis and red cell survival

295
Q

What are the features of beta thalassemia major

A

Most common in Mediterranean and parts of Africa; anemia manifests 6-9 on this after birth; major re cell hemoglobin is HbF which is elevated

296
Q

What is the morphology of beta thalassemia major

A

Blood smear shows severe red cell ab: variation in size (anisocytosis), shape (poikilocytosis), microcytosis, and hypochromia; target cells (hemoglobin collects in center); reticulocyte elevated but not to level you would think; in bones of fate and skull marrow erodes existing cortical bone and induces new bone formation “crewcut appearance” on XR; iron overload damages heart, liver and pancreas

297
Q

Are the unpaired alpha chains in beta thalassemia visible on blood smear

A

No

298
Q

What is the outcome of beta thalassemia

A

Need transfusions or die at early age; in those who survive, impt cause of death is cardiac dz - treat with iron chelators to prevent; can survive till 3rd decade but overall grim

299
Q

What are the features of beta thalassemia minor

A

Asymptomatic; hypochromic, microcytosis, basophilic stippling; increase in HbA2 (diagnostic); HbF normal or slightly increased

300
Q

What happens to newborns with alpha thalassemia

A

Excess unpaired gamma globin chains form gamma tetramers known as hemoglobin Barts; in older children and adults, excess beta chains form beta4 tetramers known as HbH; both of these are more soluble than the alpha tetramers so hemolysis and infective erythropoeisis isn’t as severe as beta thalassemia

301
Q

What is the most common cause of alpha thalassemia

A

Deletion

302
Q

What is the silent carrier state of alpha thalassemia

A

Deletion o single alpha globin gene; asymptomatic and have slight microcytosis

303
Q

What is alpha thalassemia trait

A

Deletion of 2 alpha genes from single chrom (more common in Asians) or deletion of one alpha gene from 2 chrom (Africans); people with the former have kids with a risk of HbH or hydrops fetalis; microcytic minimal anemia; HbA2 normal or low

304
Q

What is hemoglobin H dz (HbH)

A

Caused by a deletion of 3 alpha globin genes; most common in Asians; tetramers of HbH form - extremely high affinity for oxygen and therefore not useful for oxygen delivery leading to tissue hypoxia disproportionate to level of hemoglobin; HbH also prone to oxidation which causes it to precipitate and form intracellular inclusions that promote red cell sequestration; result is moderate anemia resembling beta thalassemia intermedia

305
Q

What is the most severe form of alpha thalassemia

A

Hydrops fetalis; deletion of all 4 alpha globin; gamma tetramers have high affinity for oxygen and little delivered to tissues; survival in early development due to expression of zeta chains which binds to gamma chains; fetal distress becomes apparent in 3rd trimester; infants can be saved with intrauterine transfusion; fetus shows pallor, edema, and massive HSM; lifelong dependence on blood transfusions for survival with assoc risk of iro overload

306
Q

What is paroxysmal nocturnal hemoglobinuria

A

Results from acquired mutations in phosphatidlinositol glucan complementation Group A gene (PIGA) which is essential for synthesis of membrane assoc complement regulatory proteins *only hemolytic anemia caused by acquired genetic defect; X linked - single acquired mutation needed

307
Q

What proteins are PNH blood cells deficient in

A

Decay-accelerating factor or CD55, membrane inhibitors of reactive lysis or CD59 (most important bc inhibitor of C3 convertase which prevents spontaneous activation of alternative complement pathway), and C8 binding protein

308
Q

What kind of hemolysis is seen with PNH

A

Intravascular (caused by MAC); damage by complement or lysis; chronic hemolysis without dramatic hemoglobinuria is most common; tendency for them to lyse at night is bc decreased blood pH at night which increases activity of complement; loss of heme iron in urine (hemosiderinuria) leads to iron deficiency

309
Q

What is the leading cause of disease-related death in people with PNH

A

Thrombosis; venous involving hepatic, portal or cerebral veins; some can develop AML or myelodysplastic syndrome

310
Q

How is PNH diagnosed

A

Flow cytometry; treated with eculizumab (inhibits conversion of C5 to C5a) - lowers need for transfusions and risk of thrombosis but increased risk of serious or fatal meningococcal infections; only cure is HSC transplant

311
Q

How do you test for immuonhemolytic anemia’s

A

Direct Coombs antiglobulin test; tests for abs or complement on red cells - patients red cells are mixed with abs to immunoglobulin or complement
Indirect Coombs: patients serum tested for its ability to agglutinate red cells bearing antigens

312
Q

What are the warm ab type (IgG abs active at 37 degrees C) immunohemolytic anemia

A

Primary (idiopathic)

Secondary: autoimmune disorders (SLE), drugs, lymphoid neoplasms

313
Q

What are the cold agglutinin type (IgM abs below 37 degrees C) immunohemolytic anemia’s

A

Acute (mycoplasma infection, mono)

Chronic: idiopathic, lymphoid neoplasms

314
Q

What are the cold hemolysis type (IgG abs below 37 degrees C) immunohemolytic anemia’s

A

Rare; occurs mainly in children following viral infections

315
Q

What is the most common type of immunohemolytic anemia

A

Warm ab type; mostly extravascular hemolysis; IgG coated red cells bind to Fc receptors on phagocytes which remove membrane d urging partial phagocytosis -> converts them to spherocytes which are sequestered

316
Q

What causes drug induced immunohemolytic anemia

A
  • antigenic drugs: usually follows large IV doses of drug 1-2 weeks after; ie penicillin and cephalosporins bind to red cell membrane and recognized by anti drug abs (in penicillin induced bs only bind to drug) in quinidine induced abs recognize complex of drug and membrane protein; act as opsonin
  • tolerance breaking drugs: alpha methyldopa; induces production of abs against red cell antigens (usually Rh)
317
Q

What causes cold agglutinin type immunohemolytic anemia

A

Mycoplasma, EBV, CMV, influenza and HIV - self limited in these circumstances
Chronic cold agglutinin occurs in assoc its B cell neoplasms; clinical sx result from binding of IgM to red cells in areas where temp is lower )(fingers, toes, ears); vascular obstruction caused by agglutinate red cells results in pallor, cyanosis, and raynaud

318
Q

What is the cold hemolysin type of immunohemolytic anemia

A

Responsible for paroxysmal cold hemoglobinuria; rare; causes substantial and sometimes fatal intravascular hemolysis and hemoglobinuria; abs are IgGs that bind to P blood group antigen on red cell surface in peripheral regions; complement mediated lysis occurs when cells recirculate to warm areas; most seen in kids following viral infections (in this setting, transient and most recover); treatment with removing offending agent - if doesn’t work immunosuppressive drugs and splenectomy

319
Q

What is the most significant cause of hemolysis related to trauma of RBCs

A

Cardiac valve prosthesis and microangiopathic disorders (seen with DIC but also occurs with thrombotic thrombocytopenic purpura, hemolytic uremic syndrome, malignant HTN, SLE and disseminated cancer)- mostly mechanical valves; * results in red cell fragments (schistocytes) burr cells, helmet cells, and triangle cells in blood smear

320
Q

What is the common feature of megaloblastic anemia

A

Impairment of DNA synthesis that leads to ineffective hematopoiesis and abnormally large erythroid precursors and red cells; 2 forms: pernicious (B12) and folate deficiency anemia

321
Q

What are the causes of megaloblastic anemia

A
  • decreased intake of B12 (vegetarian)
  • impaired absorption of B12: intrinsic factor deficiency (pernicious anemia, gastrectomy), malabsorption states, diffuse intestinal dz (lymphoma, systemic sclerosis), ileal resection - fish tapeworm infestation
  • folic acid deficiency (alcoholism, anticonvulsants, oral contraceptives, hemodialysis)
  • increased requirement of folic acid: pregnancy, disseminated cancer
  • methotrexate
322
Q

Why does b12 and folate def cause anemia

A

Both are cofactors for formation of thymidine

323
Q

What is the morphology of megaloblastic anemia

A

Macro-ovalocytes; lack central pallor - appear hyperchromic but MCHC is not elevated; variation in size (anisocytosis) and shape; reticulocyte count low; neutrophils are larger and show nuclear hypersegmentation; marrow is hypercellular; giant metamyelocytes and band forms

324
Q

What is pernicious anemia

A

Caused by autoimmune gastritis that impairs production of intrinsic factor

325
Q

Describe the absorption of B12

A

Freed from food via pepsin and binds haptocorrin -> in duodenum released from haptocorrin by pancreatic proteases and associates with intrinsic factor -> goes to ileum where it is endocytosed by ileal enterocytes that express cubilin (receptor for IF) -> B12 assoc with transcobalamin II and is secreted into plasma -> liver; also goes through a B12 independent pathway which is why you can treat with high doses of B12

326
Q

What is B12 required for

A
  • conversion of homocysteine to methionine; in same reaction N5methyl FH4 is converted to FH4 which is needed for conversion of dUMP to dTMP (building block of DNA); anemia improves if give folic acid
  • isomerization of methylmalonyl coA to succinylcholine coA-> def lead sto increased plasma and urine methylmalonic acid; leads to buildup of methylmalonate and propionate and formation of FA into neuronal lipids - > neuro complications of B12 def
327
Q

Are the neuro sx caused by B12 def cured with folate

A

No; may actually worsen

328
Q

What population does pernicious anemia occur in

A

All races; older adults; rare younger than 30; have tendency to form abs against multiple self antigens

329
Q

What is the pathogenesis of pernicious anemia

A

Autoimmune attack on gastric mucosa; chronic atrophic gastritis; type I ab that blocks binding of B12 to IF - found in plasma and gastric juice; type II abs ovens binding of IF-B12 to ileal receptor; type III present in most cases - recognize alpha and beta subunits of gastric proton pump but not specific for pernicious anemia; abs are used for diagnosis but not pathology; autoreactive T cells initiates injury and triggers formation of autoabs; assoc with AI adrenalitis and thyroiditis

330
Q

What can cause vit B12 def other than pernicious anemia

A

Achlorhydria; gastrectomy; loss of exocrine pancreases function

331
Q

What is the morphology of pernicious anemia

A

Fundic gland atrophy; replace by mucus secreting goblet cells (intestinalization); tongue becomes beefy, shiny, glazed (atrophic glossitis); Demyelination of dorsal and lateral spinal tracts -> spastic paresis, sensory ataxia and paresthesias in lower limbs

332
Q

What is the diagnosis of pernicious anemia based on

A

Megaloblastic anemia, leukopenia with hypersegmented granulocytes, low serum B12, and elevated serum levels of homocysteine and methylmaloic acid; confirmed with increase in hct after parenteral administration of B12

333
Q

What are elevated homocysteine levels a risk for

A

Atherosclerosis and thrombosis; B12 can cure everything except risk of gastric carcinoma

334
Q

What is the proper name for folic acid deficiency

A

Deficiency of pteroylmonoglutamic acid

335
Q

What is FH4 involved in

A

Purine synthesis, conversion of homocysteine to methionine, deoxythymidylate monophosphate (dTMP) synthesis

336
Q

What are the richest sources of folic acid

A

Green vegetables

337
Q

What is unique about folate deficiency in alcoholics with cirrhosis

A

Caused by trapping in liver, excessive urinary loss and disordered folate metabolism under these circumstances, accompanied by general malnutrition and cheilosis, glossitis and dermatitis

338
Q

What does methotrexate inhibit

A

Dihydrofolate reductase

339
Q

What are the lab values of folate def

A

Megaloblastic anemia; high homocysteine but low methylmalonate; no neuro changes

340
Q

What is important to do before beginning folate therapy for folate deficiency

A

Make sure it is not B12 deficiency (bc can worsen neuro sx)

341
Q

Who is iron deficiency anemia common in

A

Toddlers, adolescent girls and women of child bearing age

342
Q

What are the major sites of iron storage

A

Liver and mononuclear phagocytes

343
Q

Where is iron absorbed

A

Proximal duodenum

344
Q

How is iron absorbed

A

Nonheme iron converted to Fe2+ (ferrous) by b cytochromosomes and STEAP3; then transported by divalent metal transporter DMT1; transported into blood via ferry port in coupled to oxidation to Fe3+ by Hephaestin and ceruloplasmin; Fe3+ binds to transferrin and delivers it to marrow

345
Q

What does hepcidin do

A

Binds to ferroportin and inhibits iron absorption (causes it to be endocytosed)

346
Q

What are mutations that disable TMPRSS6 related to

A

Microcytic anemia; it is an hepatic transmembran serine protease that normally suppresses hepcidin production when iron stores are low; affected patients have high hepcidin resulting in reduced iron absorption and failure to respond to iron therapy

347
Q

How does secondary hemochromatosis occur in diseases assoc with ineffective erythropoiesis

A

Ineffective erythropoiesis suppresses hepcidin production even when iron stores are high

348
Q

What is the absorption of inorganic iron enhanced by

A

Ascorbic acid, citric acid, amino acids and sugars; inhibited by tannates (tea), carbonates, oxalates, and phosphates

349
Q

Where does dietary iron inadequacy occur

A
  • infants: small amounts of iron in milk - cow’s milk contains more but bioavailability is poor
  • impoverished
  • older adults: poor dentition
  • teenagers because eat junk
350
Q

What malabsorption syndromes can cause iron deficiency

A

Sprue, fat malabsorption, and chronic diarrhea; gastrectomy (decreases acidity of prox duodenum and increases speed of gut contents passing through)

351
Q

What is the most common cause of iron deficiency

A

Chronic blood loss

352
Q

What kind of anemia does iron deficiency produce

A

Hypochromic microcytic anemia

353
Q

What is the morphology of iron deficiency

A

BM shows increase in erythroid progenitors; disappearance of stainable iron from macrophages in BM (Prussian blue); “pencil cells”

354
Q

What are the clinical features of iron deficiency anemia

A

Koilonychia, alopecia, atrophic changes in tongue and gastric mucosa and intestinal malabsorption; pica - eat clay and flour and move their limbs during sleep; *esophageal webs with microcytic hypochromic anemia and atrophic glossitis - Plummer Vinson syndrome

355
Q

What does the diagnosis of iron deficiency anemia depend on

A

Lab; Hb and Hct low; iron and ferritin low and total plasma iron binding capacity is high; elevated transferrin levels but low saturation; low hepcidin

356
Q

What is the most common cause of anemia in hospitalized patients

A

Anemia of chronic disease; reduction in proliferation of erythroid progenitors and impaired iron utilization

357
Q

What chronic illnesses are associated with anemia of chronic disease

A
  • chronic microbial infections (osteomyelitis, bacterial endocarditis, lung abscess)
  • chronic immune disorders (RA, regional enteritis)
  • neoplasms (carcinomas of lung and breast and Hodgkin)
358
Q

What are the features of anemia of chronic disease

A

Low serum iron, reduced total iron binding capacity, abundant stored iron in tissue macrophages; *IL-6 stimulate increase in hepcidin production - results in progenitors starved for iron; EPO low; mild; can be normocytic and normochromic or hypochromic and microcytic; high ferritin level

359
Q

What is aplastic anemia

A

Syndrome of chronic primary hematopoietic failure and pancytopenia; mostly autoimmune

360
Q

What viral infections can cause aplastic anemia

A

Viral hepatitis of non-A non-B non-C non-G type

361
Q

What is Fanconi anemia

A

AR; defects in multiprotein complex required for DNA repair; marrow hypofunction -> aplastic anemia; accompanied by hypoplasia of kidney and spleen and bone anomalies (mostly thumbs or radii)

362
Q

What defects are seen in some cases of adult-onset aplastic anemia

A

Inherited telomerase defects; premature stem cell exhaustion and marrow aplasia

363
Q

What are most cases of aplastic anemia caused by

A

Idiopathic

364
Q

What chemical agents can cause aplastic anemia

A

Alkylating agents, antimetabolites, benzene, chloramphenicol, inorganic arsenicals; phenylbutazone, methyhlphenylethylhydantoin, carbamazepine, penicillamine, gold salts

365
Q

What physical agents can cause aplastic anemia

A

Whole body irradiation, viral infections, CMV, EBV, herpes zoster

366
Q

What is the pathogensis of aplastic anemia

A

Autoreactive T cell clones; think GPI-linked proteins may be targets and explains why aplastic anemia is assoc with PNH

367
Q

What is the morphology of aplastic anemia

A

Hypocellular BM; fat cells, fibrous stroma and scattered lymphocytes and plasma cells remain; dry tap aspiration so need BM biopsy; granulocytopenia and thrombocytopenia

368
Q

What are the clinical features of aplastic anemia

A

Any age in either sex; pancytopenia; NO splenomegaly; macrocytic and normochromic anemia; reticulocytopenia is the rule; need BM biopsy to rule out myelodysplastic syndromes (hypocellular vs hypercellular BM); bone marrow transplant is treatment of choice

369
Q

What is pure red cell aplasia

A

Primary marrow disorder in which only erythroid progenitors are suppressed; can occur in assoc with thymoma or large granular lymphocytic leukemia, drugs, AI, or parvovirus infection; resection of thymoma leads to improvement; otherwise immunosuppressive therapy; in parvovirus B19, transient and unimportant - only if immunosuppressed or already have severe hemolytic anemia

370
Q

What is myelophthisic anemia

A

Marrow failure in which space-occupying lesions replace normal marrow; *most common cause is met cancer - breast, lung and prostate; can occur with granulomatous dz; also a feature of spent phase of myeloproliferative disorders; abnormal release of nucleated erythroid precursors and immature granulocytic forms from marrow (leukoerythroblastosis) and tear drop shaped red cells

371
Q

What are the features of anemia due to chronic renal failure

A

Proportional to the severity of uremia; caused by diminished synthesis of EPO; also see platelet dysfunction due to uremia

372
Q

What is the anemia associated with liver failure

A

Macrocytic due to lipid abnormalities which cause RBCs to acquire phospholipid and cholesterol as they circulate

373
Q

What endocrine disorder is associated with anemia

A

Hypothyroidism - normochromic normocytic anemia

374
Q

What is polycythemia

A

High red cell count with increase in hemoglobin; can be relative when there is hemoconcentration due to decreased plasma volume or increase in total red cell mass

375
Q

What can cause relative polycythemia

A

Dehydration; stress polycythemia (gaisbock syndrome); usually hypertensive, obese, and anxious

376
Q

What is absolute polycythemia

A
  • primary: results from intrinsic abnormality of hematopoietic precursors
  • secondary: when red cell progenitors are responding to increased levels of EPO
377
Q

What is the most common cause of primary polycythemia

A

Polycythemia Vera

378
Q

What familial mutations can cause primary polycythemia

A

Mutations in EPO receptor that induce EPO-independent receptor activation

379
Q

What are causes of secondary polycythemia

A

EPO secreting tumors; inherited defects that lead to stabilization of HIF-1alpha

380
Q

What is prothrombin time

A

PT; assesses extrinsic and common coagulation pathways clotting of plasma after addition of exogenous source of tissue thrombophlebitis and Ca2+ ions is measured; prolonged PT can result from def of factor V, VII, X, prothrombin or fibrinogen

381
Q

What is partial thromboplastin time

A

PTT; assesses intrinsic and common clotting pathways; clotting of plasma after addition of kaolin, cephalon and Ca2+ ions measured; kaolin activates factor XII and cephalon substitutes for platelet phospholipids; prolongation of PTT results from dysfunction of factors V, VIII, IX, X, XI, XII, prothrombin or fibrinogen

382
Q

What are the absolute secondary cause of polycythemia

A

*high EPO (primary has low EPO); lung dz, high altitude living, cyanotic heart dz, paraneoplastic (RCC, HCC, cerebellar hemangioblastoma), hemoglobin mutants with high O2 affinity, chuvash polycythemia (VHL mutations), prolyl hydroxylase mutations

383
Q

What do bleeding disorders caused by vessel wall abnormalities usually present as

A

Small hemorrhages (petechiae and purpura) in skin or mucus membranes (gingiva) - usually; but can present with bleeding into joints, mm, subperiosteal locations or menorrhagia, nosebleeds GI bleeds or hematuria; platelet counts and PT PTT are normal

384
Q

What are the conditions in which abnormalities in the vessel wall cause bleeding

A
  • infections: petchial and purpura hemorrhages; *meningoccocemia, infective endocarditis and rickettsioses
  • drug reactions drug induced immune complexes deposit in vessel walls which leads to hypersensitivity (leukocytoclastic) Vasculitis
  • Scurvy and ehlers Danlos: microvascular bleeding bc weak vessel walls; cushing syndrome (protein wasting effects of corticosteroids production causes loss of perivascular supporting tissue)
  • henoch-schonlein purpura: systemic immun disorder; colicky ab pain, polyarthralgia and acute glomerulonephritis; immune comple deposition in vessels and mesangial regions
  • hereditary hemorrhagic telangiectasia (weber-Osler-rendu): AD; mutations that modulate TGFbeta signaling; dilated, tortuous blood vessels with thin walls; most common bleeding under mucous membranes of nose, tongue, mouth, and eyes and GI tract
  • perivascular amyloidosis: weaken vessel walls and cause bleeding; mostly with AL amyloidosis - mucocutaneous petechiae
385
Q

What vessel wall abnormalities is most associated with serious bleeding

A

Hereditary hemorrhagic telangiectasia

386
Q

What is the PT and PTT in thrombocytopenia

A

Normal

387
Q

Where are hemorrhages most likely as a result of thrombocytopenia

A

Small vessels; skin and mucous membranes of GI and GU tracts; intracranial bleeding*

388
Q

What are the categories of causes of thrombocytopenia

A
  • decreased platelet production: depress marrow (aplastic anemia and leukemia) drugs and alcohol;; HIV myelodysplastic syndrome
  • decreased platelet survival: immune thrombocytopenia (destruction caused by deposition of abs on platelets - alloabs an arise when platelets are transfused or cross placenta) non immune (DIC and thrombotic microangiopathies; also mechanical heart valves)
  • sequestration
  • dilution: massive transfusions can produce dilutional thrombocytopenia
389
Q

What is chronic immune thrombocytopenia purpura

A

Caused by auto ab mediated destruction of platelets; secondary forms occur in SLE, HIV, B cell neoplasms (CLL)

390
Q

What is the pathogensis of chronic immune throbocytopenic purpura

A

Auto abs against platelet membrane glycoproteins (IIb-IIIa or Ib-IX); most are IgG - act as opsonins and are phagocytosed; improved by splenectomy

391
Q

What is the morphology of chronic ITP

A

Spleen is normal size; marrow reveals modestly increased number of megakaryocytes; peripheral blood reveals abnormally large platelets

392
Q

What are the clinical features of chronic ITP

A

Adults women <40; bleeding into skin and mucosa; pinpoint hemorrhages (petechiae) ecchymoses; easy bruising, nosebleeds, bleeding into gums; may manifest as melena, hematuria or excessive menstrual low; *subarachnoid and intracerebral hemorrhage fatal complications but are rare; no splenomegaly or LAD; low platelets increased megakaryocytes; PT and PTT normal; respond to glucocorticoids; also can give anti-CD20 (rituximab); TPO mimetic can increase platelet production

393
Q

What is acute immune thrombocytopenic purpura

A

Autoabs to platelets *disease of childhood occurring equally in both sees; sx appear after viral illness; self limited resolves in 6 months

394
Q

What drugs are most commonly implicated in drug-induced thrombocytopenia

A

Quinine, quinidine, vancomycin - bind platelet glycoproteins to create antigenic determinants that are recognized by abs; also platelet inhibitory drugs that bind glycoproteins IIb-IIIa

395
Q

What is heparin induced thrombocytopenia

A
  • type I: rapidly after onset of therapy; little clinical important most likel a result of direct platelet aggregating effect of heparin
  • type II: less common but more severe; 5-14 days after therapy leads to life threatening venous and arterial thrombosis; caused by abs that recognize complexes of heparin and platelet factor 4-> activates platelets and promotes thrombosis; risk is lowered with use of low molecular weight heparin
396
Q

What is HIV associated thrombocytopenia

A

Impaired platelet production and increased destruction; CD4 and CXCR4 (receptor and coreceptor for HIV) are found on megakaryocytes which allows HIV to infect them - prone to apoptosis; also causes B cell hyperplasia which predisposes to production of autoabs

397
Q

What is thrombotic microangiopathy

A

Includes TTP and HUS; caused by insults that lead to excessive activation of platelets which deposit as thrombi in small bv

398
Q

What is TTP

A

Fever, thrombocytopenia, microangiopathic hemolytic anemia, transient neuro deficits and renal failure

399
Q

What is HUS

A

Microangiopathic hemolytic anemia and thrombocytopenia but NO neuro defects, prominent acute renal failure, occurs frequently in children

400
Q

What makes TTP and HUS different from DIC

A

Activation of coagulation cascade is not of primary importance *PTT and PT are normal

401
Q

What are the causes of TTP

A

Deficiency of ADAMTS13 - inherited or acquired

402
Q

What are the causes of HUS

A
  • typical: E. coli O157:H7 (endothelial damage by shiga like toxin)
  • atypical: alternative complement pathway inhibitor deficiency (complement factor H, membrane cofactor protein CD46 or factor I - inherited) (autoabs - acquired)
  • other assoc: drugs (cyclosporine, chemo) radiation, BM transplant, HIV, pneumococcal sepsis, SLE, lymphoid neoplasms
403
Q

What is another name for ADAMTS13

A

VWF metalloprotease - normally degrades very high molecular weight multiverses of vWF; in its absence these multiverses accumulate in plasma and promote platelet activation and aggreagation

404
Q

When does inherited TTP present

A

Adolescence; symptoms are episodic; other factors (superimposed vascular injury or prothrombotic state) must be present to trigger full blown TTP

405
Q

How is TTP treated

A

Plasma exchange (removes autoabs and provides functional ADAMTS13)

406
Q

What is the atypical HUS associated with

A

Relapsing and remitting course

407
Q

How is HUS treated

A

Typical is treated supportively

408
Q

What are the classifications of inherited disorders of platelet function

A

Defects in adhesion, defects of aggregation and disorders of platelet secretion

409
Q

What is Bernard shoulder syndrome

A

Defective adhesion of platelets to subendothelial matrix; AR caused by deficiency of platelet membrane glycoproteins complex Ib-IX (receptor for vWF); affected patients have severe bleeding tendency

410
Q

What is glanzmann thrombocytopenia

A

Defective platelet aggregation; AR; fail to aggregate in response to ADP, collagen, epi or thrombin bc of deficiency of glycoproteins IIb-IIIa (integrin that participates in bridge formation btw platelets by binding fibrinogen); severe bleeding tendency

411
Q

What are the disorders of platelet secretion

A

Characterized by defective release of mediators of platelet activation such as thromboxanes and granule bound ADP; called storage pool disorders

412
Q

What are the acquired defects in platelet function

A

Aspirin and uremia

413
Q

How does bleeding due to isolated coagulation factor deficiencies manifest

A

Large posttraumatic eccymoses or hematomas OR prolonged bleeding after a laceration; often occurs into GI and GU t facts Nd into weight bearing joints (hemarthrosis)

414
Q

What factor is deficiency in hemophilia A

A

VIII

415
Q

What factor is deficient in hemophilia B

A

IX

416
Q

What does vitamin K deficiency result in impaired synthesis of

A

Factors II, VII, IX, X and protein C; will also be deficient in these factors with severe liver dz

417
Q

What are the 2 most common inherited disorders of bleeding

A

Hemophilia A and von willebrand dz

418
Q

What is factor VIII a cofactor for

A

factor IX (which converts X to Xa)

419
Q

What is the relationship btw factor VIII and vWF

A

Factor VIII binds vWF which stabilizes factor VIII

420
Q

How is vWF measured

A

Ristocetin agglutination test - mix patients plasma with formalin-fixed platelets and ristocetin (activates vWF and induces it to bind glycoproteins Ib-IX and form bridges)

421
Q

What is the most common inherited bleeding disorder

A

Von willebrand; mild bleeding t end envy; unnoticed until Hemostasis stress (surgery or dental procedure); most common sx - spontaneous bleeding from mucous membranes, excessive bleeding from wounds, menorrhagia; AD but some AR exist

422
Q

What are the categories of Von willebrand dz

A
  • type I and III: assoc with quantitative defects in vWF; type I is AD: mild to moderate deficiency *most common type 3 is AR associated with very low levels of vWF and has severe manifestations (deletion or framshift in both alleles)
  • type 2: qualitative defects in vWF; 2A is most common of these; AD; vWF in normal amounts but missense mutations present that lead to defective mutlimer assembly - mild to moderate bleeding
423
Q

What happens to platelets in vWF

A

Normal count by have defects in platelet function; will have secondary decrease in factor VIII bc cannot be stabilized by vWF(prolongation of PTT in type 1 and 3) but bleeding into joints not seen

424
Q

How can you treat von willebrand dz

A

Desmopressin (stimulates vWF release) before surgery; or infusions of plasma concentrations containing factor VIII and vWF

425
Q

What is the most coon hereditary dz assoc with life threatening bleeding

A

Hemophilia A (Factor VIII def); some can have no family hx; most severe deficiencies result from inversion involving the X chrom that completely abolishes synthesis o factor VIII

426
Q

Are petechiae seen in hemophilia A

A

No

427
Q

What are the lab values for hemophilia A

A

Prolonged PTT but normal PT (abnormality in intrinsic pathway)

428
Q

How is hemophilia A treated

A

Infusions of recombinant factor VIII; some with severe dz develop abs that bind and inhibit factor VIII bc perceived as foreign

429
Q

What are the lab values or hemophilia B

A

Prolonged PTT;; Normal PT

430
Q

How is clotting initiated

A

Exposure of tissue factor which combines factor VII to activate factor X and IX; activation of factor X leads to generation of thrombin; thrombin converts fibrinogen to fibrin and feedsback to activate factors IX, VIII, and V, and stimulates fibrin crosslinking

431
Q

How is clotting stopped in areas that dont need it

A

As thrombin encounters uninsured vessels it is converted to anticoagulant through bindin to thrombomodulin -> activates protein C which inhibits factor V and VIII

432
Q

What 2 major mechanisms trigger DIC

A
  • Release of tissue actor or other poorly characterized procoagulants into circulation
  • widespread injury to endothelial cells
433
Q

What can the procoagulants that cause DIC be derived from

A

Placenta in obstetric complications or tissues injured by burns or trauma; mucus released by adenocarcinoma can act as procoagulants by directly activated factor X

434
Q

What is implicated in DIC of sepsis

A

TNF; induces endothelial cells to express tissue factor and to decrease expression of thrombomodulin; also upregulates adhesion molecules which can damage endothelial cells by ROS

435
Q

What cancers are most commonly associated with DIC

A

APL and adenocarcinoma of lung, pancreas, colon and stomach

436
Q

What are the consequences of DIC

A
  • widespread deposition of fibrin within microcirculation l eating to ischemia and microangiopathic hemolytic anemia
  • consumption of platelets and clotting factors and activations of plasminogen leading to hemorrhagic diathesis; plasmin cleaves fibrin and digests factors V and VIII
437
Q

What is the morphology of DIC

A

Thrombi found in brain, heart, lungs, kidneys, adrenals spleen and liver; can have bl renal cortical necrosis; PE and fibrin exudation crating hyaline membranes; can cause Waterhouse friderichsen syndrome; unusual form of DIC occurs in assoc with giant hemangiomas (kasabach-Merritt syndrome) in which thrombi form within the neoplasm

438
Q

What are the clinical features of DIC

A

Can be fulminant (endotoxic shock or amniotic fluid embolism) or insidious ad chronic (carcinomatosis or retention of dead fetus); most are obstetric patients and tends to be reversible with delivery of fetus; *microangiopathic hemolytic anemia, dyspnea, cyanosis, resp failure, convulsions, coma, oliguria, renal failure, sudden or progressive circulatory shock; acute - tends to be bleeding; chronic tends to be thrombotic

439
Q

What is the most common complication of blood transfusion

A

Febrile nonhemolytic reaction: fever and chills within 6 hours of a transfusion of red cells or platelets; caused by inflammatory mediators derived from donor leukocytes; respond to antipyretics

440
Q

What allergic reactions can occur as a result of blood transfusions

A

Occurs when blood products containing certain antigens are give to previously sensitized recipients; most likely occur in patients with IgA def* - triggered by IgG abs that recognize IgA in infused blood product
-urticarial allergic reactions may be triggered by presence of allergen in donated blood that is recognized by IgE abs in recipient; most common but generally mild; respond to antihistamines

441
Q

What causes acute hemolytic reaction to blood transfusions

A

IgM abs against donor red cells that fix complement; lead to hemoglobinuria; fever shaking and chills and flank pain appear rapidly; direct Coombs is positive unless all donor cells Lysenko; can progress to DIC, shock, acute renal failure or death

442
Q

What causes delayed hemolytic reactions of blood transfusions

A

Abs that recognize red cell antigens that the recipient was sensitized to previously; caused by IgG abs to foreign protein antigens and are assoc with positive direct Coombs test, low haptoglobin and elevated LDH; abs to Rh, Kell, and Kidd induce sufficient complement activation to cause severe and fatal reactions identical to those resulting from ABO mismatches; other abs that do not fix complement result in red cell opsonization, extravascular hemolysis and spherocytosis - minor

443
Q

What is transfusion related lung injury

A

Severe and frequently fatal complication; factors in transfused blood product trigger the activation o neutrophils in lung microvasculature; occurs more frequently in people with preexisting lung dz; *most common abs are those that bind MHC antigens (usually class I); often found in multiparous women; more likely in transfusions that contain fresh frozen plasma and platelets presentation: sudden onset resp failure during or soon after transfusion; diffuse bl pulm infiltrates that do not respond to diuretics; fever, hypotension and hypoxemia; treatment is supportive

444
Q

What are infectious complications of transfusions

A

Bacterial and viral

Bacterial more common from skin - more common in platelet preparations bc must be stored at room temp

445
Q

What are the treatments for anemia of chronic renal disease

A

Epoetin Alfa, hydryoxyurea, eculizumab

446
Q

What are the drugs for neutropenia

A

Filgrastim, pegfilgrastim, sargramostim, plerixafor

447
Q

What are the drugs for thrombocytopenia

A

Oprelvekin, Romiplastin, eltrombopag

448
Q

What are the causes of microcytic anemia (Wolff question)

A

Iron deficiency, anemia of inflammation, copper deficiency, lead poisoning, congenital sideroblastic anemia’s

449
Q

What is iron therapy

A
  • oral: 200-400 mg ferrous per day in 2-3 divided doses with only water/juice (food inhibits absorption); not enteric coated and not sustained release; causes N, constipation, anorexia, heartburn, vomiting, and diarrhea and dark stools
  • parenteral (colloid) iron: required if iron malabsorption, intolerance of oral therapy, noncompliance; iron dextran, sodium ferric gluconate and iron-sucrose and ferumoxytol; Side effects same as oral
450
Q

What is acute iron toxicity

A

Seen in young children who accidentally ingest iron tablets; sx -> necrotizing gastroenteritis with vomiting, ab pain, bloody diarrhea leading to shock lethargy and dyspnea; severe Met acidosis, coma and death; *treat its bowel irrigation and parenteral deferoxamine (chelating) DONT use charcoal

451
Q

What is a source of B12 for vegetarians

A

Fortified breakfast cereals

452
Q

How much B12 do you need per day

A

2 micrograms

453
Q

What is the effect of nitrous oxide on B12

A

Inhaled for analgesia; inactivates cyanocobalamin

454
Q

What is the treatment of B12 def

A
  • oral B12 even if pernicious anemia

- parenteral therapy used if neuro sx are present

455
Q

What can high doses of folate therapy cause

A

Hypotension and hypoglycemia

456
Q

What does epoetin alpha do

A

Stimulates erythropoiesis; increases reticulocyte count and RBC count, Hb, and Hct; used for anemia due to chronic kidney dz, cancer chemo, zidovudine treatment for HIV; administered IV or SQ; incrased risk of thrombosis, pain

457
Q

What does hydroxyurea do

A

Targets ribonucleotide reductase - S cycle arrest; boosts level of HbF; lowers concentration of HbS; administered orally; toxicities - cough or hoarseness, fever, lower back or side pain, painful or difficult urination

458
Q

What is eculizumab

A

Monoclonal ab that binds to C5 and inhibits cleavage to C5a and C5b inhibits complement mediated intravascular hemolysis in PNH and atypical HUS; given IV for 4 weeks; toxicities: infections (must give meningococcal vaccine before use), URI, MSK pain, anemia, leukopenia, HTN, HA, insomnia, UTI

459
Q

What are the presenting sx of neutropenia

A

Low grade fever, sore mouth, odynophagia, gingival pain and swelling, skin abscesses, recurrent sinusitis and otitis, sx of pneumonia, perirectal pain

460
Q

What is Neutropenic fever

A

Life threatening complication of chemo; >101; <21 points is high risk for febrile neutropenia

461
Q

What is filgrastim

A

G-CSF; regulates production of neutrophils; used to decrease incidence of infection in patients with nonmyeloid malignancies receiving myelosuppressive chemo or BM transplant; used to mobilize cells into periphery for collection; administer IV or SQ 24 hrs after chemo; can cause bone pain, splenic rupture and ARDS; pegfilgrstim is longer lasting

462
Q

What is sargamostim

A

GM-CSF; increases production of neutrophils, eosinophils Nd monocytes; used to accelerate recovery of myeloid cells after autologous BM transplant; used following chemo in patients >55 with AML to decrease incidence of infections; given IV or SQ; can cause gasping syndrome in premature infants; fluid retention, sequestration of granulocytes, SVT

463
Q

Is filgrastim or sargramostim more likely usd

A

Filgrastim has fewer effects so usually wins; use only if incidence of febrile neutropenia is estimated at > 20% (primary prophylaxis) only use i delay or reduction of chemo would prevent full doses of potentially curative chemo (secondary prophylaxis)

464
Q

What is plerixafor

A

Partial agonist of CXCR4 receptor (homing of HSC to BM); mobilizes HSC from BM to plasma; used in patients who dont mobilize SC for transplant with G-CSF; used in patients with lymphoma and multiple myeloma; SQ; hypersensitivity reaction; has potential to mobilize leukemia cells

465
Q

What drugs cannot be used for thrombocytopenia

A

Thrombopoietin (autoabs that caused severe thrombocytopenia); stem cell factor (found on mast cells and causes severe allergic reactions)

466
Q

What are the megakarytocyte growth factors used for thrombocytopenia

A
  • IL-11: oprelvekin: activates cell surface cytokine receptors to stimulate growth of multiple lymphoid and myeloid cells; used for prevention of thrombocytopenia in patients receiving chemo; can cause hypokalemia and bloodshot eyes
  • romiplostim: recombinant thrombopoietin - peptides that link to ab fragments; activates MPL thrombopoietin receptor to increase platelet count; used for patients with chronic immune thrombocytopenia purpura (ITP) who have insufficiency response to corticosteroids or splenectomy
467
Q

What is eltrombopag

A

Oral non-peptide TPO receptor agonist; increases platelet count in health individuals, ITP and hep C; used for ITP and hep C; side effects: hepatotoxicity when in combo with IFN and ribavirin

468
Q

What drugs cause hemolytic anemia

A

Cephalosporins most common (ceftriaxone and cefotetan); penicillin (esp piperacillin); Dapson, levodopa, levofloxacin, methyldopa, nitrofurantoin, NSAIDs, phenazypyridine, quinidine

469
Q

What causes drug induced thrombocytopenia

A

Immune: heparin

Non immune: quinidine, furosemide, NSAIDs, penicillin, sulfonamides, ranitidine, gold

470
Q

What are the alkylating agents

A
  • bisamines: cyclophosphamide, mechlorethamine, melphalan chlorambucil
  • nitrosoureas: carmustine, streptozocin
  • aziridines: thiotepa
  • alkylsulfonate: busulfan
  • non-classic: procarbazine, dacarbazin, bendmustine
  • platinum analogs: cisplatin, carboplatin, oxaliplatin
471
Q

What are the antimetabolites

A
  • antifolates: methotrexate, pemetrexed, pralatrexate
  • fluoropyrimidines: 5FU, capecitbine, TAS-102
  • deoxycytidine analogs: cytarabine and gemcitabine
  • purine antagonists: 6 thiopurines, fludarabine and cladribine
472
Q

What natural products are used for cancer

A
  • vinca alkaloids: vinblastine, vincristine, vinorelbine
  • taxanes and other anti-microtubule: paciltaxel, docetaxel, carbazitaxel, ixabepilone, eribulin
  • epipodophyllotoxins: etoposide
  • camptothecins: topotcan, irinotecan
473
Q

What are the anti-tumor abx

A
  • anthracyclines: doxorubicin, daunorubicin, idarubicin, epirubicin, mitoxantrone, deraxane
  • mitomycin
  • bleomycin
474
Q

What are the tyrosine kinase inhibitors

A

Imatinib, dasatinib, nilotinib, bosutinib, ponatinib

475
Q

What are the growth factor receptor inhibitors

A

-mab, erlotinib, afatininb, zip-afliberceept sorafenib, sunitinib, pazopanib

476
Q

When is primary chemo used

A

If no surgical option; Hopkins and NHL, choriocarcinoma, germ cell cancer, AML, burkitt’s wilms tumor, embryonal rhabdo, ALL

477
Q

What are the mechanisms of resistance for alkylating agents

A
  • increase in DNA repair enzymes (MGMT)
  • decreased transport of drug
  • increased expression of glutathione
478
Q

What are the mechanisms of resistance of antimetabolites

A
  • inhibition of metabolism into active metabolites (cytarabine)
  • decreased drug transport
  • decreased formation of polyglutamate metabolites by folyl polyglutamate synthase (needed for methotrexate, pemetrexed and pralatrexate)
479
Q

What are the drugs with a MOA disruption of tubulin polymerization

A

Vinblastine, vincristine, vinorelbine

480
Q

Which drugs enhance tubulin polymerization

A

Paclitaxel, docetaxel, cabazitaxel

481
Q

What are the topoisomerase inhibitors

A

I: topetecan and irinotecan
II: etoposide

482
Q

Which natural product has neurotoxicity as a side effect

A

Vincristine

483
Q

Which natural products have hypersensitivity as a side effect

A

Paclitaxel and docetaxel

484
Q

Which natural product has diarrhea as a side effect

A

Topotecan and irinotecan

485
Q

What is the MOA of antitumor abx

A
  • inhibition of topoisomerase 2, generation of free radicals - doxorubicin
  • induce DNA cross links: mitomycin
  • DNA fragmentation and strand breaks due to free radical formation: bleomycin
486
Q

What are the mechanisms of resistance of antitumor abx

A

Point mutations in topoisomerase II (doxo), increased expression of MDRP (doxo and mitomycin), upregulation of bleomycin hydrolase (inhibits bleomycin iron binding)

487
Q

What antitumor abx has an adverse effect of blue discoloration of nails, sclera and urine

A

Mitoxantrone

488
Q

What are the immune checkpoint inhibitors

A
  • PD-1 inhibitors: Nivolumab and pembrolizumab - used for melanoma, NSCLC, Hodgkin
  • PD-L1 inhibitors: atezolizumab, avelumab, durvalumab: used for bladder cancer, NSCLC, Merle cell skin cancer
489
Q

What is the treatment for ALL

A

one of: 6 mercaptopurine, cyclophosphamide, vincristine, daunorubicin

Combo of vincristine and prednisone with one above agent used to induce remission

490
Q

What is used to prevent CNS leukemia (ALL)

A

Intrathecal Methotrexate

491
Q

What is the treatment for AML

A

Cytarabine in combo with anthracycline - most active
-idarubicin is preferred

During period of induction do platelet transfusion to prevent bleeding, GSF,, abx

492
Q

What do you do after remission is achieved in AML

A

High dose cytarabine or HSC transplant

493
Q

What is the first line therapy for CML

A

Imatinib
Dasatinib and nilotinib
Busulfan also effective

494
Q

What is the treatment for CLL

A

Chlorambucil (with prednisone), cyclophosphamide

COP: cyclophosphamide, vincristine, prednisone
CHOP: cyclophosphamide, vincristine, prednisone, doxorubicin
Bendamustine also approved
Fludarabine also effective alone or in combo with cyclyophosphamide or mitoxantrone or rituximab

495
Q

What are the treatments for stage I and IIa Hodgkin

A

ABVD: doxorubicin, bleomycin, vinblastine, dacarbazine

496
Q

What are the treatments for stage III and IV Hodgkin

A

MOPP: mechlorethamine, vincristine, procarbazine and prednisone
ABVD: doxorubicin, bleomycin, vinblastine, decarbazine
Stanford V: doxorubicin, vinblastine, mechlorethamine, vincristine, bleomycin, etoposide and prednisone

497
Q

What is the treatment for NHL (Diffuse)

A

CHOP: cyclophosphamide, doxorubicin, vincristine and prednisone

Or R CHOP

498
Q

What is the treatment for nodular (follicular) NHL

A

Bedamustine and rituximab

499
Q

What is the treatment for multiple myeloma

A

Melphalan and prednisone

500
Q

What is the treatment for breast cancer

A

Stage I: surgery

stage II: CMF - cyclophosphamide, methotrexate, 5FU; FAC: 5FU, doxorubicin, cyclophosphamide

501
Q

What is the treatment for prostate cancer

A

Mitoxantrone and prednisone

*docetaxel and prednisone is standard of care

502
Q

What is the treatment for colorectal cancer

A
  • FOLFOX: leucovorin, 5FU and oxaliplatin

- XELOX: capecitabine and oxaliplatin

503
Q

What is the treatment for metastatic colorectal cancer

A
  • FOLFIRI: leucovorin, 5FU, irinotecan; ziv-aflibercept added if progression observed with oxaplatin based chemo
  • FOLFOX or FOLFIRI with bevacizumab or cetuximab/panitumumab
  • TAS-102
504
Q

What is the treatment for NSCLC

A

Bevacizumab in combo with carboplatin and paclitaxel with non-squamous histo

  • if squamous - cisplatin or carboplatin with cetuximab
  • maintenance chemo: pemetrexed after stabilized with 4 cycles of platinum based chemo
505
Q

What is the treatment of NSLCL with molecular testing available

A
  • erlotinib: first line with EGFR mutations
  • afatinib: metastatic NSCLC with EGFR deletions
  • osimertinib: met EGFR
506
Q

What is the treatment for SCC of the lung

A

Cisplatin and gemcitabine with necitumumab

-nivolumab used for cancers that have progressed after standard treatment

507
Q

What is the treatment for small cell lung cancer

A
  • cisplatin + etoposide or cisplatin + irinotecan

- topotecan is second line if failed

508
Q

What is the treatment for ovarian cancer

A
  • stage I: radiotherapy; cisplatin and cyclophosphamide
  • stage III and IV: carboplatin and paclitaxel
  • recurrent: topotecan or doxorubicin
509
Q

What is the treatment for testicular cancer

A

PEB: cisplatin, etoposide, bleomycin

510
Q

What is the treatment for malignant melanoma

A

Dacarbazine, temozolomide, cisplatin, IFN alpha, IL-2

-nivolumab, pembrolizumab for unresectable or met

511
Q

What is the treatment for brain cancer

A

Carmustine
PCV: procarbazine, lomustine, vincristine
-temozolomide for glioblastoma
-bevacizumab alone or in combo with chemo