You are on page 1of 24

Pathology Diseases of White Blood Cells, Lymph Nodes, Spleen, & Thymus

AY 2018-2019 Dr. HA Saguil


1st Shifting Exam 03/23/2019

OUTLINE daughter cell


I. Development and Maintenance of Hematopoietic Tissues (p.1) → The newly committed cell lose the capacity for self renewal
and leads to terminal differentiation and death
II. Disorders of White Blood Cells and Lymph Nodes (p.1)
→ As progenitors differentiate, they also proliferate more rapidly
III. Spleen (p.19) in response to growth factors
IV. Thymus (p.21) → Stem cell factor (KIT lgand) and FLT3-ligand-act through
receptors on very early committed cells
I. DEVELOPMENT AND MAINTENANCE OF → EPO, GM-CSF, G-CSF and Thrombopoeitin- act through
HEMATOPOEITIC TISSUES receptors expressed on committed progenitors
→ Feedback loops-tune marrow output for maintenance of
formed elements
Time Characteristics • Many diseases alter the production of blood cells
• First appearance of blood cell → Primary tumors-most important diseases that interfere with
progenitors marrow function
• Long lived tissue macrophages → Genetic diseases, infections, toxins and nutritional
(e.g.microglial cells and Kupffer cells)- deficiencies as well as chronic inflammation can also
derived from the yolk sac but decrease blood cells in marrow.
3rd week contribution of yolk sac to blood • Tumors of hematopoietic origin are often associated with
embryogenesis formation is only transient mutations that block progenitor cell maturation or abrogate
• Hematopoietic stem cells (HSCs)- their growth factor dependence
arise in the mesoderm of the
intraembryonic aorta/gonad/mesonephric Net effect is an unregulated clonal expression of hematopoietic
region weeks later elements replacing normal cells and spreads to other
hematopoietic tissues
• HSCs migrate to the liver
- (becomes the chief site of blood cell II. DISORDERS OF WBC AND LYMPH NODES
3rd month formation until before birth)
A. LEUKOPENIA
• HSC also reside at fetal placenta
• Usually results from reduced number of neutrophils
4th month • HSC goes to bone marrow (neutropenia, granulocytopenia)
• Marrow is haematopoietically active and → Lymphopenia- less common; most commonly observed in
Birth advanced HIV; stems from lymphocyte redistribution rather
hepatic hematopoiesis dwindles
than decrease
• Haematopoietically active marrow is
Puberty → Granulocytopenia- more common and often associated with
found throughout the skeleton, then
diminished granulocyte function
restricts to axial skeleton
• About half of the marrow space is
Adults NEUTROPENIA / AGRANULOCYTOSIS
haematopoietically active.

• Formed elements (red cells. Granulocytes, monocytes, platelets • Neutropenia can be caused by:
and lymphocytes)- have common origin from HSCs, pluripotent 1. inadequate or ineffective
cells that sit at the apex of a hierarchy of bone marrow granulopoiesis 2. increased destruction
progenitors. or sequestration of neutrophils in the
• HSCs give rise to several kinds of early progenitor cells with a periphery
restricted differentiation potential, such that they ultimately • Observed in:
produce mainly myeloid cells or lymphoid cells a. Suppression of hematopoietic
• Colony forming units (CFUs)- produce colonies composed of stem cells (anemia and infiltrative
specific kinds of mature cells when grown in culture marrow disorders)
• HSCs have two essential properties that are required for the b. Suppression of committed
maintenance of hematopoiesis: pluripotency and the capacity granulocytic precursors by
for self-renewal. exposure to certain drugs
c. Disease states assoc with
→ Pluripotency- ability of a single HSC to generate all mature
PATHOGENESIS ineffective hematopoiesis
blood cell
(megaloblastic anemia and
▪ When an HSC divides, at least one daughter cell must
myelodysplastic syndromes)
renew to avoid stem cell depletion
d. Rare congenital conditions (
→ Self-renewing divisions occur within a specialized marrow
Kostmann syndrome)
niche, in which stromal cells and secreted factors nurture and
• Occurs with
protect the HSCs
a. Immunologically mediated injury to
• The marrow response to short-term physiologic needs is
neutrophils (can be idiopathic,
regulated by hematopoietic growth factors through effects
immunologic and exposure to
on the committed progenitors
drugs)
→ Mature blood elements must continually be replenished b. Splenomegaly
→ Some HSCs give rise to multipotent progenitors-more c. Increased peripheral utilization
proliferative than HSCs but lesser self renewal capacity (bacterial, fungal, or rickettsial
→ Division of multipotent progenitors- give rise to at least one infection)

S4T2 Systemic Pathology Transers 1 of 24


• Most common cause of LYMPHADENITIS
agranulocytosis is DRUG TOXICITY • Following their initial development from precursors in the central
(primary) lymphoid organs, (the bone marrow for B cells) and
• Signs and symptoms related to (the thymus for T cells), lymphocytes circulate through the blood
infection includes malaise, chills and and, under the influence of specifc cytokines and chemokines,
fever followed by weakness and home to lymph nodes, spleen, tonsils, adenoids, and
CLINICAL
fatigability Peyer’s patches, which constitute the peripheral (secondary)
• Agranulocytosis- infections are lymphoid tissues
FEATURES
overwhelming; may cause death • Lymph nodes
within hours/days → Most widely distributed and easily accessible lymphoid tissue
• Serious infections- neutrophil → The activation of resident immune cells leads to morphologic
count <500mm3 changes in lymph nodes
→ Within several days of antigenic stimulation, the primary
• Neutropenia - reduction in number of neutrophils in blood follicles enlarge and develop pale-staining germinal centers
• Agranulocytosis- clinically significant reduction in neutrophils • Germinal Centers
• Makes individual susceptible to bacterial and fungal infections → Highly dynamic structures in which B cells acquire the
capacity to make high-affinity antibodies against specific
B. REACTIVE PROLIFERATION OF WBC AND LYMPH antigens
NODES • Paracortical T-cell zones
→ May also undergo hyperplasia
LEUKOCYTOSIS • The degree and pattern of the morphologic changes are
dependent on the inciting stimulus and the intensity of the
• Increase in the number of white cells in the blood
response
• Common reaction to a variety of inflammatory states
→ Trivial injuries - subtle changes
→ Significant infections -nodal enlargement and sometimes
• Factors that influence Peripheral
leave residual scarring
Blood Leukocyte Count:
• Lymph nodes in adults are almost never “normal” or “resting,”
o Size of the myeloid and lymphoid
and it is often necessary to distinguish morphologic changes
precursor and storage cell pools in
secondary to past experience from those related to present
bone marrow, thymus, circulation
disease
and peripheral tissues
o Rate of release of cells from
storage pools into circulation A. Acute Nonspecific Lymphadenitis
o Proportion of cells that are • Cervical region acute lympadenitis
adherent to blood vessel walls at → Most often due to drainage of microbes or microbial
any time (marginal pool) products from infections of the teeth or tonsils
o Rate of extravasation of cells from • Axillary or Inguinal regions
the blood into tissues → Most often caused by infections in the extremities
• Leukocyte hemostasis is maintained
by: • Mesenteric lymph nodes
o Cytokines → Draining acute appendicitis
o Growth factors → Other self-limited infections may also cause acute
o Adhesion molecules mesenteric adenitis and induce symptoms mimicking acute
o Through their effects on their appendicitis, a differential diagnosis that plagues the surgeon
commitment, proliferation,
differentiation and extravasation of • Systemic viral infections (particularly in children) and
leukocytes and their progenitors bacteremia often produce acute generalized lymphadenopathy
• In sepsis or severe inflammatory
PATHOGENESIS
disorders (ex. Kawasaki disease), • The nodes are swollen, gray-red and
leukocytosis is often accompanied by engorged
morphologic changes in the • Microscopically, there is prominence
neutrophils, such as toxic granulations, of large reactive germinal centers
Döhle bodies, and cytoplasmic containing numerous mitotic figures
vacuoles • With less severe reactions, scattered
o Toxic granules - coarser and neutrophils infiltrate about the follicles
darker than the normal neutrophilic and accumulate within the lymphoid
granules, represent abnormal PATHOGENESIS sinuses
azurophilic (primary) granules • Endothelial cells lining the sinuses
o Döhle bodies are patches of undergo hyperplasia
dilated endoplasmic reticulum that • Enlarged and painful nodes and the
appear as sky-blue cytoplasmic overlying skin is red
“puddles.” • Sometimes, suppurative infections
• Acute viral infections, particularly in can produce draining sinuses
children, can cause the appearance of • Healing of such lesions is associated
large numbers of activated with scarring
lymphocytes that resemble neoplastic
lymphoid cells
• In severe infections, many immature B. Chronic Nonspecific Lymphadenitis
granulocytes appear in the blood, • Lymph nodes in chronic reactions are nontender, as nodal
mimicking a myeloid leukemia enlargement occurs slowly over time and acute inflammation
(leukemoid reaction) with associated tissue damage is absent

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 2 of 24
(continuation of Chronic Nonspecific Lymphadenitis) C. NEOPLASTIC PROLIFERATIONS OF WBC
• Common in inguinal and axillary nodes, which drain relatively
large areas of the body and are frequently stimulated by
CATEGORY CHARACTERISTICS
immune reactions to trivial injuries and infections of the
extremities • B-cell, T-cell, and NK-cell origin
• Promote the appearance of organized collections of immune • Phenotype of the neoplastic
LYMPHOID
cells in nonlymphoid tissues (tertiary lymphoid organs) cell closely resembles that of a
NEOPLASMS
→ A classic example is that of chronic gastritis caused by particular stage of normal
Helicobacter pylori, in which aggregates of mucosal lymphocyte maturation
lymphocytes are seen that simulate the appearance of • Arise from early hematopoietic
Peyer patches progenitors
→ Rheumatoid arthritis, in which B- cell follicles often appear in 1. Acute myeloid leukemias
the inflamed synovium • Immature progenitor cells
→ Lymphotoxin, a cytokine required for the formation of normal accumulate in the bone marrow
Peyer patches, is probably involved in the establishment of (↑ Immature cells in marrow)
these extranodal” inflammation-induced collections of 2. Myelodysplastic syndromes
lymphoid cells • Associated with ineffective
hematopoiesis and resultant
HEMOPHAGOCYTIC LYMPHOHISTIOCYTOSIS MYELOID peripheral blood cytopenias (↓
NEOPLASMS mature cells in blood)
• Marked by cytopenias and s/sx of systemic inflammation related
to macrophage activation 3. Chronic myeloproliferative
disorders
• Also called Macrophage Activation Syndrome
• Increased production of one or
• Some forms are familial and may appear early in life, even in
more terminally
infants, while other forms are sporadic and may affect people of
differentiated myeloid elements
any age.
(e.g., granulocytes) usually
leads to elevated peripheral
blood counts (↑ mature cells in
• Systemic activation of macrophages and
blood)
CD8+ cytotoxic T cells
• Uncommon proliferative lesions
• The activated macrophages
of macrophages and dendritic
phagocytose blood cell progenitors in
cells
the marrow and formed elements in the
peripheral tissues, while the “stew” of
HISTIOCYTOSES • Langehans Cell
mediators released from macrophages Histiocytoses
and lymphocytes suppress o Langerhans cell (immature
hematopoiesis and produce symptoms dendritic cell) proliferation.
of systemic inflammation
• These effects lead to cytopenias and a ETIOLOGIC & PATHOGENIC FACTORS IN WBC
PATHOGENESIS
shock-like picture, sometimes referred to NEOPLASIA
as “cytokine storm” or the systemic
inflammatory response syndrome
• The most common trigger for HLH is
A. Chromosomal Translocations and other
infection, particularly with Epstein-Barr Acquired Mutations
virus (EBV)
• Most patients present with an acute • Mutated or altered genes often play crucial roles in the
febrile illness associated with development, growth, or survival of the normal counterpart of
splenomegaly and hepatomegaly the malignant cell.
• Hemophagocytosis is usually seen on → Certain mutations are strongly associated with specific tumor
bone marrow examination, but is neither types
suffcient nor required to make the → Mutation produces a “dominant-negative” protein that
diagnosis interferes with a normal function (a loss of function)
• Anemia, thrombocytopenia, and → In others the result is an inappropriate increase in some
CLINICAL very high levels of plasma ferritin and normal activity (a gain of function).
FEATURES soluble IL-2 receptor, • Oncoproteins created by genomic aberrations often block
both indicative of severe normal maturation, turn on pro-growth signaling pathways, or
inflammation, as well as elevated protect cells from apoptotic cell death.
liver function tests and triglyceride → Many oncoproteins cause an arrest in differentiation, often at
levels, both related to hepatitis a stage when cells are proliferating rapidly.
• Coagulation studies may show → Example: In acute leukemias, dominant-negative oncogenic
evidence of disseminated mutations involving transcription factors are often present
intravascular coagulation which can that interfere with early stages of lymphoid or myeloid cell
lead to death differentiation.
• “Mild” chemotherapy → Other mutations in transcriptional regulators seem to directly
• Patients with germline mutations that enhance the self-renewal of tumors cells, giving such cells
TREATMENT
cause HLH or who have stem-cell–like properties.
persistent/resistant disease are → Mutations that inhibit apoptosis are prevalent in certain
candidates for hematopoietic stem hematologic malignancies
cell transplantation • Proto-oncogenes are often activated in lymphoid cells by errors
that occur during antigen receptor gene rearrangement and
diversification.
→ Oncogenic mutations occur most frequently in germinal

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 3 of 24
center B cells during attempted antibody diversification • Due to exposure to carcinogens (i.e. benzene in tobacco
→ B cells enter upregulate the expression of activation-induced smoke)
cytosine deaminase (AID)
▪ A specialized DNA-modifying enzyme that is essential for
2 types of immunoglobulin (Ig) gene modifications: LYMPHOID NEOPLASMS
▪ Class switching: an intragenic recombination event in
which the IgM heavy-chain constant gene segment is A. Definitions and Classifications
replaced with a different constant segment (e.g., IgG3),
leading to a switch in the class (isotype) of antibody
• Leukemia
produced
→ Neoplasms that present with widespread involvement of the
▪ Somatic hypermutation: which creates point mutations
bone marrow and (usually, but not always) the peripheral
within Ig genes that may increase antibody affinity for
blood.
antigen
• Lymphoma
→ AID enzyme is also responsible for some oncoprotein
translocation resulting to activation of proto-oncogenes → Used for proliferations that arise as discrete tissue masses.
▪ c-MYC →activated in germinal center B-cell lymphomas → Hodgkin lymphoma
by chromosomal translocations → Non-hodgkin lymphomas (nhls).
▪ BCL6 → activated in germinal center B-cell lymphomas by • Plasma cell neoplasms
point mutations → Most often arise in the bone marrow and only infrequently
involve lymph nodes or the peripheral blood.
B. Inherited Gene Factors • Clinical presentation of lymphoid neoplasms is determined by
the anatomic distribution of disease.
→ Two thirds of nhls and virtually all hodgkin lymphomas
• Genetic diseases that promote genomic instability and
present as enlarged nontender lymph nodes (often > 2 cm).
predispose at increased risk of acute leukemia:
→ The remaining one third of nhls present with symptoms
→ Bloom syndrome
related to the involvement of extranodal sites (e.g., skin,
→ Fanconi anemia stomach, or brain).
→ Ataxia telangiectasia → Lymphocytic leukemias have signs and symptoms related to
→ Down syndrome (trisomy 21) the suppression of normal hematopoiesis by tumor cells in
→ Type I neurofibromatosisAll caps the bone marrow.
→ Multiple myeloma causes bony destruction of the skeleton
C. Viruses and often presents with pain due to pathologic fractures.
→ Plasma cell tumors are related to secretion of Igs
Lymphotopic → Hodgkin lymphoma is associated with release of
Associated Lymphoma inflammatory cytokines.
Viruses
Human T-cell • Adult T-cell leukemia/ lymphoma.
leukemia The WHO Classification of Lymphoid Neoplasms
virus-1 (HTLV-1) • Uses morphologic, immunophenotypic, genotypic, and
clinical features
Epstein-Barr virus • Burkitt lymphoma • Grouped lymphoid neoplasms into 5 broad categories,
(EBV) • Hodgkin lymphoma (HL) which are separated according to the cell of origin:
• B-cell lymphomas
• Rare NK-cell lymphomas 1. Precursor B-cell neoplasms (neoplasms of immature
Kaposi sarcoma • Unusual B-cell lymphoma B cells)
herpesvirus/human presenting as a malignant 2. Peripheral B-cell neoplasms (neoplasms of mature B
herpesvirus-8 effusion in the pleural cavity cells)
(KSHV/HHV-8) 3. Precursor T-cell neoplasms (neoplasms of immature
T cells)
4. Peripheral T-cell and NK-cell neoplasms
D. Chronic Inflammation (neoplasms of mature T cells and NK cells)
5. Hodgkin lymphoma (neoplasms of Reed-Sternberg
cells and variants)
Agents Associated Lymphoma
H. pylori infection Gastric B-cell lymphomas
Gluten-sensitive enteropathy Intestinal T-cell lymphomas,
HIV infection B-cell lymphomas that may
arise within any organ
Severe T-cell immunodeficiency B-cell lymphomas (particularly
those associated with EBV and
KSHV/HHV-8)

E. Iatrogenic Factors
• Ironically, radiation therapy and certain forms of chemotherapy
used to treat cancer increase the risk of subsequent myeloid
and lymphoid neoplasms.
• Due to the mutagenic effects of ionizing radiation and
chemotherapeutic drugs on hematolymphoid progenitor cells.

F. Smoking
• The incidence of acute myeloid leukemia is increased 1.3- to 2-
fold in smokers

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 4 of 24
Figure 1. Origin of lymphoid neoplasms

Principles of Lymphoid Neoplasms


• Histologic examination of lymph nodes or other involved
tissues is required for diagnosis
• Antigen receptor gene rearrangement generally precedes
transformation of lymphoid cells; hence, all daughter cells
derived from the malignant progenitor share the same
antigen receptor gene configuration and sequence, and
synthesize identical antigen receptor proteins (either Igs or T-
cell receptors).
→ Analyses of antigen receptor genes and their protein
products can be used to distinguish reactive (polyclonal) and
malignant (monoclonal) lymphoid proliferations.
• Most lymphoid neoplasms resemble some recognizable
stage of B- or T-cell differentiation, a feature that is used in
their classification.
• The vast majority (85% to 90%) of lymphoid neoplasms are of
B-cell origin, with most of the remainder being T-cell tumors;
only rarely are tumors of NK cell origin encountered.

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 5 of 24
• Lymphoid neoplasms are often associated with immune
abnormalities.
o Loss of protective immunity (susceptibility to
infection) and a breakdown of tolerance • Morphology:
(autoimmunity) can be seen, sometimes in the same → In leukemic presentations, the marrow is hypercellular and
patient.
packed with lymphoblasts, which replace the normal
o Individuals with inherited or acquired
immunodeficiency are at high risk of developing marrow elements.
certain lymphoid neoplasms, particularly those → Mediastinal thymic masses (50% to 70% of T-ALLs) are
caused by oncogenic viruses (e.g., ebv). also more likely to be associated with lymphadenopathy and
• Neoplastic b and t cells tend to recapitulate the behavior of splenomegaly.
their normal counterparts. → Scant basophilic cytoplasm and nuclei somewhat larger than
→ Like normal lymphocytes, neoplastic b and t cells home to those of small lymphocytes
certain tissue sites, leading to characteristic patterns of → Nuclear chromatin is delicate and finely stippled, and nucleoli
involvement. are usually small and often demarcated by a rim of
→ For example, follicular lymphomas home to germinal centers condensed chromatin.
in lymph nodes, whereas cutaneous t-cell lymphomas home → Nuclear membrane is deeply subdivided, imparting a
to the skin. Like their normal counterparts, particular convoluted appearance.
adhesion molecules and chemokine receptors govern the → Interspersed macrophages ingesting apoptotic tumor cells
homing of the neoplastic lymphoid cells. imparts a “starry sky” appearance
→ Exceptions to this rule include: → Because of their different responses to chemotherapy, ALL
▪ Hodgkin lymphomas, which are sometimes restricted to must be distinguished from acute myeloid leukemia (AML)
one group of lymph nodes ▪ AML is a neoplasm of immature myeloid cells that can
▪ Marginal zone B-cell lymphomas, which are often cause identical signs and symptoms.
restricted to sites of chronic inflammation. ▪ Compared with myeloblasts, lymphoblasts have more
condensed chromatin, less conspicuous nucleoli, and
• Hodgkin lymphoma spreads in an orderly fashion, whereas smaller amounts of cytoplasm that usually lacks granules.
most forms of NHL spread widely early in their course in a ▪ Definitive diagnosis still relies on stains performed with
less predictable fashion. antibodies
→ Lymphoma staging is of most useful in guiding therapy in ▪ specific for B- and T-cell antigens
Hodgkin lymphoma. → Histochemical stains are also helpful, in that (in contrast to
myeloblasts) lymphoblasts are myeloperoxidasenegative and
B. PRECURSOR B- AND T- CELL NEOPLASMS often contain periodic acid-Schiff–positive cytoplasmic
material.
Diagnosis Cell of Genotype Salient
Origin Clinical • Pathogenesis
Features → Up to 70% of T-ALLs have gain-of-function mutations in
B-cell acute Bone Diverse Predominantly
NOTCH1, a gene that is essential for T-cell development.
lymphoblastic marrow chromosomal children;
leukemia/lym precursor translocations; symptoms → high fraction of B-ALLs have loss-of-function mutations in
phoma* B cell t(12;21) relating to genes that are required for B-cell development, such as
involving marrow PAX5, E2A, and EBF, or a balanced t(12;21) involving the
RUNX1 and replacement genes ETV6 and RUNX1, two genes that are needed in very
ETV6 and early hematopoietic precursors.
present in pancytopenia;
→ mutations disturb the differentiation of lymphoid precursors
25% aggressive
T-cell acute Precursor T Diverse Predominantly → promote maturation arrest→ induce increased self-
lymphoblastic cell (often chromosomal adolescent renewal, a stem cell–like phenotype.
leukemia/lym of thymic translocations, males; thymic → single mutations are not sufficient to produce ALL. (Early
phoma origin) NOTCH1 masses and returns suggest that fewer than 10 mutations are sufficient to
mutations variable bone produce full-blown ALL)
(50%-70%) marrow
→ Approximately 90% of ALLs have numerical or structural
involvement;
aggressive chromosomal changes.
→ Most common is hyperploidy (>50 chromosomes), but
Acute Lymphoblastic Leukemia/Lymphoma (ALL) hypoploidy and a variety of balanced chromosomal
translocations are also seen.
• Neoplasms composed of immature B (pre-B) or T (pre-T)
cells, which are referred to as lymphoblasts.
• Immunophenotype
• B-ALLs (85%): manifest as childhood acute “leukemias.”
→ Immunostaining for terminal deoxynucleotidyl-transferase
• T-ALLs (~15%) tend to present in adolescent males as thymic
(TdT) (specialized DNA polymerase expressed only in pre-B
“lymphomas.”
and pre-T cells)
• Most common cancer of children.
• 3 times more common in whites than black Markers
• More frequent in boys than girls B-ALLs Very immature B-ALLs
• Hispanic – highest incidence of any ethnic group • CD19, PAX5
• Less frequently in adults of all ages Mature “late pre-B” ALLs

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 6 of 24
• CD10, CD19, CD20,
IgM heavy chain (μ • Prognosis
chain)
→ With aggressive chemotherapy about 95% of children with
T-ALLs More immature T-ALLs
• CD1, CD2, CD5, CD7 ALL obtain a complete remission, and 75% to 85% are
“Late” pre-T ALLs cured.
• CD3, CD4 CD8 → remains the leading cause of cancer deaths in children, and
only 35% to 40% of adults are cured.
• Clinical Features → Factors for worse prognosis:
→ Abrupt stormy onset within days to a few weeks of the first ▪ <2 years old (strong association of infantile ALL with
symptoms translocations involving MLL gene)
→ Symptoms related to depression of marrow function ▪ Presentation in adolescence or adulthood
▪ fatigue due to anemia ▪ Peripheral blood blast counts >100,000 (reflects high
▪ feversecondary to neutropenia tumor burden)
▪ bleeding due to thrombocytopenia → Factors for favorable prognosis:
→ Mass effects caused by neoplasticinfiltration ▪ 2-10 years old
▪ bone pain resulting from marrow expansion and infiltration ▪ Low white cell count
of the subperiosteum ▪ Hyperdiploidy
▪ generalized lymphadenopathy ▪ Trisomy of chromosomes 4, 7, 10
▪ splenomegaly ▪ Presence of t(12;21)
▪ hepatomegaly → Philadelphia chromosome T(9;22) creates a fusion gene →
▪ testicular enlargement constitutively active BCR-ABL tyrosine kinase (stronger
▪ In T-ALL, complications related to compression of large tyrosine activity in B-ALL than CML)
vessels and airways in the mediastinum ▪ Treatment: BCR-ABL kinase inhibitor + conventional
→ Central nervous system manifestations chemotherapy
▪ headache ▪ Older adults cannot tolerate very intensive chemotherapy
▪ vomiting that are curative in children
▪ nerve palsies resulting from meningeal spread
C. PERIPHERAL B-CELL NEOPLASMS

Chronic Lymphocytic Leukemia (CLL) and Small


Lymphocytic Lymphoma (SLL)
• Differ only in degree of peripheral blood lymphocytosis
• CLL: Absolute Lymphocyte count >5000cmm
→ MOST COMMON LEUKEMIA OF ADULTS IN THE
WESTERN WORLD
→ Median age of diagnosis: 60y/o
→ 2:1 Male predominance
• SLL:
→ 4% ONLY of all Non-Hodgkins’ Lymphoma

• Pathogenesis, Morphology, and Clinical Presentation


→ Most common genetic anomalies are deletions of 13q14.3, 11q,
17p, and trisomy 12q
Figure 2. Acute lymphoblastic leukemia/lymphoma → Proliferation Centers- PATHOGNOMONIC FOR CLL/SLL
→ SMUDGE CELLS- disruption of cells during smears
→ CLL/SLL disrupts normal immune function by uncertain
mechanisms (ie Hypogammaglobulinemia→predisposes to
bacterial infections)
→ Richter Syndrome-transformation to Diffuse Large B Cell
Lymphoma in 5-10% of patients

Follicular Lymphoma
• In the US, most common form of indolent NHL
• Usually presents in middle age
• Affects males and females equally
• LESS COMMON in Europe; RARE in Asian populations

• Pathogenesis, Morphology, and Clinical Presentation


→ Follicular Lymphoma likely arises from germinal center B cells,
Figure 3. Burkitt lymphoma.
and strongly associated with chromosomal translocations of
BCL2

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 7 of 24
→ In 90% of cases, there were mutations in the MLL2 gene → All endemic (AFRICAN) Burkitt Lymphomas are latently
(encodes for histone methyltransferase, regulating gene infected with EBV (also present in 25% of HIV-associated
expression) tumors, and 15-20% of sporadic cases
→ Two types of cells are present: Centrocytes (small cells with → Tumor exhibits HIGH mitotic index with numerous apoptotic
cleaved nuclear contours and little cytoplasm) and cells
Centroblasts (larger cells with open nuclear chromatin, several → Starry Sky pattern- pathognomonic for these phagocytes due to
nucleolei, and modest amts of cytoplasm) abundant cytoplasm
→ Centrocytes > Centroblasts → With bone marrow involvement: cells with royal blue
→ Bone marrow is involved in 85% of cases cytoplasm with clear cytoplasmic vacuoles
→ 30-50% of cases transform to Diffuse Large B-Cell → Both endemic and sporadic Burkitt lymphoma are found mainly
Lymphoma in children and young adults
→ Endemic BL often presents as a mass of the mandible, and
Diffuse Large B-Cell Lymphoma (DLBCL) involves kidneys, ovaries, and adrenal glands
• Most common form of NHL → Sporadic BL often involves ileocecum and peritoneum
• Slight predominance in males
• Median age is 60y/0 Plasma Cell Neoplasms and Related Disorders
• Expresses IgG, CD19 and CD20, with variable expression of • These B-Cell proliferations contain neoplastic plasma cells
CD10 and BCL6 which always secrete monoclonal Ig (M component;
myeloma) or Ig fragment, acting as tumor markers
• Subtypes • Dyscrasias- collective term for plasma cell neoplasm; accounts
→ Immunodeficiency-associated large B Cell Lymphoma- in T- for 15% of deaths by lymphoid neoplasms
cell immunodeficiency (eg in HIV) • Multiple Myeloma- most common and deadly lymphoid
→ Primary Effusion Lymphoma- malignant pleural, or ascitic neoplasm
effusion, mostly in HIV or elderly patients (HHV8 is involved) • Neoplastic cells often synthesize excess light chains along with
complete Igs, excreted in urine, termed Bence-Jones Proteins
• Pathogenesis, Morphology, and Clinical Presentation
→ Attributed to dysregulation of BCL6, a DNA-binding Zinc-finger • Clinicopathologic entities
transcriptional repressor that is required for the formation of → Multiple Myeloma
normal germinal centers → Waldenstrom Macroglobulinemia’
→ 10-20% of tumors are associated with t(14;18)→overexpression → Heavy Chain disease and Mediterranean lymphoma
of the antiapoptotic BCL2 (as in Follicular lymphoma) → Primary or immunocyte-associated amyloidosis
→ 5% of DLBCLs are associated with translocations involving → Monoclonal gammopathy of undetermined significance (MGUS)
MYC (as in Burkitt’s Lymphoma), and are treated similarly as
Burkitt’s Lymphoma • Multiple Myeloma
→ Deep sequencing of DLBCL genomes identified mutations in → Plasma cell neoplasm commonly associated with lytic bone
histone acetyltransferases such as p300 and CREBP. lesions, hypercalcemia, renal failure, and acquired immune
→ Features: Large Cell Size, and diffuse pattern of growth abnormalities
→ Presents as rapidly enlarging mass at a nodal or extranodal site → Incidence higher in men and in those of African descent
→ Waldeyer Ring (lymphatic tissue, including tonsils and → Associated with frequent rearrangements involving IgH locus
adenoids) is commonly involved and various proto-oncogenes
→ Involvement of liver or spleen → destructive masses → Factors produced by neoplastic plasma cells mediate bone
destruction (major pathologic feature)
Burkitt Lymphoma → Usually presents as destructive plasma cell tumors
• Very aggressive tumor of mature B Cells that usually arises at (plasmacytomas) involving axial skeleton
extranodal sites → Bone lesions appear as punched-out defects
• Fastest growing human tumor → Plasmablasts and bizarre, multinucleated cells may
• Strongly associated with translocations involving MYC gene predominate
(regulates aerobic glycolysis, aka Warburg effect→ synthesis of → Flame Cells- cells with red cytoplasms
all building blocks, lipids, nucleotides, and proteins for growth → Mott cells- with multiple grapelike cytoplasmic droplets, and
and cell division) cells with a variety of other inclusions, including fibrils,
• Unlike other tumors of germinal center origin, BL mostly fails to crystalline rods, and globules
express antiapoptotic protein BCL2 → Globular inclusions: Russell bodies (cytoplasmic) and
Dutcher bodies (nuclear)
• Subtypes → Roleaux formation may be observed (RBCs in linear array)
→ African (endemic) Burkitt Lymphoma → May give rise to Plasma Cell leukemia
→ Sporadic (nonendemic) Burkitt Lymphoma → Myeloma kidney- due to Bence-Jones proteins
→ Subset of aggressive lymphomas occurring in HIV patients
• Solitary Myeloma (Plasma Cytoma)
• Pathogenesis, Morphology, and Clinical Presentation → Solitary lesion of bone or soft tissue
→ All forms of Burkitt’s Lymphoma are highly associated with → Occur in the same locations
translocations of MYC gene on chromosome 8→ Inc. MYC → Extraosseous lesions are often located in the lungs,
protein levels oronasopharynx, or nasal sinuses.

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 8 of 24
→ Modest elevations of M proteins in the blood or urine may be ▪ Visual impairment associated with venous congestion,
found in some patients. which is reflected by striking tortuosity and distention of
→ Solitary osseous plasmacytoma almost inevitably progresses retinal veins; retinal hemorrhages
to multiple myeloma, but this can take 10 to 20 years or ▪ Neurologic problems such as headaches, dizziness,
longer. deafness, and stupor
→ Extraosseous plasmacytomas, particularly those involving ▪ Bleeding related to the formation of complexes between
the upper respiratory tract, are frequently cured by local macroglobulins and clotting factors as well as interference
resection. with platelet functions
▪ Cryoglobulinemia resulting from the precipitation of
• Smoldering Myeloma macroglobulins at low temperatures, which produces
→ Defines a middle ground between multiple myeloma and symptoms such as raynaud phenomenon and cold
monoclonal gammopathy of uncertain significance. urticaria
→ Plasma cells make up 10% to 30% of the marrow cellularity, ▪ Incurable progressive disease
and the serum M protein level is greater than 3 gm/dL, but ▪ Tumor growth can be controlled for a time with low doses
patients are asymptomatic. of chemotherapeutic drugs and immunotherapy with anti-
→ About 75% of patients progress to multiple myeloma over a CD20 antibody.
15-year period → Morphology
▪ Marrow contains an infiltrate of lymphocytes, plasma
• Monoclonal Gammopathy of Uncertain Significance cells, and plasmacytoid lymphocytes in varying
proportions, often accompanied by mast cell hyperplasia
→ Most common plasma cell dyscrasia, occurring in about 3%
▪ Some tumors also contain a population of larger lymphoid
of persons older than 50 years of age and in about 5% of
cells with more vesicular nuclear chromatin and prominent
individuals older than 70 years of age.
nucleoli.
→ Patients are asymptomatic and the serum M protein level is
▪ Periodic acid-schiff-positive inclusions containing Ig are
less than 3 gm/dL.
frequently seen in the cytoplasm (Russell bodies) or the
→ Approximately 1% of patients with MGUS develop a
nucleus (Dutcher bodies) of some of the plasma cells
symptomatic plasma cell neoplasm, usually multiple
myeloma, per year.
→ Clonal plasma cells in MGUS contain many of the same
chromosomal translocations and deletions that are found in
fullblown multiple myeloma

• Lymphoplasmacytic Lymphoma
→ B-cell neoplasm of older adults that usually presents in the
sixth or seventh decade of life.
→ With superficial resemblance to CLL/SLL but differs in that a
substantial fraction of the tumor cells undergo terminal
differentiation to plasma cells.
→ Plasma cell component secretes monoclonal IgM, often in
amounts sufficient to cause a hyperviscosity syndrome
known as Waldenström macroglobulinemia. Figure 4. Lymphoplasmacytic Lymphoma
→ Pathogenesis
▪ Associated with acquired mutations in MYD88 - Mantle Cell Lymphoma
encodes an adaptor protein that participates in signaling • Usually presents in the fifth to sixth decades of life and shows a
events that activate NF-κb and also augment signals male predominance.
downstream of the B-cell receptor (Ig) complex, both of • Tumor cells closely resemble the normal mantle zone B
which may promote the growth and survival of the tumor cells that surround germinal centers.
cells.
→ Immunophenotype • Pathogenesis
▪ Lymphoid component expresses B-cell markers such as → Usually presents in the fifth to sixth decades of life and
CD20 and surface Ig shows a male predominance.
▪ Plasma cell component secretes the same Ig that is → Tumor cells closely resemble the normal mantle zone B cells
expressed on the surface of the lymphoid cells. Usually that surround germinal centers.
IgM but can also be IgG or IgA. → Have an (11;14) translocation involving the IgH locus on
→ Clinical Features chromosome 14 and the cyclin D1 locus on chromosome
▪ Weakness, fatigue, and weight loss 11 that leads to overexpression of cyclin D1.
▪ Lymphadenopathy, hepatomegaly, and splenomegaly,
anemia • Immunophenotype
▪ Autoimmune hemolysis caused by cold agglutinins, igm → Express high levels of cyclin D1
antibodies that bind to red cells at temperatures of less → Most tumors are also express CD19, CD20, and
than 37°c. moderately high levels of surface Ig (usually IgM and IgD
▪ Igm-secreting tumors greatly increases the viscosity with κ or λ light chain).
of the blood, giving rise to a hyperviscosity
syndrome.

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 9 of 24
→ It is usually CD5+ and CD23−, which help to distinguish it clone emerges that still depends on antigen-stimulated T-
from CLL/SLL. helper cells for signals that drive growth and survival.
→ The IgH genes lack somatic hypermutation, supporting an → Tumors may acquire additional mutations that render their
origin from a naive B cell. growth and survival antigen-independent, such as the
• Clinical features (11;18), (14;18), or (1;14) chromosomal translocations,
→ Most common presentation is painless which are relatively specific for extranodal marginal zone
lymphadenopathy lymphomas.
→ Symptoms related to involvement of the spleen (present in → All of these translocations up-regulate the expression and
~50% of cases) and gut are also common. function of BCL10 or MALT1, protein components of a
→ Prognosis is poor; the median survival is only 3 to 4 years. signaling complex that activates NF-κb and promotes the
→ Not curable with conventional chemotherapy, and most growth and survival of B cells.
patients eventually succumb to organ dysfunction caused by
tumor infiltration. Hairy Cell Lymphoma
→ Blastoid variant and a “proliferative” expression profiling • Distinctive B-cell neoplasm constitutes about 2% of all
signature are associated with even shorter survivals. leukemias.
→ Hematopoietic stem cell transplantation and proteasome • It is predominantly a disease of middle-aged white males, with
inhibitors are newer therapeutic approaches a median age of 55 and a male-to-female ratio of 5:1
• Pathogenesis
• Morphology → Associated in more than 90% of cases with activating point
→ Nodal tumor cells may surround reactive germinal centers to mutations in the serine/threonine kinase BRAF, which is
produce a nodular appearance at low power, or diffusely positioned immediately downstream of RAS in the MAPK
efface the node. signaling cascade
→ Typically, the proliferation consists of a homogeneous → The specific mutation, a valine to glutamate substitution at
population of small lymphocytes with irregular to residue 600, is also found in diverse other neoplasms,
occasionally deeply clefted (cleaved) nuclear contours including many melanomas and Langerhans cell histiocytosis
→ Large cells resembling centroblasts and proliferation centers • Immunophenotype
are absent, distinguishing mantle cell lymphoma from → Express the pan-B-cell markers CD19 and CD20, surface
follicular lymphoma and CLL/SLL, respectively. Ig (usually IgG), and certain relatively distinctive
→ nuclear chromatin is condensed, nucleoli are inconspicuous, markers, such as CD11c, CD25, CD103, and annexin A1
and the cytoplasm is scant. supporting an origin from a naive B cell.
→ Tumors composed of intermediate-sized cells with more
open chromatin

Figure 6 Hairy Cell Lymphoma (Phase-Contrast Microscope)


Figure 5. Mantle Cell Lymphoma

Marginal Cell Lymphoma


• Heterogeneous group of B-cell tumors that arise within lymph
nodes, spleen, or extranodal tissues.
• The extranodal tumors were initially recognized at mucosal sites
and are often referred to as mucosa-associated lymphoid
tumors (or “MALTomas”).
• Tumor cells show evidence of somatic hypermutation and are
considered to be of memory B-cell origin. Figure 7. Hairy Cell Lymphoma (Stained Smear)
• Pathogenesis:
→ They often arise within tissues involved by chronic • Clinical Features
inflammatory disorders of autoimmune or infectious → Splenomegaly, hepatomegaly, lymphadenopathy
etiology → About one third of those affected present with infections.
→ They remain localized for prolonged periods, spreading → There is an increased incidence of atypical mycobacterial
systemically only late in their course. infections, possibly related to frequent unexplained
→ They may regress if the inciting agent (e.g., Helicobacter monocytopenia.
pylori) is eradicated. → Exceptionally sensitive to “gentle” chemotherapeutic
→ The disease begins as a polyclonal immune reaction. With regimens, which produce long lasting remissions.
the acquisition of still-unknown initiating mutations, a B-cell → Tumors often relapse after 5 or more years, yet generally
respond well when retreated with the same agents

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 10 of 24
→ BRAF inhibitors appear to produce excellent responses in • Typically composed of large anaplastic cells, some containing
tumors that have failed conventional chemotherapy. horseshoe-shape nuclei and voluminous cytoplasm (so-called
→ The overall prognosis is excellent hallmark cells)
→ Leukemic cells with fine hairlike • The tumor cells often cluster about venules and infiltrate
→ projections that are best recognized under the phase- lymphoid sinuses, mimicking the appearance of metastatic
contrast microscope carcinoma.
→ On routine peripheral blood smears, hairy cells have round, • ALK is not expressed in normal lymphocytes; thus, the detection
oblong, or reniform nuclei and moderate amounts of pale of ALK protein in tumor cells is a reliable indicator of an ALK
blue cytoplasm with threadlike or bleblike extensions gene rearrangement.
→ The number of circulating cells is highly variable. • Defined by the presence of rearrangements in the ALK gene on
→ The marrow is involved by a diffuse interstitial infiltrate of chromosome 2p23.
cells with oblong or reniform nuclei, condensed chromatin, • These rearrangements break the ALK locus and lead to the
and pale cytoplasm. formation of chimeric genes encoding ALK fusion proteins,
→ Because these cells are enmeshed in an extracellular matrix constitutively active tyrosine kinases that trigger the RAS and
composed of reticulin fibrils, they usually cannot be JAK/STAT signaling pathways
aspirated (a clinical difficulty referred to as a “dry tap”) and • T-cell lymphomas with ALK rearrangements tend to occur in
are only seen in marrow biopsies. children or young adults, frequently involve soft tissues, and
→ The splenic red pulp is usually heavily infiltrated, leading carry a very good prognosis (unlike other aggressive peripheral
to obliteration of white pulp and a beefy red gross T-cell neoplasms).
appearance. • The cure rate with chemotherapy is 75% to 80%. Inhibitors of
→ Hepatic portal triads are also involved frequently ALK have been developed and are being evaluated as a form of
selective, targeted therapy.
D. PERIPHERAL T-CELL AND NK CELL NEOPLASMS

Peripheral T-cell lymphoma, Unspecified


• Not easily categorized and are lumped into a “wastebasket”
diagnosis, peripheral T-cell lymphoma, unspecified.
• No morphologic feature is pathognomonic, but certain findings
are characteristic
• These tumors efface lymph nodes diffusely and are typically
composed of a pleomorphic mixture of variably sized malignant
T cells (Figure 6)
• There is often a prominent infiltrate of reactive cells, such as
eosinophils and macrophages, probably attracted by tumor-
derived cytokines. Figure 9. Anaplastic Large cell lymphoma
• Brisk neoangiogenesis may also be seen
• They usually express CD2, CD3, CD5, and either αβ or γδ T- Adult T-cell leukemia/Lymphoma
cell receptors.
• This neoplasm of CD4+ T cells is only observed in
• Some also express CD4 or CD8; such tumors are taken to be of
adultsinfected by human T-cell leukemia retrovirus type 1
helper or cytotoxic T-cell origin, respectively.
(HTLV-1)
• DNA analysis is used to confirm the presence of clonal T-cell • Mainly in regions where HTLV-1 is endemic, namely southern
receptor rearrangements. Japan, West Africa, and the Caribbean basin.
• Most patients present with generalized lymphadenopathy, • Common findings include skin lesions, generalized
sometimes accompanied by eosinophilia, pruritus, fever, and lymphadenopathy, hepatosplenomegaly, peripheral blood
weight loss. lymphocytosis, and hypercalcemia.
• These tumors have a significantly worse prognosis than
• Appearance of the tumor cells varies, but cells with multilobated
comparably aggressive mature B-cell neoplasms (e.g., diffuse
nuclei (“cloverleaf” or “flower” cells) are frequently observed
large B-cell lymphoma).
• Tumor cells contain clonal HTLV-1 provirus, which is believed to
play a critical pathogenic role. Notably, HTLV-1 encodes a
protein called Tax that is a potent activator of NF-κB which
enhances lymphocyte growth and survival.
• Most patients present with rapidly progressive disease that is
fatal within months to 1 year despite aggressive chemotherapy.
• Less commonly, the tumor involves only the skin and follows a
much more indolent course, like that of mycosis fungoides
• In addition to adult T-cell leukemia/lymphoma, HTLV-1 infection
sometimes gives rise to a progressive demyelinating disease of
the central nervous system and spinal cord
Figure 8. Peripheral T-cell lymphoma, unspecified
Mycosis Fungosis/Sezary syndrome
Anaplastic Large Cell Lymphoma (ALK Positive) • Different manifestations of a tumor of CD4+ helper T cells that
home to the skin.

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 11 of 24
• Histologically, the epidermis and upper dermis are infiltrated by • Large azurophilic granules are seen in the cytoplasm of the
neoplastic T cells, which often have a cerebriform appearance tumor cells that resemble those found in normal NK cells
due to marked infolding of the nuclear membrane. • Extranodal NK/T-cell lymphoma is highly associated with EBV.
• The tumor cells express the adhesion molecule cutaneous • Tumor originates from a single EBV-infected cell.
leukocyte antigen (CLA) and the chemokine receptors CCR4 • How EBV gains entry is uncertain, since the tumor cells do not
and CCR10, all of which contribute to the homing of normal express CD21, the surface protein that serves as the B-cell EBV
CD4+ T cells to the skin. receptor.
• Although cutaneous disease dominates the clinical picture, • Most tumors are CD3− and lack T-cell receptor rearrangements
sensitive molecular analyses have shown that the tumor cells and express NK-cell markers, supporting an NK-cell origin. No
circulate through the blood, marrow, and lymph nodes even consistent chromosome aberration has been described.
early in the course. • Most extranodal NK/T-cell lymphomas are highly aggressive
• Median survival of 8 to 9 years. neoplasms that respond well to radiation therapy but are
• Transformation to aggressive T-cell lymphoma occurs resistant to chemotherapy.
occasionally as a terminal event. • Prognosis is poor in patients with advanced disease.
• Clinically, the cutaneous lesions of mycosis fungoides typically
progress through three somewhat distinct stages, an E. HODGKIN’S LYMPHOMA
inflammatory premycotic phase, a plaque phase, and a tumor • Hodgkin lymphoma (HL) encompasses a group of lymphoid
phase neoplasms that differ from NHL in several respects
• Late disease progression is characterized by extracutaneous • HL arises in a single node or chain of nodes and spreads first to
spread, most commonly to lymph nodes and bone marrow. anatomically contiguous lymphoid tissues.
• Sézary syndrome is a variant in which skin involvement is • characterized by the presence of neoplastic giant cells called
manifested as a generalized exfoliative erythroderma. Reed- Sternberg cells. These cells release factors that induce
• In contrast to mycosis fungoides, the skin lesions rarely proceed the accumulation of reactive lymphocytes, macrophages, and
to tumefaction, and there is an associated leukemia of “Sézary” granulocytes, which typically make up greater than 90% of the
cells with characteristic cerebriform nuclei. tumor cellularity.
• In the vast majority of HLs, the neoplastic Reed-Sternberg cells
Large, Granular, lymphocytic leukemia are derived from germinal center or postgerminal center B cells.
• T-cell and NK-cell variants of this rare neoplasm are recognized, • The average age at diagnosis is 32 years. It is one of
both of which occur mainly in adults. • the most common cancers of young adults and adolescents,but
• The tumor cells are large lymphocytes with abundant blue also occurs in the aged.
cytoplasm and a few coarse azurophilic granules, best seen in • It was the first human cancer to be successfully treated with
peripheral blood smears. The marrow usually contains sparse radiation therapy and chemotherapy, and is curable in most
interstitial lymphocytic infiltrates, which can be difficult to cases.
appreciate without immunohistochemical stains.
• Infiltrates are also usually present in the spleen and liver. As
might be expected, T-cell variants are CD3+, whereas NK-cell
large granular lymphocytic leukemias are CD3−, CD56+.
• Recent work has shown that 30% to 40% of large granular
lymphocytic leukemias have acquired mutations in the
transcription factor STAT3, which functions downstream of
cytokine receptors.
• Individuals with T-cell disease usually present with mild to
moderate lymphocytosis and splenomegaly.
• Lymphadenopathy and hepatomegaly are usually absent
• Despite the relative paucity of marrow infiltration, neutropenia Pathogenesis
and anemia dominate the clinical picture. • Ig genes of Reed-Sternberg cells have undergone both V(D)J
• Neutropenia is often accompanied by a striking decrease in late recombination and somatic hypermutation, establishing an
myeloid forms in the marrow. Rarely, pure red cell aplasia is origin from a germinal center or postgerminal center B cell
seen. • Despite having the genetic signature of a B cell, the Reed-
• There is also an increased incidence of rheumatologic Sternberg cells of classical HL fail to express most B-cell–
disorders. specific genes, including the Ig genes
• Some patients with Felty syndrome, a triad of rheumatoid arthritis, • cause of this wholesale reprogramming of gene expression is
splenomegaly, and neutropenia,have this disorder as an underlying presumably the result of widespread epigenetic changes of
cause uncertain etiology
• Activation of the transcription factor NF-κB is a common
Extranodal NK/T-cell Lymphoma event in classical HL. This can occur by several mechanisms:
• Rare in the United States and Europe, but 3% of NHLs in Asia. → NF-κB may be activated either by EBV infection or by
• Presents most commonly as a destructive nasopharyngeal some other mechanism and turns on genes that promote
mass lymphocyte survival and proliferation.
→ EBV tumor cells express latent membrane protein-1
• Less common sites: testis and the skin
(LMP-1), a protein encoded by the EBV genome that
• The tumor cell infiltrate surrounds and invades small vessels, transmits signals that up-regulate NF κB.
leading to extensive ischemic necrosis.

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 12 of 24
→ Activation of NF-κB may occur in EBV-tumors as a result
of acquired loss-of-function mutations in IκB or A20 (also
known as TNF alpha-induced protein 3, or TNFAIP3),
which are both negative regulators of NF-κB.
→ It is hypothesized that activation of NF-κB by EBV or
other mechanisms rescues “crippled” germinal center B
cells that cannot express Igs from apoptosis, setting the
stage for the acquisition of other unknown mutations that
collaborate to produce Reed-Sternberg cells. • Lymphohistiocytic variants (L&H cells / popcorn cells)
→ Seen in the lymphocyte predominance subtype
• EBV-infected B cells resembling Reed-Sternberg cells are found → Polypoid nuclei, inconspicuous nucleoli, and moderately
in the lymph nodes of individuals with infectious mononucleosis, abundant cytoplasm
strongly suggesting that EBV-encoded proteins play a part in
the remarkable metamorphosis of B cells into Reed-Sternberg
cells.
• The florid accumulation of reactive cells in tissues involved by
classical HL occurs in response to a wide variety of cytokines
(e.g., IL-5, IL-10, and M-CSF) chemokines (e.g.,eotaxin), and
other factors (e.g., immunomodulatory factor galectin-1) that are
secreted by Reed-Sternberg cells. Once attracted, the reactive
cells produce factors that support the growth and survival of the
tumor cells and further modify the reactive cell response. B. Subtypes of Hodgkin Lymphoma
• eosinophils and T cells express ligands that activate the CD30 1. Nodular sclerosis type
and CD40 receptors found on Reed-Sternberg cells, producing → Most common form (65-70% of cases)
signals that up-regulate NF- B. → Tends to involve the lower cervical, supraclavicular, and
• Someof the factors produced by RS cells give rise to a state of mediastinal lymph nodes
immunodeficiency by impairment of T helper and cytotoxic cells → Collagen is deposited in bands that divide involved lymph
nodes into circumscribed nodules
and enhancing the generation of regulatory T cells
• Reed-Sternberg cells are aneuploid and possess diverse clonal
chromosomal aberrations. Disease stage I or II
• Copy number gains in the REL proto-oncogene on chromosome Cellular infiltrate Heterogenous
2p are particularly common and may also contribute to RS cells Lacunar cells
increases in NF-κB activity. Immunophenotype Classic
Associated with No
EBV?
A. Morphology of Reed-Stenberg Cells
More common in… Young adults; equal
• Classic diagnostic RS cells occurrence in males &
→ Large cells (45 µm in diameter) with multiple nuclei females
→ A single nucleus with multiple nuclear lobes, each with a “Classic” immunophenotype = (+) PAX5, CD15, CD30
large inclusion-like nucleolus about the size of a small
lymphocyte (5 to 7 µm in diameter)

• Mononuclear variant
→ Single nucleus with a large inclusion-like nucleolus

2. Mixed-cellularity type
→ 20-25% of cases
→ Associated with systemic symptoms (i.e. night sweats &
weight loss) and advanced tumor stag
• Lacunar cells Disease stage III or IV
→ Seen in the nodular sclerosis subtype Cellular infiltrate Heterogenous
→ More delicate, folded, or multilobate nuclei RS cells Diagnostic & mononuclear
→ Abundant pale cytoplasm that is often disrupted during Immunophenotype Classic
the cutting of sections, leaving the nucleus sitting in an Associated with EBV? Yes; 70% of cases
empty hole (a lacuna) More common in… Males, biphasic incidence
(peak in young adults &
>55 yrs old)

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 13 of 24
• Most cases present with cutaneous immune
unresponsiveness (anergy)
→ Results from depressed cell-mediated immunity
(because factors released by RS cells suppress TH1
immune responses)

• Patients with the nodular sclerosis & lymphocyte predominance


types:
→ Tend to have stage I-II disease; free of systemic
symptoms
3. Lymphocyte-rich type • Patients with disseminated disease (stages III-IV) or the mixed
→ Uncommon cellularity or lymphocyte depletion types:
Cellular infiltrate Mostly reactive → More likely to have constitutional symptoms, such as
lymphocytes fever, night sweats, and weight loss
RS cells Diagnostic & mononuclear
Immunophenotype Classic • Spread of HL:
Associated with EBV? Yes; 40% of cases → Nodal disease → splenic disease → hepatic disease →
More common in… Males, older adults involvement of marrow & other tissues
--------------------------------------------------------------
4. Lymphocyte depletion type Clinical Staging of Hodgkin and Non-Hodgkin Lymphomas
→ Least common type (<5% of cases) (Ann Arbor Classification)
→ Often presents with advanced disease
Stage 1: Involvement of a single lymph node region (I) or a single
extralymphatic organ or site (IE)

Stage 2: Involvement of 2 or more lymph node regions on the


same side of diaphragm alone (II) or localized involvement of an
extralymphatic organ or site (IIE)

Stage 3: Involvement of lymph node regions on both sides of the


diaphragm without (III) or with (IIIE) localized involvement of an
extralymphatic organ or site
Cellular infiltrate Scarce Stage 4: Diffuse involvement of 1 or more extralymphatic organs
RS cells Diagnostic & other variants or sites with or without lymphatic involvement
Immunophenotype Classic
Associated with EBV? Yes; 90% of cases • All stages are further divided on the basis of the absence (A) or
More common in… Older males, HIV(+) presence (B) of the following symptoms:
patients, developing
countries → Unexplained fever
→ Drenching night sweats
5. Lymphocyte predominance type → Unexplained weight loss of greater than 10% of normal
→ Uncommon (5% of cases) body weight

Disease stage I or II • Tumor stage is the most important prognostic variable.


Cellular infiltrate Small lymphocytes • Stage I-II: cure rate = ~90%
admixed with • Advanced disease (Stage IVA & IVB) cure rate = 60-70%
macrophages, follicular • Low-stage localized HL can be cured with involved field
dendritic cells, reactive B radiotherapy.
cells
→ However, long-term survivors have a higher incidence of
RS cells L&H / popcorn cells
certain malignancies, including lung cancer, melanoma,
Immunophenotype (+) for CD20 & BCL6 and breast cancer.
(iba!!) (-) for CD15 & CD30
→ Non-neoplastic complications of radiotherapy
Associated with EBV? No include pulmonary fibrosis and accelerated
More common in… Young males with cervical atherosclerosis.
or axillary → Chemotherapy with alkylating agents also increases the
lymphadenopathy; incidence of secondary tumors.
mediastinal
MYELOID NEOPLASMS
• Common feature: origin = hematopoietic progenitor cells
• Primarily involve the marrow and, to a lesser degree, the
secondary hematopoietic organs (spleen, liver, lymph nodes)
• 3 categories
→ Acute myeloid leukemias
o Accumulation of immature myeloid forms (blasts) in
the bone marrow suppresses normal hematopoiesis
→ Myelodysplastic syndromes (MDS)
Clinical Features o Defective maturation of myeloid progenitors gives
• HL most commonly presents as painless lymphadenopathy. rise to ineffective hematopoiesis, leading to
cytopenia

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 14 of 24
→ Myeloproliferative disorders • The number of leukemic cells in the blood is highly variable.
o Increased production of one or more types of blood Occasionally, blasts are entirely absent from the blood
cells (aleukemic leukemia)
• Manifestations of myeloid neoplasms are influenced by:
→ The position of the transformed cell within the hierarchy Types of myeloid blasts:
of progenitors (i.e., a pluripotent hematopoietic stem cell A. Myeloblasts
versus a more committed progenitor) → Delicate nuclear chromatin, 2-4 nucleoli, more
→ The effect of the transforming events on differentiation, voluminous cytoplasm than lymphoblasts
which may be inhibited, skewed, or deranged by o Fine, Peroxidase-positive azurophilic granules
particular oncogenic mutations

A. ACUTE MYELOID LEUKEMIA


• Tumor of hematopoietic caused by acquired oncogenic
mutations that impede differentiation, leading to the
accumulation of immature myeloid blasts in the marrow.
• Replacement of the marrow with blasts produces marrow failure
and complications related to anemia, thrombocytopenia, and
neutropenia. AML occurs at all ages, but the incidence rises
throughout life, peaking after 60 years of age B. Auer rods
→ Needle-like azurophilic granules
CLASSIFICATION → Numerous in AML with the t(15;17) (acute
• SEE APPENDIX FOR FULL WHO CLASSIFICATION promyelocytic leukemia)

PATHOGENESIS
• Recurrent genetic aberrations seen in AML disrupt genes
encoding transcription factors that are required for normal
myeloid differentiation. For example:
→ RUNX1 and CBFB genes encode polypeptides that bind
one another to form a RUNX1/CBFB1β transcription
factor that is required for normal hematopoiesis.
→ The two most common chromosomal rearrangements,
t(8;21) and inv(16), disrupt the RUNX1 and CBFB
genes, respectively. C. Monoblasts
→ Folded or lobulated nuclei; lack Auer rods
• Mutations that lead to activation of growth signaling pathways + → Nonspecific esterase-positive
transcription factor aberrations = AML
• Mutations affecting factors that impact the state of the
“epigenome”, suggesting the epigenetic alterations have a
central role in AML
→ DNA methylation
→ Posttranslational modifications of histones (e.g.,
acetylation, methylation, phosphorylation)

• Immunophenotype of AML
→ Because it can be diffiult to distinguish myeloblasts and • The number of leukemic cells in the blood is highly variable.
lymphoblasts morphologically, the diagnosis of AML is • Blasts may be more than 100,000/mm3, but are under
confimed by performing stains for myeloid-specific 10,000/mm3 in about 50% of patients
antigens (flow sytometry). • Occasionally, blasts are entirely absent from the blood
• Cytogenetic analysis (aleukemic leukemia).
→ AMLS arising de novo in younger adults • For this reason, a bone marrow examination is essential to
o Associated with balanced chromosomal
exclude acute leukemia in pancytopenic patients.
translocations, particularly t(8;21), inv(16), and
t(15;17)
→ AMLs following MDS or exposure to DNA-damaging
agents (chemo/radiation therapy) CLINICAL FEATURES
o Deletions or monosomies involving chromosomes 5 • Most patients present within weeks or a few months of the onset
and 7 of symptoms with complaints related to anemia, neutropenia,
o Lack chromosomal translocations and thrombocytopenia, most notably fatigue, fever, and
→ AMLs occurring after treatment with topoisomerase II spontaneous mucosal and cutaneous bleeding
inhibitors
• Very similar to those produced by ALL. Thrombocytopenia
→ Translocations involving the MLL gene on chromosome
11q23 • Results in a bleeding diathesis, which is often prominent.
• Cutaneous petechiae and ecchymoses, serosal hemorrhages
MORPHOLOGY into the linings of the body cavities and viscera, and mucosal
hemorrhages into the gingivae and urinary tract are common
• The diagnosis of AML is based on the presence of at least 20%
myeloid blasts in the bone marrow.

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 15 of 24
• Procoagulants and fibrinolytic factors released by leukemic 2. RNA Splicing Factors
cells, especially in AML with the t(15;17), exacerbate the - involves components of 3’ end of the
bleeding tendency. RNA splicing machinery
• Infections are frequent, particularly in the oral cavity, skin, lungs, 3. Transcription Factors
kidneys, urinary bladder, and colon, and are often caused by - TFs that are required for normal
opportunists such as fungi, Pseudomonas, and commensals. myelopoiesis resulting to deranged
• Signs and symptoms related to involvement of tissues other differentiation (feature of MDS)
than the marrow are usually less striking in AML than in ALL, 4. TP53 (tumor suppressor gene)
but tumors with monocytic differentiation often infiltrate the skin - loss-of-function mutation
(leukemia cutis) and the gingiva; - seen in 10% of MDS cases
• Central nervous system spread is less common than in ALL. - results to presence of a complex
• AML occasionally presents as a localized soft-tissue mass karyotype
known variously as a myeloblastoma, granulocytic sarcoma, or and poor clinical outcomes
chloroma.
• Without systemic treatment, such tumors inevitably progress to Primary and t-MDS
full-blown AML over time. - associated with recurrent chromosomal
• abnormalities including:
Prognosis. a. monosomies 5 and 7
• AML is generally a difficult disease to treat; about 60% of b. deletions of 5q, 7q, and 20q
patients achieve complete remission with chemotherapy, but c. trisomy 8
only 15% to 30% remain free of disease for 5 years.
• Outcome varies markedly among different molecular subtypes. • Marrow may be: hypercellular
• With targeted therapy using all-trans retinoic acid and arsenic (common), normocellular or hypocellular
salts AMLs with the t(15;17) now have the best prognosis of (rare)
any type, being curable in more than 80% of patients.
• AMLs with t(8;21) or inv(16) have a relatively good prognosis Most characteristic finding:
with conventional chemotherapy, particularly in the absence of disordered/dysplastic differentiation
KIT mutations. affecting erythroid, granulocytic, monocytic
• In contrast, the prognosis is dismal for AMLs that follow MDS or and megakaryocytic lineages
genotoxic therapy, or that occur in older adults, possibly
because in these contexts the disease arises out of a Abnormalities in RBCs:
background of hematopoietic stem cell damage or depletion. a. Ring sideroblasts
- Erythroblasts with iron-laden
• “Highrisk” forms of AML (as well as relapsed AML of all types)
mitochondria visible as perinuclear
are treated with hematopoietic stem cell transplantation when
granules in Prussian blue-stained aspirates
possible.
or biopsies
• Sequencing of AML genomes has recently revealed new
b. Megaloblastoid maturation
molecular predictors of outcome.
- Resembles abnormality in vitamin B12
• Insights gained from DNA sequencing will have an increasingly
and folate deficiency
important role in selecting therapy and stratifying patient in
MORPHOLOGY c. Nuclear budding abnormalities
clinical trials of new therapeutics, such as drugs that target the
- Nuclei with misshapen, often polyploid,
tumor epigenome.
outlines
Abnormalities in neutrophils:
B. MYELODYSPLASTIC SYNDROMES
- Decreased numbers of secondary
• Group of clonal stem cell disorders characterized by maturation
granules
defects that are associated with ineffective hematopoiesis and a
- Toxic granulations
high risk of transformation to AML
- Dohle bodies
• Bone marrow is partly or wholly replaced by clonal progeny of
- Pseudo-Pelger-Hut cells (neutrophils with
neoplastic multipotent stem with an ineffective and disordered
only two nuclear lobes)
differentiation
- Lack of nuclear segmentation 

→ Abnormal cells stay within bone marrow progressing to
peripheral blood cytopenia
Abnormalities in megakaryocytes:
• Primary vs Secondary MDS
- Single nuclear lobes or multiple separate
→ Primary – Idiopathic
nuclei (pawn ball megakaryocytes)
→ Secondary – previous genotoxic drug or radiation therapy
• All can transform to AML by t-MDS has the highest risk of Abnormalities in myeloid blasts:
transformation - Increased (>20%) overall marrow
→ Morphological changes and chromosomal aberrations are cellularity
often observed Primary MDS
Mutations in: - Disease of older adults (>70 yrs)
1. Epigenetic Factors CLINICAL
PATHOGENESIS - Weakness and infections due to
- includes factors the regulate DNA FEATURES
cytopenia
methylation and histone modifications - Poor prognosis for higher blast counts

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 16 of 24
and sever cytopenia
- Median survival: 9-29 months - Characteristic finding: presence of
- Death due to bleeding scattered macrophages with abundant
(thrombocytopenia) or infections wrinkled, green-blue cytoplasm (sea-blue
(neutropenia) histiocytes)

t-MDS Blood: leukocytosis (>100,000 cells/mm3)


- Poorest prognosis consisting of neutrophils, band forms,
metamyelocytes, myelocytes, eosinophils and
- Median survival: 4-8 months
basophils
- Characterized by severe cytopenia and
rapid progression to AML
- Platelets: increased
- Spleen: greatly enlarged due to extensive
Tx: extramedullary hematopoiesis and often with
Younger patients – allogeneic infarcts
hematopoietic stem cell transplantation
Older patients – supportive therapy only - Disease of adults but also occurs in children
and adolescents
C. MYELOPROLIFERATIVE DISORDERS - Peak incidence: Fifth to sixth decades of
• Common pathogenic feature: mutated, constitutively activated life
tyrosine kinases or acquired aberrations in signaling pathways - Onset is insidious
that lead to growth factor independence - Mild to moderate anemia and
hypermetabolism due to increased cell
→ Mutated tyrosine kinases → growth factor-independent
turnover lead to fatigability, weakness, weight
proliferation and survival of marrow progenitors → loss and anorexia
↑production of one or more mature blood elements - First symptoms: dragging sensation in
→ Does not impair differentiation abdomen due to splenomegaly or acute onset
• Common features: of LUQ pain due to splenic infarction
→ Increased proliferative drive in bone marrow
→ Extramedullary hematopoiesis Natural history:
→ Marrow fibrosis and peripheral blood cytopenia Slow Progression (for 3 years)
→ Transformation to acute leukemia Accelerated Phase, increasing anemia and
thrombocytopenia (for 6-12 months)
CLINICAL
Blast Crisis (resembling acute leukemia)
Chronic Myelogenous Leukemia FEATURES - In 50% of cases, blast crisis occur without
• Presence of chimeric BCR-ABL gene derived from portions of an accelerated phase
BCR gene on chromosome 22 and the ABL gene on
chromosome 9 Tx:
→ BCR-ABL – directs synthesis of constitutively active BCR- BCR-ABL inhibitors (not clear if truly
ABL tyrosine kinase curative)
→ In 90% of cases, BCR-ABL is created by a reciprocal - Markedly decrease BCR-ABL positive cells
(9;22)(q34;q11) translocation = Philadelphia chromosome in marrow but do not extinguish CML “stem
cell” which persists at low levels
(Ph) 

Hematopoietic stem cell transplantation
• Cell origin: Pluripotent hematopoietic stem cell
- Should be performed in stable phase (slow
Dimerization domain of BCR-ABL self- progression phase) 

associates
- In younger patients

ABL tyrosine kinase activation
Treatments have less favorable outlook after

slow progression phase due to the extensive
ABL tyrosine phosphorylates proteins that
damage and rapid progression of the disease
induce signaling through the same pro-growth
and pro-survival pathways, RAS & JAK/STAT
PATHOGENESIS
pathways, (turned on by hematopoietic
growth factors)

- Preferentially drives the proliferation of Polycythemia Vera


granulocytic and megakaryocytic progenitors • Strongly associated with activating point mutations in tyrosine
- Causes abnormal release of immature kinase JAK2
granulocytic forms from marrow into blood • Characterized by increased marrow production of red cells,
granulocytes and platelets (panmyelosis)
- Marrow is hypercellular due to massive
• Increase in red cells (polycythemia) is responsible for most
increased numbers of maturing granulocytic
clinical symptoms 

precursors
MORPHOLOGY - Megakaryocytes: increased; small, Point mutation in JAK2 (valine-to-phenylalanine
dysplastic forms substitution)
PATHOGENESIS
- Erythroid progenitors: normal or mildly ↓
decreased Constitutive JAK2 signaling

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 17 of 24

Increased marrow production of RBC Essential Thrombocytosis
independent of growth factors • Associated with activating point mutations in JAK2 (50% of

cases), MPL (5-10% of cases) or calreticulin (remaining cases)
Decreased requirements for erythropoietin and
→ MPL – a receptor tyrosine kinase that is activated by
other hematopoietic growth factors
thrombopoietin

Low serum erythropoietin levels → Calreticulin – protein with functions in endoplasmic
reticulum and cytoplasm
Secondary forms of polycythemia is → JAK2 and calreticulin mutations are mutually exclusive hence
characterized by high erythropoietin levels calreticulin mutations are hypothesized to cause increase
JAK-STAT signaling
- Elevated hematocrit leads to increased blood Point mutations in JAK2, MPL or calreticulin
viscosity and sludging ↓
- Thrombocytosis with abnormal platelet function Constitutive JAK2 or MPL signaling
leads to increased risk for thrombosis and ↓
PATHOGENESIS
bleeding Renders progenitors thrombopoietin-
- Marrow is hypercellular with some residual fat independent
- Increase in red cell progenitors is subtle and ↓
usually accompanied by an increase in Hyperproliferation
granulocytic precursors and megakaryocytes - Marrow cellularity is mildly increased but
- In 10% of cases, moderate to marked megakaryocytes are markedly increased
increased in reticulin fibers is seen and with abnormally large forms
- Delicate reticulin fibers are seen but overt
- Mild organomegaly MORPHOLOGY fibrosis (in primary myelofibrosis) is absent
- Due to congestion - Mild leukocytosis
MORPHOLOGY - Blood contains increased basophils and - Mild organomegaly (50% of cases) due to
abnormally large platelets extramedullary hematopoiesis
- Late PCV: “Spent Phase”
- extensive marrow fibrosis that - Incidence: 1-3 per 100,000 per year
displaces hematopoietic cells - Usually occurs in 60 years old or older
- increased extramedullary - Long asymptomatic periods with occasional
hematopoiesis in spleen and liver prominent thrombotic or hemorrhagic crises
organomegaly - Median survival: 12-15 years

- Transformation to AML is rare (1%) 
 - Major clinical manifestations: Thrombosis


- Uncommon (1-3 per 100,000 per year) CLINICAL and hemorrhage due to platelet dysfunctions
- Insidious onset, usually in adults of late middle FEATURES
age - Separated from PCV and primary
- Increased red cell mass and hematocrit + myelofibrosis based on: absence of
increased total blood volume → abnormal blood polycythemia and marrow fibrosis,
flow (esp. on veins → becomes distended) respectively 

- Cyanosis due to stagnation and deoxygenation
of blood in peripheral vessels Tx: Gentle chemotherapeutic agents that
- Headache, dizziness, hypertension and GI suppress thrombopoiesis
symptoms
- Intense pruritus and peptic ulceration due to
histamine release of basophils Primary Myelofibrosis
- Hyperuricemia due to high cell turnover
• Hallmark:
- Increased risk of bleeding and thrombosis due
to abnormal blood flow and platelet function. → Development of obliterative marrow fibrosis
CLINICAL ▪ Reduces bone marrow hematopoiesis → cytopenias and
Without treatment, death occurs within months of
FEATURES extensive extramedullary
diagnosis
• hematopoiesis
Activating mutations of JAK2 (50-60% of
cases), MPL (1-5% of cases) or calreticulin (remaining cases)
Lab findings: = similar to ET
- Hemoglobin concentration: 14-28 g/dL Chief pathologic feature:
- Hematocrit: >/=60% - Extensive deposition of collagen in
- WBC count: 12,000-50,000 cells/mm3 marrow by non- neoplastic fibroblasts
- Platelet count: > 500,000 platelets/mm3 caused by inappropriate release of fibrogenic
- Platelets exhibit morphologic abnormalities factors (PDGF & TGF-β) from neoplastic
(giant forms) and defective aggregation PATHOGENESIS megakaryocytes
- TGF-β – promotes collagen deposition and
Tx: causes angiogenesis
- Phlebotomy maintains red cell mass at near
normal levels - Fibrosis displaces hematopoietic elements
- JAK2 inhibitors (stem cells) causing marrow failure

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 18 of 24
- Circulating hematopoietic stem cells take up LANGERHANS CELL HISTIOCYTOSIS
residence in spleen, liver and lymph nodes • Histiocytosis” – variety of proliferative disorders of dendritic cells
extramedullary hematopoiesis
or macrophages 

- Moderate to severe anemia due to
disordered red cell production in
• A spectrum of proliferations of a special type of immature
extramedullary sites and suppression of dendritic cell called Langerhans cell
marrow function • Majority of mutations are oncogenic
Early myelofibrosis: → Aberrant expression of chemokine receptors
- Marrow is hypercellular due to increasing ▪ a factor that contributes to homing of neoplastic
maturing hematopoietic cells (similar to PCV) Langerhans cells
- Erythroid and granulocytic precursors: • Most common mutation: activating valine-to-glutamate
normal substitution at residue 600 in BRAF
- Megakaryocytes: large, dysplastic and • Less common mutation: TP53, RAS and tyrosine kinase MET
abnormally clustered • Proliferating Langerhans cells: abundant, vacuolated cytoplasm
- Minimal fibrosis
and vesicular nuclei with linear grooves or folds 

- Blood: leukocytosis and thrombocytosis
→ Presence of Birbeck granules in cytoplasm is characteristic
Late myelofibrosis: → Birbeck granules: pentalaminar tubules with dilated terminal
- Marrow is more hypercellular and end producing a tennis racket-like appearance which contain
diffusely fibrotic the protein langerin
- Clusters of atypical megakaryocytes with
unusual nuclear shapes (“cloud-like”) A. MULTIFOCAL MULTISYSTEM LANGERHANS CELL
MORPHOLOGY - Hematopoietic elements found within dilated HISTIOCYTOSIS (LETTERER-SIWE)
sinusoids
• Occurs most frequently before 2 years of age but occasionally
Very late myelofibrosis: affects adults 

- Fibrotic marrow space is converted into • Dominant clinical feature: development of cutaneous
bone (osteosclerosis) lesions resembling a seborrheic eruption
- Features are identical to spent phase of → Caused by infiltrates of Langerhans cells over the front and
other myeloproliferative disorders back of trunk and on scalp
- Marrow distortion due to fibrosis leads to • Most have hepatosplenomegaly, lymphadenopathy, pulmonary
premature release of nucleated erythroid and lesions and destructive osteolytic bone lesions 

early granulocyte progenitors
• Extensive infiltration of marrow → anemia, thrombocytopenia
(leukoerythroblastosis)
and predisposition to recurrent infections 

Teardrop-shaped red cells (dacryocytes) • In some instances, the tumor cells are anaplastic = Langerhans
- Cells that are damaged during irthing cell sarcoma 

process in fibrotic marrow
- Less common than PCV and ET B. UNIFOCAL AND MULTIFOCAL UNISYSTEM
- Usually occurs in individuals older than 60 LANGERHANS CELL HISTIOCYTOSIS
years
• Characterized by proliferations of Langerhans cells admixed
- Median survival: 3-5 years
with variable numbers of eosinophils, lymphocytes, plasma
- Progressive anemia and splenomegaly
(sensation of fullness in LUQ)
cells and neutrophils.
- Fatigue, weight loss and night sweats due to → Eosinophils are usually a prominent component of infiltrate
CLINICAL
increase in metabolism associated with • Unifocal lesions commonly affects the skeletal system in older
FEATURES
expanding mass of hematopoietic cells children or adults
- Hyperuricemia and secondary gout due to → Asymptomatic or may cause pain, tenderness and pathologic
high rate of cell turnover fractures
→ May heal spontaneously or be cured by local excision or
Lab findings: irradiation
- Moderate to severe normocytic
• Multifocal unisystem disease usually affects young children
normochromic anemia accompanied by
→ Presents with multiple erosive bony masses that may expand
leukoerythroblastosis
to adjacent tissues
- WBC count: normal or mildly reduced but
can be markedly elevated (80,000- 100,000 → Involvement of the posterior pituitary stalk that may lead to
cells/mm3) early in the course diabetes insipidus
- Platelet count: normal or elevated
but → Hand-Schuller-Christian triad
continuation of thrombocytopenia as disease progresses ▪ Calvarial bone defects
CLINICAL ▪ Diabetes insipidus
FEATURES Tx: ▪ Exophthalmos
JAK2 inhibitors – effective in decreasing
symptoms C. PULMONARY LANGERHANS CELL HISTIOCYTOSIS
Hematopoietic stem cell transplantation • Most often seen in adult smokers
only effective for young patients
→ May regress spontaneously upon smoking cessation
• Reactive proliferations of Langerhans cells
• 40% are associated with BRAF mutations (neoplastic in origin)

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 19 of 24
III. SPLEEN
• 5 to 6 inches in length, 4 inches width, 150 g weight
• Storage and removal of worn-out erythrocytes
• Recycling of iron contained in the hemoglobin
• Blood forming organ in fetus

A. COMPONENTS OF THE SPLEEN


RED PULP
• primarily a filter designed to screen and eliminate defective
or foreign cells
• Traversed by numerous thin walled vascular sinusoids
• Separated by splenic cords or cords of Billroth
• Endothelial lining-discontinuous
• Macrophages creates a physical and functional filter
• Major repository mononuclear phagocytic cell

WHITE PULP
• Site where lymphoid cells can be found
• Is composed of masses of T and B lymphocytes
• Ensheathing a central artery
• Represented by the splenic nodule
• Each nodule has a peripheral or marginal zone of densely
packed lymphocytes

B. FUNCTIONS OF THE SPLEEN


1. Phagocytosis of blood cells and particulate matter
• In conditions in which red cell deformability is decreased,
red cells become trapped in the cords and phagocytosed
by macrophages.
• Splenic macrophages are also responsible for “pitting” of
red cells, the process by which inclusions bodies are
excised, and for the removal of particles, such as
bacteria, from the blood.
2. Antibody production.
• Dendritic cells in the periarterial lymphatic sheath trap
antigens and present them to T lymphocytes.
• T- and B-cell interaction at the edges of white pulp
follicles leads to the generation of antibody-secreting
plasma cells within the sinuses of the red pulp
3. Hematopoiesis.
• During fetal development, the spleen may be a minor site
of hematopoiesis, but this normally disappears by birth. Nonspecific Acute Splenitis
However, the spleen can become a major site of
compensatory extramedullary hematopoiesis in the ETIOLOGY • occurs in any blood-borne infection
setting of severe chronic anemia and in patients with • caused both by the microbiologic agents
myeloproliferative disorders, such as chronic PATHOGENESIS themselves and by cytokines that are
myelogenous leukemia and primary myelofibrosis. released as part of the immune response
4. Sequestration of formed blood elements • Spleen is enlarged (200-400 gm) and
• Normal spleen contains only about 30 to 40 mL of red soft
cells, but this volume increases greatly with • major feature: acute congestion of the
splenomegaly. red pulp
• Normal spleen also harbors approximately 30% to 40% • Neutrophils, plasma cells, and
of the total platelet mass. MORPHOLOGY eosinophils are usually present
→ With splenomegaly up to 80% to 90% of the total platelet throughout the white and red pulp
mass can be sequestered → thrombocytopenia. • At times the white pulp follicles may
• Enlarged spleen can trap white cells → leukopenia undergo necrosis, particularly when the
causative agent is a hemolytic
streptococcus
C. DISEASES OF THE SPLEEN
SPLENOMEGALY/HYPERSPLENISM Congestive Splenomegaly
• When sufficiently enlarged, the spleen causes a dragging ETIOLOGY Chronic venous outflow obstruction
sensation in the left upper quadrant and, through pressure on
Causes of obstruction:
the stomach, discomfort after eating.
• Intrahepatic d/o reducing portal venous
• Hypersplenism - increased sequestration of formed elements
drainage
and the consequent enhanced phagocytosis by the splenic PATHOGENESIS • Extrahepatic d/o that directly impinge
macrophages
upon the portal or splenic veins
→ characterized by anemia, leukopenia, thrombocytopenia,
• Sytemic or central venous congestion- in
alone or in combination right-sided cardiac decompensation

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 20 of 24
• Liver cirrhosis - main cause of massive • Usually precipitated by blunt trauma
congestive splenomegaly • Spontaneous ruptures - occur in the apparent absence of a
• Spontaneous portal vein thrombosis physical blow.
• Pylephlebitis - inflammation of the portal → stem from some minor physical insult to a spleen made
vein fragile by an underlying condition.
• Long-standing congestion produces → The most common predisposing conditions are infectious
marked enlargement (1000 to 5000 gm) mononucleosis, malaria, typhoid fever, and lymphoid
• The organ is firm, and the capsule is
neoplasms, which may cause the spleen to enlarge rapidly,
usually thickened and fibrous
producing a thin, tense capsule that is susceptible to rupture.
• Microscopically, the red pulp is
congested early in the course but • Chronically enlarged spleens are unlikely to rupture
MORPHOLOGY becomes increasingly fibrotic and cellular because of the toughening effect of extensive reactive
with time fibrosis
• The elevated portal venous pressure
stimulates the deposition of collagen in IV. THYMUS
the basement membrane of the • Lobulated organ covered by a capsule; irregularly pyramidal
sinusoids, which appear dilated because • From the 3rd and 4th pair of pharyngeal pouches
of the rigidity of their walls • Weight:10 to 35g at birth
→ Maximum weight of 20 to 50g at puberty
Splenic Infarcts
→ Then undergoes progressive involution with a weight of
Occlusion of the major splenic artery or any of
5 – 15g, although it never disappears completely
its branches
ETIOLOGY
• most often caused by emboli that arise → May also involute in children and young adults in
from the heart response to severe illness and HIV infection
• Bland infarcts - pale, wedge-shaped, and • Two fused, well-encapsulated lobes
subcapsular in location. The overlying • Divisions:
capsule is often covered with fibrin. → Outer cortical layer: cortical, peripheral, epithelial cells are
MORPHOLOGY
• Septic infarcts - appearance is modified polygonal in shape, has an abundant cytoplasm, with
by the development of suppurative dendritic extension that contacts adjacent cells and abundant
necrosis thymocytes
→ Central medulla – the epithelial cells are densely packed
spindle-shaped; scant cytoplasm devoid of interconnecting
processes
• 2 major cell types:
→ Thymic epithelial cells at the periphery
→ Bone marrow-derived / immature T lymphocytes
• Whorls of medullary epithelial cells create Hassall
corpuscles
• Progenitor cells of marrow origin→ migrate to the thymus→
mature into T cells → exported to the periphery
• During adulthood the thymic production of T cells slowly
declines as the organ atrophies
• Other components found inside the thymus:
Fig. 10 Splenic infarct
Recent infarcts are hemorrhagic, whereas older, more fibrotic infarcts are a → Macrophages
pale yellow-gray color. → Dendritic cells
→ Minor population of B lymphocytes o rare neutrophils and
NEOPLASMS eosinophils
• Neoplastic involvement of the spleen is rare except in myeloid → Small population of neuroendocrine cells, which may
and lymphoid tumors. explain how neuroendocrine tumors arise in this organs
• Benign fibromas, osteomas, chondromas, lymphangiomas,
→ Scattered myoid (muscle-like) cells
and hemangiomas may arise
→ Most common: lymphangiomas and hemangiomas (often → Myoid cells are of particular interest because of the suspicion
that they play some role in the development of myasthenia gravis
cavernous in type)

A. DEVELOPMENTAL DISORDES
CONGENITAL ANOMALIES
THYMIC HYPOPLASIA OR APLASIA
• Complete absence of the spleen is rare and is usually
associated with other congenital abnormalities, such as • seen in DiGeorge syndrome
situs inversus and cardiac malformations. → marked by severe defects in cell-mediated immunity and
• Hypoplasia is a more common finding. variable abnormalities of parathyroid development
• Accessory spleens (spleniculi) - small, spherical associated with hypoparathyroidism
structures that are histologically and functionally identical to → often associated with other developmental defects as part of
the normal spleen. the 22q11 deletion syndrome
→ great clinical importance in some hematologic disorders,
such as hereditary spherocytosis and immune
ISOLATED THYMIC CYSTS
thrombocytopenia purpura, where splenectomy is used as a
treatment • usually discovered incidentally postmortem or during
surgery
• rarely exceed 4 cm in diameter, can be spherical or
RUPTURE arborizing, and are lined by stratified to columnar epithelium

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 21 of 24
• Fluid contents - serous or mucinous and are often modified
by hemorrhage

B. THYMIC HYPERPLASIA
• The term thymic hyperplasia is a bit misleading, since it
usually applies to the appearance of B-cell germinal centers
within the thymus, a finding that is referred to as thymic
follicular hyperplasia
• Most frequently encountered in myasthenia gravis, found
in 65% to 75% of cases
→ Similar thymic changes are sometimes encountered in
Fig. 11 Benign thymoma (medullary type)
Graves disease, systemic lupus erythematosus,
scleroderma, rheumatoid arthritis, and other autoimmune
disorders

C. THYMOMAS
• The designation “thymoma” is restricted to tumors of thymic
epithelial cells
→ also contain benign immature T cells (thymocytes)
• Classification relies on the most important prognostic
features, surgical stage and presence or absence of overt
cytologic features of malignancy.
• Three histologic subtypes: Fig. 12 Malignant thymoma, type I
→ Tumors that are cytologically benign and noninvasive
→ Tumors that are cytologically benign but invasive or Clinical features:
metastatic • 40% of thymomas - present with symptoms stemming from
impingement on mediastinal structures.
→ Tumors that are cytologically malignant (thymic
• 30% to 45% - detected in the course of evaluating patients
carcinoma) with myasthenia gravis.
• In all categories, the tumors usually occur in adults older • The rest - discovered incidentally during imaging or surgery
than 40 years of age (M=F); are rare in children • Other associated autoimmune disorders
• Most arise in the anterior superior mediastinum, but → hypogammaglobulinemia
sometimes they occur in the neck, thyroid, pulmonary hilus, → pure red cell aplasia
or elsewhere.
→ Graves disease
MORPHOLOGY → pernicious anemia
• most often composed of medullary-type → dermatomyositis-polymyositis
epithelial cells or a mixture of medullary → Cushing syndrome.
and cortical type. • Thymocytes that arise within thymomas give rise to long-lived
NONINVASIVE • The medullary type epithelial cells are CD4+ and CD8+ T cells, and cortical thymomas rich in
elongated or spindle-shaped. There is thymocytes → more likely to be associated with autoimmune
THYMOMA
usually a sparse infiltrate of thymocytes disease
• Mixed thymomas - there is an admixture • Llikely that abnormalities in the selection of T cells maturing
of polygonal cortical type epithelial cells within the environment of the neoplasm contribute to the
and a denser infiltrate of thymocytes. development of diverse autoimmune disorders.
• epithelial cells are most commonly of the
cortical variety, with abundant cytoplasm REFERENCES
and rounded vesicular nuclei, and are Robbins
usually mixed with numerous thymocytes.
2020 Trans (Incomplete; completed in this trans)
INVASIVE • invasive thymomas penetrate through
the capsule into surrounding structures.
THYMOMA
• With minimal invasion, complete excision
yields a 5-year survival rate of greater
than 90%, whereas extensive invasion is
associated with a 5-year survival rate of
less than 50%.
• Macroscopically: usually fleshy, obviously
invasive masses, sometimes
accompanied by metastases to sites
such as the lungs
• Microscopically: most are squamous cell
THYMIC carcinomas
CARCINOMA • The next most common variant is
lymphoepitheliomalike carcinoma, a
tumor composed of sheets of cells with
indistinct borders that bears a close
histologic resemblance to
nasopharyngeal carcinoma.

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 22 of 24
APPENDIX

Table 1. Subtypes of Hodgkin Lymphoma

Figure 1. Signals mediating cross talk in Hodgkin Lymphoma

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 23 of 24
Table 2. Subtypes of AML

Patho Diseases of White Blood Cells, Lymph Nodes, Spleen, and Thymus 24 of 24

You might also like