You are on page 1of 21

WBC, Lymph Nodes, Spleen, and Thymus

Far Eastern University – Nicanor Reyes Medical Foundation  Pluripotency and capacity for self-renewal are essential
Pathology A – Diseases of WBC, Lymph Nodes, Spleen and Thymus properties of HSCs for maintenance of hematopoiesis
Cherry Tan M.D.  Marrow response to short-term physiologic needs is regulated by
hematopoietic growth factors through the effects on committed
INTRODUCTION progenitors
 Myeloid tissues are the bone marrow and cells derived from it  Multipotent progenitors are more proliferative than HSCs but
 Lymphoid tissues are thymus, lymph nodes and spleen have a lesser capacity for self-renewal
 Stem cell factor (KIT ligand), FLT3-ligand act through receptors
DEVELOPMENT AND MAINTENANCE OF HEMATOPOIETIC TISSUES expressed in very early committed progenitors
 Blood cell progenitors first appear during third week of  Erythropoietin, GM-CSF, G-CSF and thrombopoietin act through
development receptors of restricted committed progenitors
 Hematopoietic stem cells arise several week later in the  Primary tumors of hematopoietic cells are the most important
mesoderm of the intraembryonic aorta / gonad / mesonephros disease the interfere marrow function
region  Tumors of hematopoietic origin are often associated with
 Liver becomes the chief site of hematopoiesis from the third mutations that block progenitor cell maturation or abrogate
month and shortly before birth growth factor dependence
 HSC also resides in the placenta and at the fourth month occurs at
the bone marrow DISORDERS OF WHITE CELLS
 At birth, the marrow serves as the chief site of hematopoiesis  Proliferative disorders – expansion of leukocytes
until puberty, where it only occur at the axial skeleton  Reactive proliferations – in the setting of infections or
 Formed elements of blood have a common origin from HSCs, the inflammation
pluripotent cells that sit at the apex of a hierarchy of bone  Neoplastic proliferations – less frequent, more important
marrow progenitors clinically
 Colony forming units produce colonies composed of specific kinds  Leukopenias – deficiency of leukocytes
of mature cells when grown in culture
Leukopenia
 An abnormally low WBC count that results from reduced numbers
of neutrophils (neutropenia, granulocytopenia)
 Lymphopenia is less common, but can be congenital or acquired,
commonly observed in HIV infection, drug therapy, autoimmune
disorders, malnutrition and acute viral infections
 Granulocytopenia is more common and often associated with
diminished granulocyte function

Neutropenia, Agranulocytosis
 Neutropenia is the reduction in the number of neutrophils in the
blood
 Agranulocytosis is the clinically significant reduction in neutrophils
which makes the individual susceptible for bacterial and fungal
infections

Pathogenesis
 Caused by:
1. Inadequate or ineffective granulopoiesis
 Suppression of HSCs – occurring in aplastic anemia and
infiltrative marrow disorders. Accompanied by anemia and
thrombocytopenia
 Suppression of committed granulocytic precursors –
exposure to drugs
 Ineffective hematopoiesis – defective precursors die in the
marrow as seen in megaloblastic anemia and
myelodysplastic syndromes
 Rare congenital conditions – defects in genes as seen in
Kostmann syndrome

Page 1 of 21
2. Accelerated destruction or sequestration of neutrophils REACTIVE PROLIFERATIONS OF WHITE CELLS AND LYMPH NODES
 Immunologically mediated injury – can be idiopathic, seen
in SLE or drug exposure Leukocytosis
 Splenomegaly – leads to modest neutropenia, associated  Increase in the number of WBC in the blood
with anemia and thrombocytopenia  Common reaction to a variety of inflammatory stress
 Increased peripheral utilization – bacterial, fungal or
rickettsial infections Pathogenesis
 Drug toxicity is the most common cause of agranulocytosis:
 Generalized suppression of hematopoiesis is due to
alkylating agents and anti-metabolites used in cancer
treatment
 Idiosyncratic reaction is caused by aminopyrine,
chloramphenicol, sulfonamides, thiouracil, chlorpromazine
and phenylbutazone
 Chlorpromazine and phenylbutazone produces a toxic
effect on granulocytic precursors on the marrow
 Sulfonamides, aminopyrine, thiouracil and antibodies
induce immune destruction of mature PMNs
 Monoclonal proliferations of large granular lymphocytes (LGL
leukemia) can also produce severe neutropenia

Morphology
 Alterations on the bone marrow vary with the etiology
 Hypercellular due to:  Factors that influence blood leukocyte count:
 Compensatory increase in granulocytic precursors due to  Size of myeloid and lymphoid precursor and storage cell pools
destruction of mature granulocyte in the marrow, thymus, circulation and peripheral tissues
 Ineffective granulopoiesis as seen in megaloblastic anemia  Rate of release of cells to the circulation
and myelodysplastic syndromes
 Proportion of cells adherent to vessel walls at any time
 Hypocellular as seen in agranulocytosis induced by suppression or (marginal pool)
destruction of granulocytic precursors  Rate of extravasation of cells into the tissue
 Infections are common consequences of agranulocytosis  Factors that cause leukocytosis:
 Severe and life threatening infections occur in the lungs,
urinary tract and kidneys
Neutrophilic Acute bacterial infections, especially those
 Increased risk for fungal infections caused by Candida and leukocytosis caused by pyogenic organisms; sterile
Aspergillus inflammation caused by, for example, tissue
 Site of infection has little WBC response and cause an necrosis (myocardial infarction, burns)
inflamed or enlarged regional lymph node Eosinophilic Allergic disorders such as asthma, hay fever,
 Ulcerating necrotizing lesions of the oral cavity (agranulocytic leukocytosis parasitic infestations; drug reactions; certain
angina) (eosinophilia) malignancies (e.g., Hodgkin and some non-
Hodgkin lymphomas); automimmune disorders
 Deep, undermined and gray to green-black necrotizing
(e.g., pemphigus, dermatitis herpetiformis) and
necrotic membranes inhabited by bacteria and fungi
some vasculitides; atheroembolic disease
 Can also occur in the skin, vagina, anus or GIT (transient)
Basophilic Rare, often indicative of a myeloproliferative
Clinical Features leukocytosis disease (e.g., chronic myelogenous leukemia)
 Infection, malaise, chills and fever followed by marked weakness (basophilia)
and fatigability Monocytosis Chronic infections (e.g., tuberculosis), bacterial
 Agranulocytosic infections may cause death within hours to days endocarditis, rickettsiosis, and malaria;
3 autoimmune disorders (e.g., systemic lupus
 Most serious infections present <500 per mm PMNs while <1,000
3 erythematosus); inflammatory bowel diseases
per mm is quite worrisome
(e.g., ulcerative colitis)
 Broad spectrum antibiotics are given whenever signs or symptoms Lymphocytosis Chronic infections (e.g., tuberculosis), bacterial
appear endocarditis, rickettsiosis, and malaria;
 G-CSF stimulates the production of granulocytes from marrow autoimmune disorders (e.g., systemic lupus
precursor and is given for myelosuppresive chemotherapy erythematosus); inflammatory bowel diseases
(e.g., ulcerative colitis)

Page 2 of 21
 In sepsis or severe inflammatory disorders, leukocytosis is  Neutrophils are prominent and centers of follicles undergo
accompanied by: necrosis due to pyogenic organisms
 Toxic granules – coarser and darker than the normal  Endothelial cells undergo hyperplasia
neutrophilic granules and represents abnormal azurophilic
(primary) granules b. Chronic nonspecific lymphadenitis
 Dohle bodies – patches of dilated ERs that appear as sky blue  Lymph nodes are nontender and inflammation is absent
cytoplasmic puddles  Common in the inguinal and axillary nodes
 Cytoplasmic vacuoles  Promote the appearance of organized collections of immune cells
in nonlymphoid tissues (tertiary lymphoid organs)

Morphology
1. Follicular hyperplasia – seen in humoral immune response
 Presence of large oblong germinal centers surrounded by naive
B cells
 Due to rheumatoid arthritis, toxoplasmosis and early stage HIV
 Morphologically similar to follicular lymphoma except for:
intact architecture, marked variation in size and shape of
follicles and frequency of mitotic figures, macrophages and light
and dark zones
 Acute viral infections in children cause appearance of large  Marginal zone b cell hyperplasia extends to the mantle zone
numbers of activated lymphocytes that resemble neoplastic 2. Paracortical hyperplasia – caused by T-cell mediated immune
lymphoid cells response
 Leukemoid reaction is the increase in granulocytes in blood due  Contains immunoblasts that are three to four times the site of
to severe infections resting lymphocytes with round nuclei, open chromatin,
prominent nucleoli and moderate amount of pale cytoplasm
Lymphadenitis  T cell zones expand and efface B cell follicles
 Activation of resident immune cells leads to morphological  Hypertrophy of sinusoidal and vascular endothelial cells
changes in lymph nodes accompanied by macrophages and eosinophils
 Primary follicles enlarge and develop pale staining germinal 3. Reticular hyperplasia – sinus histiocytosis, refers to an increase in
centers that are highly dynamic structures in which B cells make the size and number of cells lining the sinusoids
antibodies  Prominent in nodes draining cancers
 Paracortical T-cell zones may also undergo hyperplasia  Lining lymphatic endothelial cells are hypertrophied and
 Degree and pattern of morphologic changes are dependent on the macrophages are in great numbers
inciting stimulus and intensity of the response  Expansion and distension of the sinuses

a. Acute Nonspecific Lymphadenitis c. Hemophagocytic Lymphohistiocytes (HLH)


 Often occurs in the cervical region due to drainage of microbes or  Reactive condition marked by cytopenias and systemic
microbial products from infections of the teeth or tonsils inflammation related to macrophage activation
 In the axillary or inguinal region, often caused by infections of the  Referred to as macrophage activation syndrome
extremities
 Mesenteric nodes are affected due to acute appendicitis Pathogenesis
 Acute generalized lymphadenopathy is due to systemic viral  Common feature of all forms of HLH is systemic activation of
infections, particularly in children, and bacteremia macrophages and CD8+ cytotoxic T cells
 Leads to cytopenias and a shock-like picture referred to as
Morphology “cytokine storm” or SIRS
 Nodes involved are large, swollen, gray-red and painful, when  Familial forms are associated with several different mutations,
abscess forms, nodes become fluctuant and the overlying skin is impacting the ability of cytotoxic T cells and NK to form or deploy
red cytotoxic granules
 Suppurative infections penetrate through the capsule of the node  Most common trigger for HLH is infection, particularly EBV
and tract to the skin producing draining sinuses
 Healing is associated with scarring
 Prominence of large reactive germinal centers containing
numerous mitotic figures
 Macrophages often contain debris from bacteria and necrotic cells

Page 3 of 21
Clinical Features:  Can be gain of function of inappropriate increase in some
 Acute febrile illness associated with splenohepatomegaly normal activity.
 Hemophagocytosis is seen on bone marrow examination  Oncoproteins created by genomic aberrations often block
 Anemia, thrombocytopenia and very high levels of plasma ferritin normal maturation, turn on pro-growth signals, or protect cells
and soluble IL-2e receptor from apoptosis.
 Elevated liver function tests and triglyceride levels  Dominant negative oncogenic mutations in acute leukemia,
 DIC may be seen in coagulation studies involve transcription factors often present that interfere with
 Can progress rapidly to multiple organ failure, shock and death early stage of lymphoid or myeloid differentiation.
 Transcriptional regulator mutations directly enhance the self-
Neoplastic Proliferations of WBC: renewal of tumor cells, giving such stem-cell-like properties.

 Malignancies are clinically the most important disorders of WBC. Often involve tyrosine kinases that activate RAS and its
two downstream pathways PIK3/AKT and MAPK, driving
 Lymphoid Neoplasms cell to growth and Warburg metabolism.

Include diverse group of tumors of B-cells, T-cells, NK-cell  Proto-oncogenes are often activated by errors that occur during
origins. The phenotype closely resembles that of a particular antigen receptor gene rearrangement and diversification.
stage of normal lymphocyte maturation.
 Potentially oncogenic mutations occur most frequently in the
 Myeloid Neoplasms germinal center B cells during attempted Ab diversification.
 Arise from early hematopoietic progenitors.
 After stimulation, B cells enter the centers and upregulate
 Three categories are recognized:

expression of Activation-Induced Cytosine Deaminase (AID),
Acute Myeloid Leukemia – Immature progenitor cells
accumulate in the bone marrow. a DNA-modifying enzyme that is essential for Ig modification


Myeloid Dysplastic Syndromes – Associated with Class Switching – Intragenic recombination event which the
IgM heavy chain constant gene segment is replaced.
ineffective hematopoiesis and resultant peripheral blood 
cytopenias. Somatic Hypermutation – Created point mutations with Ig

Genes that may increase Ab affinity for antigens.
Chronic Myeloproliferative Disorders – Increased
production of one or more terminally differentiated  MYC oncogenes are also activated in germinal B-cell
myeloid element, usually leading to elevated counts. lymphomas by translocations.
 Histiocytoses
 Uncommon proliferative lesions of macrophages and 2.) Inherited Genetic Factors
dendritic cells.  Genetic diseases that promote genomic instability, such as
 A special type of immature dendritic cell, Langhan cells, give Bloom Syndrome, Fanconi anemia, and Ataxia telangiectasia are
rise to neoplastic disorders termed as Langhan cell at increased risk of acute leukemia.
histiocytoses.  Down syndrome and Type 1 neurofibromatosis are associated
with increased incidence of childhood leukemia.
ETIOLOGY AND PATHOGENESIS OF WHITE CELL NEOPLASIA
3.) Viruses
 Three lymphotrophic viruses have been implicated as causative
agents in particular lymphomas:
 HTLV1 (Human T-cell Leukemia Virus-1)

Associated with adult T-cell leukemia/lymphoma.
 EBV (Epstein-Barr Virus)

Found in a subset of Burkitt Lymphoma

Found in 30-40% of Hodgkin’s Lymphoma

Many B-cell lymphomas arising in the setting of T-cell
immunodeficiencies

Rare NK-cell lymphomas
 KSHV/HHV-8 (Kaposi sarcoma herpesvirus/Human herpesvirus 8)

Presents as malignant effusion, often in the pleural cavity.

4.) Chronic Inflammation


1.) Chromosomal Translocations and Acquired Mutations  Helicobacter pylori infections are associated with gastric B-cell
lymphomas, gluten-sensitive enteropathy, and intestinal T-cell
 Non-random chromosomal abnormalities, most commonly
translocations, are present in majority of WC neoplasia. lymphomas.
 HIV infection is associated with an increased risk of B-cell
 Mutated genes often play crucial roles in the development,
lymphomas that may arise virtually within any organ.
growth, or survival of the normal cells.
 Can be loss of function due to mutation of a dominant
negative protein that interferes with normal function.
Page 4 of 21
 T-cell dysregulation by HIV causes a systemic hyperplasia of  Lymphoid neoplasia can be suspected from features but
germinal center B cells associated with increased incidence of histologic examination of the lymph nodes or other tissus is
germinal center B-cell lymphomas. required for diagnosis.
 In AIDS, severe T-cell immunodeficiency further elevates the risk  Antigen receptor gene rearrangement generally precedes
of B-cell lymphoma, particularly those associated with EBV and transformation of lymphoid cells
KSV/HHV-8.  All daughter cells derived from malignant progenitor share
the same antigen receptor gene configuration and
5.) Iatrogenic Factors sequence, and synthesize identical antigen receptor proteins
 Radiation therapy and certain form of chemotherapy used to (Igs or T-cell receptors).
treat cancer, increases the risk of subsequent myeloid and  Most lymphoid neoplasms resemble some recognizable stage of
lymphoid neoplasms. B-cell or T-cell differentiation.
 This stems from the mutagenic effects of ionizing radiation and  85-90% are of B cell origin, the remainder from T-cell.
chemotherapeutic drugs on hemato-lymphoid progenitor cells.  Rarely are tumors of NK cell origin encountered.
 Markers recognized by Ab’s are helpful in characterization of
6.) Smoking lymphomas and leukemia.
 Incidence of acute myeloid leukemia is increased 1.3-2-fold in  Lymphoid Neoplasms are often associated with immune
smokers due to exposure to carcinogens (e.g. benzene). abnormalities.
 Loss of protective immunity (susceptibility to infection) and
Lymphoid Neoplasms breakdown of tolerance (autoimmunity) can be seen.
 Ironically, those with acquired immunodeficiency are at high
DEFINITIONS AND CLASSIFICATION risk of developing lymphoid neoplasms, particularly those
 Leukemia is a term used for neoplasms that present with caused by viruses.
widespread involvement of the bone marrow and usually but  Neoplastic B and T cells tend to recapitulate the behavior of
not always the peripheral blood. their normal counterpart.
 Lymphoma is used for proliferations that arise as discrete tissue  In follicular lymphomas, home to germinal centers.
masses.  In cutaneous T-cell lymphomas, home to the skin.
 With large group of lymphomas, Hodgkin Lymphoma (HL) is  Adhesion molecules and chemokine receptors govern the
segregated from all other forms, which constitute the Non- homing of the neoplastic cells.
Hodgkin Lymphoma (NHL).  They also recirculate through the lymphatics and peripheral
 Plasma cell neoplasm often arises in the bone marrow and only blood to the distant sites; as a result they are widely
infrequently involves lymph nodes or the peripheral blood. disseminated at the time of diagnosis.
 Clinical presentations :  Exceptions are:

Hodgkin Lymphoma – restricted to a group of lymph node
 2/3 of NHL and all HL present as enlarged non-tender lymph 
Marginal Zone B-cell Lymphoma – restricted to sites of chronic
nodes often >2cm in size. inflammation.
 Remaining 1/3 of NHL present with extranodal involvement.  HL spreads in an orderly fashion, while most forms of NHL
 Lymphocytic leukemia has signs and symptoms related to the spread widely in their early course in a less predictable fashion.
suppression of normal hematopoiesis by tumor cells in the
bone marrow. Precursor B- and T-cell Neoplasms
 Most common plasma cell neoplasm, multiple myeloma, 1.) Acute Lymphoblastic Leukemia / Lymphoma
present with bony destruction associated with pain due to  ALLs are neoplasms composed of immature B, or T cells which
pathologic fractures. are referred to as lymphoblasts.
 Other symptoms related to lymphoid tumors can be  About 80% are B-ALLs, which manifest as childhood acute
associated with proteins secreted by the tumors: leukemia.
 Whole Ig fragments in plasma cell tumors.  Less common T-ALLs tend to present in adolescent males as
 Cytokines from inflammatory cells in HL thymic lymphomas.
 Currently, the WHO classification scheme uses morphologic,  B-ALL uncommonly presents as a mass in the skin or a bone,
immunophenotypic, genotypic, and clinical features to sort the many T-ALLs present with or evolve to a leukemic picture.
lymphoid neoplasm into broad categories:  ALL is the most common cancer in children.
1. Precursor B-cell neoplasms (immature B-cells)
2. Peripheral B-cell neoplasms (mature B-cells) Pathogenesis
3. Precursor T-cell neoplasms (immature T-cells)  Chromosomal aberrations seen in ALL dysregulate the
4. Peripheral T-cell and NK-cell neoplasms (mature T-cells and expression and function of normal transcription factors required
NK-cells) for normal B- and T-cell development.
5. Hodgkin Lymphoma (Reed-Sternberg cells and variants)

Page 5 of 21
 70% have gain of function mutation in NOTCH1, a gene essential  Very immature T-ALLs are negative for CD3, 4, and, 8.
for T-cell development. Clinical Features
 B-ALLs have loss of function mutations in genes that are required  ALL and AML are genetically and immunophenotypically distinct,
for B-cell development, such as PAX5, E2A, and EBF, or a but they are clinically very similar.
balanced t(12;21) involving ETV6 and RUNX1.  Accumulation of neoplastic blasts in the bone marrow
 These mutations disturb the differentiation and promote suppresses the normal hematopoiesis by physical crowding,
maturation arrest; they induce increased self-renewal, stem competition with growth factors, and other unknown
cell-like phenotypes. mechanisms.
 Single mutations are not sufficient to produce ALLs.  ALLs have these more common characteristics:
Approximately 90% of ALLs have numerical or structural  Abrupt stormy onset within days to a few weeks.
chromosomal changes.  Symptoms related to depression of marrow function, such
 Most common is hyperploidy (>50 chromosomes), but as anemia, fever, infection due neutropenia, bleeding due to
hypoploidy and balanced translocations are also seen. thrombocytopenia.
 B-ALL and T-ALL are associated with completely different sets of  Mass effects caused by neoplastic infiltration, more
translocations, indicating that they are pathogenetically distinct. common in ALL, includes bone pain, generalized
lymphadenopathy, splenomegaly, hepatomegaly, testicular
Morphology enlargement, and in T-ALL compression of vessels and
airways in the mediastinum.
 CNS manifestations such as headache, vomiting, nerve
palsies due to meningeal spread, common in ALL.

Prognosis
 Aggressive chemotherapy cures 75-80% of cases, 95% obtain a
complete remission in children, 35-40% of adults are cured.
 Factors associated with worse prognosis:
 Age younger than 2 y/o, associated with infantile ALL with
translocations involving the MLL gene.
 In leukemic presentations, the marrow is hypercellular and  Presentation in adolescence or adulthood.
packed with lymphoblasts, which replace the normal marrow  Peripheral blood blast counts greater than 100,000,
elements. reflecting high tumor burden.
 Mediastinal thymic masses occur in 50-70% of T-ALLs, associated  Factors with favorable prognosis:
with lymphadenopathy and splenomegaly.  Age between 2-10 y/o
 Both T-ALL and B-ALL have tumor cells which have scant  Low WBC count
basophilic cytoplasm and nuclei larger than those small  Hyperdiploidy
lymphocytes.  Trisomy of chromosomes 4,7, and 10
 Nuclei chromatin is delicate and finely stippled; nucleoli are  Presence of a t(12;21)
usually small and often demarcated by a rim of condensed
chromatin. Peripheral B-cell Neoplasms
 Nuclear membrane is deeply subdivided. 1.) Chronic Lymphocytic Leukemia (CLL), Small Lymphocytic
 Mitotic rate is high, hence with the aggressive clinical behavior. Lymphoma (SLL)
 ALL vs. AML  CLL and SLL only differ in the degree of peripheral blood
 Compared to myeloblasts, lymphoblasts have more lymphocytosis.
condensed chromatin, less conspicuous nucleoli, and smaller  Most affected patients have sufficient lymphocytosis to fulfill
amounts of cytoplasm that usually lacks granules. 3
the diagnostic requirement for CLL >5000 per mm .
 CLL is the most common leukemia of adults in the Western.
Immunophenotype
 Immunostaining for Deoxynucleotidyl Transferase (TdT), a Pathogenesis
specialized DNA polymerase, expressed only in pre-B and pre-T  Chromosomal translocations are rare in CLL./SLL.
lymphoblasts, positive in 95% of cases.  Most common genetic anomalies are deletions of 13q14.3, 11q,
 B-ALLs are arrested at different stages. Usually express the B-cell 17p, and trisomy 12q.
marker CD19 and transcription factor PAX5 as well as CD10.  Two microRNAs, miR-15a and miR-16-1 are possible tumor
 CD10 is negative in very immature B-ALLs. suppressor genes involved in CLL/SLL.
 T-ALLs are also arrested at different stages. Usually expresses
CD1, 2, 5, and 7.

Page 6 of 21
 Ig genes of some CLL/SLL are somatically hypermutated, some  Hypogammaglobulinemia is common and contributes to an
are not, suggesting that the cell of origin may be either a increased susceptibility to infections.
postgerminal center memory B cell or a naïve B cell.  10-15% develop hemolytic anemia or thrombocytopenia due
 Tumors from naïve B cells pursue a more aggressive course. to autoantibodies made by non-neoplastic B cells.
 There is also gain of function in NOTCH1 in 10-18% of tumors.
 The growth of CLL/SLL is largely confined to proliferation centers, Prognosis
where tumor cells receive critical cue from microenvironment.  Extremely variable and depend primarily on the clinical stage.
 Stromal cells seem to express variety of factors that stimulate  Overall survival is 4-6 years, but more than 10 years in
the activity of the transcription factor NF-κB which promotes cell individuals with minimal tumor burdens.
growth and survival.  Factors with worse prognosis:
 Presence of deletions of 11q, 17p.
Morphology  A lack of somatic hypermutation.
 Expression of ZAP-70, a protein that augments signals
produced by the Ig receptors.
 Presence of NOTCH1 mutations.
 Another factor that impacts patient survival is the tendency of
CLL/SLL to transform to a more aggressive tumor.
 Most commonly takes the form is to diffuse large B-cell
lymphoma, so called Richter Syndrome.

2.) Follicular Lymphoma


 The most common form of NHL in the US.
 Lymph nodes are diffusely effaced by an infiltrate of  Presents in the middle age and afflicts males and females
predominantly small lymphocytes 6-12 µm in diameter with equally.
round to slightly irregular nuclei, condensed chromatin, and
scant cytoplasm. Pathogensis
 Admixed with activated lymphocytes that often gathers in loose  Arises from germinal center B cells, strongly associated with
aggregates referred to as proliferation centers, which contain chromosomal translocations involving BCL2.
mitotically active cells.  Hallmark is a (14;18) translocation that juxtaposes the IGH locus
 Proliferation centers are pathognomonic for CLL/SLL. on chromosome 14 and the BCL2 locus on chromosome 18.
 Some of the cells in the blood usually disrupted in the process of  BCL2 becomes overexpressed and antagonizes apoptosis and
making smears produce a so-called smudge cells. promotes survival of follicular lymphoma.
 Bone marrow is almost always involved by interstitial infiltrates  Follicular lymphoma is devoid of apoptotic cells.
or aggregates of tumor cells, which are also always seen in the  MLL2 gene mutations are present in 90%, it encodes for histone
splenic white/red pulp and hepatic portal tracts. methyltransferases that regulate gene expression, suggesting
epigenetic abnormalities.
Immunophenotypes  Follicular lymphoma cells grow in lymph nodes within a network
 CLL/SLL express the pan B-cell markers CD19 and CD20, as well of reactive follicular dendritic cells admixed with macrophages
as CD23, CD5. and T-cells.
 Low levels expression of surface Ig (usually IgM or IgM and IgD)
is also typical. Morphology
 Predominantly nodular or nodular and diffuse growth pattern
Clinical Features observed in lymph nodes.
 Patients are often asymptomatic at diagnosis.  Two principal cell types are present in varying proportions:
 Symptoms are nonspecific, including easy fatigability, weight  Small cells with irregular or cleaved nuclear contous and
loss, and anorexia. scant cytoplasm referred to as centrocytes.
 Generalized lymphadenopathy and splenomegaly.  Large cells with open nuclear chromatin, several nucleoli, and
 Leukocyte count is highly variable, leukopenia can be seen in modest amounts of cytoplasm called centroblasts.
individuals with SLL, and marrow involvement.  Small cleaved cells are in the majority.
3  Peripheral blood involvement sufficient to produce
 Counts in excess of 200,000/mm are sometimes seen in CLL
3
patients with heavy tumor burdens. lymphocytosis usually <20,000/mm is seen in 10% of cases.
 A small monoclonal Ig spike is present in the blood.  Bone marrow involvement in 85% of cases characteristically
 CLL/SLL disrupts the normal immune function through takes the form of paratrabecular lymphoid aggregates.
uncertain mechanisms.  Splenic white pulp and hepatic portal triads are also involved.

Page 7 of 21
Immunophenotype Subtypes
 Neoplastic cells closely resemble normal germinal center B-cells. 1.) Immunodeficiency-associated large B-cell lymphoma
 Expresses CD19, CD20, CD10, surface Ig, and BCL6.  Occurs in the setting of severe T-cell deficiency (e.g. HIV)
 Unlike CLL/SLL, CD5 is not expressed.  Neoplastic B cells are usually infected with EBV, which plays a
Clinical Features critical pathogenic role.
 Tends to present with painless, generalized lymphadenopathy.  Restoration of T-cell immunity may lead to regression.
 Involvement of extranodal sites, such as GIT, CNS, or testis is
relatively uncommon. 2.) Primary effusion lymphoma
 Although incurable, it follows an indolent waxing and waning  Presents as malignant pleural or ascetic effusion.
course.  Mostly in patients with advanced HIV infection or older adults.
 Survival is not improved by aggressive therapy; hence, the usual  Tumor cells are often anaplastic, typically fail to express surface
approach is to palliate the patient with low-dose chemo- or markers, but have clonal IgH gene rearrangements.
immunotherapy when they become symptomatic.  Tumor cells are infected with KSHV/HHV-8.
 Histologic transformation occurs in 30-50%, most commonly to
diffuse large B-cell lymphoma. Less commonly, tumors Clinical Features
resembling Burkitt lymphoma emerge associated with  Typically presents as a rapidly enlarging mass at a nodal or
translocations involving MYC. extranodal site (GIT, skin, bone, brain).
 Median survival is less than 1 year after transformation.  Waldeyer’s ring (Oropharyngeal Lymphoid Tissue) is commonly
involved.
3.) Diffuse Large B-Cell Lymphoma  Primary or secondary liver or spleen involvement takes the form
 DLBCL is the most common form of NHL. of destructive masses.
 Median patient age is about 60 years old.  Bone marrow involvement is uncommon and usually occurs late.
 Male is more affected then females.  Rarely, a leukemic picture emerges.
 Rapidly fatal without treatment.
Pathogenesis  Intensive combination chemotherapy, 60-80% have
 DLCBL is a molecularly heterogeneous disease. remission and 40-50% are cured.
 Frequent pathogenic event is dysregulation of BCL6, a DNA-
binding zinc-finger transcriptional repressor required for the 4.) Burkitt Lymphoma
formation of normal germinal center.  Categories:
 Another 10-20% have t(14;18) mutations, leading to  African Burkitt Lymphoma (Endemic)
overexpression of BCL2, an antiapoptotic protein.  Sporadic Burkitt Lymphoma (Non-endemic)
 Aggressive lymphomas in HIV-infected individuals
Morphology
 Common features are relatively large cell size, usually 4-5 times Pathogenesis
the diameter of small lymphocyte, and a diffuse pattern of  All forms are highly associated with translocations of the MYC
growth. gene on chromosome 8 leading to increased MYC protein levels.
 Tumor cells have round or oval nucleus that appears vesicular  MYC is a transcriptional regulator that increases expression of
due to migration of chromatin to nuclear membrane, but large genes required for aerobic glycolysis, so-called Warburg effect.
multilobated or cleaved nuclei are prominent.  It is believed to be the fastes growing human tumor.
 Nucleoli may be 2-3 times in number and located adjacent to the  The translocation partner for MYC is usually the IgH locus, t(8;14)
nuclear membrane.  Essentially, all endemic Burkitt lymphoma, are latently infected
 Cytoplasm is usually abundant and may be pale or basophilic. with EBV, which is also present in 25% of HIV-associated tumors
 More anaplastic tumors may even contain multinucleated cells and 20% of sporadic cases.
with large inclusion-like nucleoli resembling Reed-Sternberg cells
Morphology
Immunophenotype  Tissues are effaced by diffuse infiltrate of intermediate-sized
 Mature B-cell tumors expressing CD19 and CD20 and show lymphoid cells 10-25 µm in diameter with round or oval nuclei,
variable expression of germinal center B-cell markers, such as coarse chromatin, several nucleoli, and a moderate amount of
CD10 and BCL6. cytoplasm.
 Most have surface Igs.  Tumor exhibits a high mitotic index and contains numerous
apoptotic cells.
 The nuclear remnants are phagocytosed by interspersed benign
macrophages. These have abundant clear cytoplasm, creating a
starry sky appearance.

Page 8 of 21

 When bone marrow is involved, aspirates reveal tumor cells with Smoldering myeloma refers to another uncommon variant
defined by lack of symptoms and high plasma M
slightly clumped nuclear chromatin, 2-5 distinct nucleoli, and a component.
royal blue cytoplasm containing clear vacuoles.
 Waldenstrom macroglobulinemia

A syndrome in which high levels of IgM lead to symptoms
Immunophenotype related to hyperviscosity of the blood.

 These are mature B cell tumors expressing surface IgM, CD19, It occurs in older adults,most commonly in association with
lymphoplasmacytic lymphoma.
CD20, CD10, and BCL6.
 Heavy-chain disease

 Burkitt lymphoma almost always fails to express the anti- A rare monoclonal gammopathy.
apoptotic protein BCL2, unlike other germinal center tumors. 
Seen with lymphoplasmacytic lymphoma and unusual
small bowel marginal zone lymphoma occurring in
malnourished populations (Mediterranean Lymphoma).
Clinical Features 
Common feature is the synthesis and secretion of free heavy-
 Endemic and sporadic are found mainly in children or young chain fragment.
adults, overall it accounts for 30% of childhood NHL.  Primary or Immunocyte-Associated amyloidosis
 Most manifest extranodal sites. 
Results from a monoclonal proliferation of plasma cell
secreting light chain (usually λ isotype) that are deposited
 Endemic often presents as a mass involving the mandible, and as amyloid.
shows an unusual predilection for involvement of abdominal 
Some have overt multiple myeloma but others have only a
viscera particularly the kidneys, ovaries, and adrenals. minor clonal population of plasma cells in the marrow.
 Sporadic most often appears as mass involving the ileocecum  Monoclonal Gammopathy of Undetermined Significance
and peritoneum. (MGUS)

 Involvement of the bone marrow and peripheral blood is Applied to patients without signs or symptoms who have
small to moderately large M components in the blood.
uncommon. 
Very common in older adults and has a low but constant rate
 This is very aggressive but responds well to intensive of transformation to multiple myeloma.
chemotherapy.
1.) Multiple Myeloma
Plasma Cell Neoplasms and Related Disorders  A plasma cell neoplasm commonly associated with lytic bone
 These B-cell proliferations contain neoplastic plasma cells that lesions, hypercalcemia, renal failure, and acquired immune
virtually always secrete a monoclonal Ig or Ig fragment which abnormalities.
serves as a tumor marker and often have pathologic
consequences. Pathogenesis
 Plasma cell neoplasms (often called dyscrasia) account for 15%  Associated with frequent rearrangements involving the IgH locus
of deaths caused by lymphoid neoplasm. and various proto-oncogenes.
 Monoclonal Ig in the blood is called the M component, in  Included are frequent translocations with Ig heavy-chain on
reference to myeloma. 14q2, cyclin D1 on chromosome 11q13, and cuclin D3 on
 They have 160,000 molecular weight making them restricted chromosome 6p21.
to the plasma and ECF, and not in the urine in the absence of  A deletion of chromosome 17p that involves the TP53 tumor
glomerular damage. suppressor locus also occurs.
 Neoplastic cells often synthesize excess light chains along with  The proliferation and survival of myeloma cells are dependent
complete Igs. on several cytokines, most notably IL-6, which is an important
 Highly sensitive for free light chains in the blood is available. growth factor for plasma cells.
 The level of free light chains is usually elevated and is  High serum levels of IL-6 are seen in patients with active
markedly skewed toward one light chain at the expense of disease, and associated with poor prognosis.
the seconds.  Factors produced by neoplastic plasma cells mediate bone
Since free light chains are small, they can be concentrated in destruction, the major pathologic feature of multiple myeloma:
the urine, referred to as Bence-Jones proteins.  Myeloma derived MIP1α upregulates the expression of
 Abnormal proteins are associated with the ff. entities: RANKL by bone marrow stromal cells that in turn activates
 Multiple Myeloma osteoclasts.

Also called Plasma Cel Myeloma.

 Modulators of Wnt pathway are potent inhibitors of
Most important plasma cell neoplasm, usually presents as osteoblast functions.
tumorous masses scattered throughout the skeletal
system.  Net effect is marked increase in bone resorption, which leads

Solitary myeloma (Plasmacytoma) is an infrequent variant to hypercalcemia.
that presents as a single massin bone or soft tissue.

Page 9 of 21
Morphology  Renal insufficiency trails only infections.
 Usually presents as destructive plasma cell tumors  Pathogenesis of renal involvement is multifactorial.
(Plasmacytomas) involving the axial skelton.  Most important is the Bence-Jones proteinuria, as the
 Bones commonly affected (in descending order) are vertebral excreted light Ig chains are toxic to renal tubular epithelial
column, ribs, skull, pelvis, femur, clavicle, and scapula. cells.
 It begins in the medullary cavity, erode cancellous bone, and  Certain light chains are prone to cause amyloidosis of the AL
progressively destroy the bony cortex. type, which can exacerbate renal dysfunction.
 Bone lesions appear radiographically as punched-out defects  In 99% of patients, lab analyses reveal increased levels of Igs in
usually 1-4 cm in diameter, and consist of soft, gelatinous, red the blood, or light chain Igs in the urine.
tumor masses.  The monoclonal Igs are usually first detected as abnormal
 The marrow contains an increased number of plasma cells, proteins pikes in the serum.
which usually constitute 30% of the cellularity.  Most myelomas are associated with more than 3gm/dL of
 Plasma cells may infiltrate the interstitium. serum Ig, or more than 6 mg/dL of Bence jones urine protein.
 Malignant plasma cells have perinuclear clearing due to  Most common is IgG, followed by IgA.
prominent Golgi bodies and have an eccentrically placed  IgM, IgD, or IgE are rare.
nucleus.  Marrow involvement often gives rise to a normocytic
 Plasmablasts may appear normal with vesicular nuclear normochromic anemia, sometimes accompanied by leukopenia
chromatin and a prominent single nucleolus or appear as and thrombocytopenia.
bizzare, multinucleated cell, such variants include:  Prognosis is variable.

2.) Solitary Myeloma (Plasmacytoma)


 3-5% of plasma cell neoplasms present as a solitary lesion of
bone or soft tissue.
 Tend to occur in same locations in multiple myeloma.
 Extraoseus lesions often involve the lungs, oronasopharynx, or
nasal sinuses.
 Modest elevations of M proteins in the blood or urine.
 Solitary osseous plasmacytoma inevitably progress to multiple
 Flame Cells – with fiery red cytoplasm myeloma, but it can take 10-20 years or longer.
 Mott cells – with multiple grape-like cytoplasmic droplets,  Extraosseous plasmacytomas are cured by local resection.
containing fibris, crystalline rods, and globules. These
inclusions are known as Russell Bodies (cytoplasmic) or 3.) Smoldering Myeloma
Dutcher Bodies (nuclear).  Defines a middle ground between multiple myeloma and
 High levels of M proteins cause red cells to form rouleaux. monoclonal gammopathy of uncertain significance.
 Rarely, tumor cells flood the peripheral blood giving rise to  Plasma cells make up 10-30% of marrow cellularity.
plasma cell leukemia.  Serum M protein is > 3 gm/dL, but patients are asymptomatic.
 Bence jones proteins are excreted in the urine contribute to a  75% progress to multiple myeloma over 15 year period.
form of renal disease called myeloma kidney.
4.) Monoclonal Gammopathy of Uncertain Significance (MGUS)
Immunophenotype  The most common plasma cell dyscrasia.
 Positive for CD138, an adhesion molecule known as syndecan-1.  Approximately 1$ develop a symptomatic plasma cell neoplasm,
 They also often express CD56. usually multiple myeloma.

Clinical Features 5.) Lymphoplasmacytic Lymphoma


 The presentation of myeloma arise from: th th
 A B-cell neoplasm usually presents in the 6 or 7 decade of life.
 Effects of plasma cell growth in tissues particularly the bone.  Superficially resembles CLL/SLL but differs because a substantial
Production of excessive Igs, which often have abnormal fraction of tumor cells undergo terminal differentiation to
physicochemical properties. plasma cells.
 Suppression of normal humoral immunity.  Most commonly, the plasma cell secretes monoclonal IgM, often
 Bone resorption leads to pathologic fractures and chronic pain. in amounts to cause hyperviscosity syndrome known as
 The attendant hypercalcemia can give rise to neurologic Waldenstrom macroglobulinemia.
manifestations (e.g. confusion, weakness, lethargy, constipation,  Unlike multiple myeloma, complications from light chain Igs are
and polyuria). rare and bone destruction does not occur.
 Decreased normal Ig leads to recurrent bacterial infection.

Page 10 of 21
Pathogenesis  Results to upregulation of D1 to promote G1-S phase
 Associated with acquired mutations in MYD88, which encodes progression.
an adaptor protein that participates in signaling events that
activate NF-kB, and also augment signals downstream of the B- Morphology
cell receptor which may promote the growth and survival of  Generalized lymphadenopathy, 20-40% have peripheral blood
tumor cells. involvement.
 Frequent sites of extranodal involvement include bone, marrow,
Morphology spleen, liver, and gut.
 Marrow contains an infiltrate of lymphocytes, plasma cells, and  Mucosal involvement of small bowel produces polyp-like lesion
plasmacytoid lymphocytes in varying proportions. (lymphomatoid polyposis), of all NHL, mantle cell lymphoma is
 Often accompanied by mast cell hyperplasia. most likely to spread in this fashion.
 Some contain a population of larger lymphoid cells with more  Nodal tumor cells may surround reactive germinal centers to
vesicular nuclear chromatin and prominent nuclei. produce nodular appearance.
 Period acid-schiff positive inclusions contain Ig frequently seen in  Typically, proliferations consist of a homogeneous population of
the cytoplasm (Russell bodies) or the nucleus (Dutcher bodies). small lymphocytes with irregular to occasionally deeply clefted
 At diagnosis, dissemination to lymph node, spleen and liver has unclear contours.
occurred.  Centroblasts/proliferative centers are absent unlike that of
 Infiltration of nerve roots, meninges, or rarely the brain. CLL/SLL, respectively.
 In most cases, nuclear chromatin is condensed, nucleoli are
Immunophenotype inconspicuous, and cytoplasm is scant.
 Expresses B-cell markers CD20 and surface Ig, whereas the
plasma cell secretes the same Ig that is expressed on the surface Immunophenotype
of lymphoid cells.  Express high levels of cyclin D1.
 Usually IgM but can also be IgG or IgA.  Most express CD19, CD20, and moderately high levels of surface
Ig (usually IgM and IgD).
Clinical Features  Usually CD5+ and CD23-.
 Dominant presenting complains are non-specific.  IgH lacks somatic hypermutation, supporting an origin from a
 Half of patients have lymphadenopathy, hepatomegaly, and naïve B cell.
splenomegaly.
 Anemia caused by marrow infiltration is common. Clinical Features
 10% have autoimmune hemolysis caused by cold agglutinins,  Most common is painless lymphadenopathy.
IgM antibodies that bind to red cells.  Related to involvement of the spleen and gut.
 IgM have additional signs and symptoms, because of its large  Prognosis is poor, median survival is only 3-4 years.
size, at high concentrations IgM increases viscosity of the blood  Non-curable with conventional chemo, most patient succumb to
giving rise to hyperviscosity syndrome leading to: organ dysfunction.
 Visual Impairment associated with venous congestion.
 Neurologic Problems due to sluggish blood flow. Marginal Zone Lymphomas
 Bleeding due to formation of complexes.  Encompasses a heterogeneous group of B-cell tumors that arise
 Cryoglobulinemia resulting from precipitation of Ig. within lymph nodes, spleen, or extranodal tissues.
 Lymphoplasmacytic lymphoma is incurable progressive dse.  Initially recognized at mucosal sites referred to as mucosa-
 Elevated IgM can be alleviated by plasmapheresis. associated lymphoid tumors (MALTomas).
 Tumor growth can be controlled.  Most tumor cells show evidence of somatic hypermutation and
 Median survival is about 4 years. considered to be of memory B-cell origin.
 Three exceptional characteristics:
Mantle Cell Lymphoma  Often arise within tissues involved by chronic inflammatory
 Uncommon lymphoid neoplasm. disorders of autoimmune or infectious etiology.

Example is salivary gland in Sjogren Syndrome.
 Closely resemble the normal mantle zone B cells that surround 
Thyroid gland in Hashimoto’s thyroiditis
germinal centers. 
Stomach in Helicobacter gastritis.
 Remain localized for prolonged periods.
Pathogenesis  May regress if inciting agent is eradicated.
 All have an (11;14) translocation involving the IgH locus on  These suggest extranodal margicnal lymphomas arising in
chromosome 14 and cyclin D1 on chromosome 11, leading to chronically inflamed tissues lie on a continuum between
overexpression of cyclin D1. reactive lymphoid hyperplasia and full-blown lymphoma.

Page 11 of 21
 The disease begins as polyclonal immune reaction, with the Clinical Features
acquisition of still unknown initiating mutations, a B-cell clone  Results from infiltration of the bone marrow, liver, & spleen.
emerges that sill depends on antigen-stimulated T-helper cell  Splenomegaly is often massive, most common physical finding.
signals.  Hepatomegaly is less common.
 At this stage, removal of the inciting agent may cause tumor  Lymphadenopathy is rare.
involution.  Pancytopenia resulting from marrow involvement and splenic
sequestration is seen.
Hairy Cell Leukemia  1/3 of affected patients present with infections.
 Rare, distinctive B-cell neoplasm constitutes 2% of al leukemia.  Increased incidence of atypical mycobacterial infections.
 Predominantly a disease of middle-aged white males, median  Related to frequent unexplained monocytopenia.
age of 55 years old.  Exceptionally sensitive to gentle chemotherapy, which produces
 Male to female ratio of 5:1 long lasting remissions.
 The tumor replaces after 5 years but still responds to the
Pathogenesis same agent.
 Associated with 90% of cases with activating point mutation in  BRAF Inhibitors produce excellent responses.
the serine/threonine kinase BRAF, which is immediately
downstream of RAS in the MAPK cascade. Peripheral T-Cell and NK-Cell Neoplasms
 Specific mutation is a Valine to Glutamate substitution at the  .
th
600 residue (also found in melanomas and langerhan cell
histiocytosis). 1.) Peripheral T-Cell Lymphoma, Unspecified
 Under wastebasket diagnosis, since these are not easily
Morphology categorized.
 No morphologic feature is pathognomonic.
 These tumors efface the lymph nodes diffusely and are typically
composed of a pleomorphic mixture of variably sized malignant
T cells.
 Brisk neoangiogenesis can be seen.
 All peripheral T-cell lymphomas are derived from mature T-cells,
usually express CD3, CD5, and either αβ or γ∆ T-cell receptors.
 Most patients present with generalized lymphadenopathy,
sometimes accompanied with eosinophilia, pruritus, fever, and
weight loss.

 Appearance of leukemic cells which have fine hair-like 2.) Anaplastic Large-Cell Lymphoma (ALK Positive)
projections that are best recognized under phase-contrast  Uncommon entity is defined by the presence of rearrangements
microscope. in the ALK gene on chromosome 2p23.
 Hairy cells have round, oblong, or reniform nuclei and moderate  These break the ALK locus and lead to formation of chimeric
amounts of pale blue cytoplasm with thread-like or bleb-like genes encoding for ALK fusion proteins.
extensions.  The tumor is typically composed of large anaplastic cells, some
 Marrow is involved by a diffuse interstitial infiltrate of cells with contain horseshoe-shaped nuclei and voluminous cytoplasm (so-
oblong and reniform nuclei. called hallmark cells).
 Usually cannot be aspirated because these cells are enmeshed in  Tumor cells often cluster about venules and infiltrate lymphoid
an ECM composed of reticulin fibrils ( a clinical difficulty called as sinuses, mimicking the appearance of metastatic carcinoma.
dry tap), only seen in marrow biopsies.  T-cell lymphomas with ALK rearrangement occur in children or
 Splenic red pulp is usually heavily infiltrated, with obliteration of young adult involving soft tissues, and have good prognosis.
the white pulp; hepatic portal triads are also affected.  Cure rate is 75-80%.
 Both ALK+ and ALK- tumors express CD30, a member of TNF
Immunophenotype receptor family, recombinant antibodies bind and kill CD30-
 Express the pan-B-cell markers CD19 and CD20, surface Ig, and expressing cells.
certain relatively distinctive markers such as CD11c, CD25,
CD103, and annexin A1.

Page 12 of 21
3.) Adult T-cell leukemia /lymphoma 6.) Extranodal NK/T-Cell Lymphoma
 Neoplasm of CD4+ T cells is only observed in adults infected by  Rare, presents most commonly as a destructive nasopharyngeal
HTLV-1. mass, less common site include the testis and the skin.
 Common findings include skin lesions, generalized  Tumor cells infiltrate typically surrounds and invades small
lymphadenopathy, hepatosplenomegaly, peripheral blood vessels, leading to extensive ischemic necrosis.
lymphocytosis, and hypercalcemia.  Large azurophilic granules are seen in the cytoplasm of the cells.
 Appearance of tumor cells has multilobated nuclei (cloverleaf or  High associated with EBV, no consistent chromosome aberration
flower cell). has been described.
 They often contain clonal HTLV-1 provirus.  Most are highly aggressive neoplasms that respond well to
 HTLV encodes for a protein called Tax that activate NF-kb, which radiation but are resistant to chemotherapy.
enhances lymphocyte growth and survival.
 Most patients present with rapidly progressive disease that is Hodgkin Lymphoma
fatal within months to a year.

4.) Mycosis Fungoides / Sezary Syndrome


 These are different manifestations of a tumor of CD4+ T-cells
that home to the skin.
 Cutaneous lesions of Mycosis fungoides progress through three
distinct stages:
 Inflammatory premycotic phase.
 Plaque Phase
 Tumor Phase
 Encompasses a group of lymphoid neoplasms that differ from
 Histologically, the dermis and upper dermis are infiltrated by NHL. HL arises in a single nod or chain of nodes and spread first
neoplastic cells having a cerebriform appearance due to marked to anatomically contiguous lymphoid tissue.
infolding of the nuclear membrane.  It is characterized by the presence of neoplastic cells called
 Late disease progression leads to extracutaneous spread to Reed-Sternberg Cells (RS Cells).
lymph nodes or bone marrow.  These cells release factors that induce accumulation of reactive
 Sezary syndrome, a variant in which skin involvement manifests lymphocytes, macrophages, and granulocytes.
as generalized exfoliative erythroderma, and there is associated  In most HLs, the RS cells are mostly derived from germinal
leukemia of Sezary cells with characteristic cerebriform nuclei. center or post-germinal center B cells.
 Tumor cells express adhesion molecule cutaneous leukocyte  Average age at diagnosis is 32 years old.
antigen (CLA) and chemokine receptor CCR4 and CCR10.  First human cancer to be successfully treated with radiation and
chemotherapy, and is curable in most cases.
5.) Large Granular Lymphocytic Leukemia
 T-cell and NK-cell variants of this rare neoplasm occur in adults. Classification
 Usually present with mild to moderate lymphocytosis and 1. Nodular Sclerosis
splenomegaly. 2. Mixed Cellularity
 Lymphadenopathy and hepatomegaly are usually absent. 3. Lymphocyte-Rich
 30-40% have STAT3 mutations, which functions as downstream 4. Lymphocyte-Depletion
of cytokine receptors 5. Lymphocyte-Predominance
 Tumor cells are large lymphocytes with abundant blue cytoplasm
and a few coarse azurophilic granules.
 In the first four subtypes, the RS cells have similar
 Marrow usually contains sparse interstitial lymphocytic immunophenotype, these are often clumped together in the
infiltrates, which can be difficult to appreciate without classical HL.
immunohistochemical stain.  The remaining, lymphocyte predominance, have RS cells that
 T-cell variants are CD3+, NK-cell variants are CD3- and CD56+ have distinctive B-cell immunophenotype.
 Neutropenia and anemia dominate the clinical picture.
 Neutropenia is accompanied by striking decrease in late Pathogenesis
myeloid forms in the marrow.
 The Ig genes of RS cells have undergone both V(D)J
 Rarely, pure red cell aplasia is seen. recombination and somatic hypermutation, establishing an
 Increased incidence of rheumatologic disorder origin from a germinal center or post-germinal center B cell.
 Felty syndrome is seen in some patients, a triad of  RS cells fail to express most B-cell-specific genes, including the Ig.
rheumatoid arthritis, splenomegaly, and neutropenia.  Presumably it results from widespread epigenetic changes.
 Activation of NF-kB is a common event in classical HL.

Page 13 of 21
 This can occur via: 1.) Nodular Sclerosis
 Activated by EBV infection or by some other mechanisms  Most common form, 65-70% of cases.
that promote lymphocyte survival and proliferation.  Characterized by the presence of lacunar variant RS cells and the
 EBV expresses Latent Membrane Protein 1 (LMP1) that deposition of collagen in bands that divide the involved lymph
transmit signals that upregulate NF-kB. node into circumscribed nodules.
 Activation of NF-kb may occur in EBV tumors as a result of  Fibrosis may be scant t abundant.
loss of function in IkB or A20 which are negative regulators of  RS cells are found in a polymorphous background of T cells,
NF-kB. eosinophils, plasma cells, and macrophages.
 It is hypothesized that activation of NF-kB by EBV rescues  This subtype is uncommonly associated with EBV.
crippled germinal center B cells that cannot express Igs from  Immunophenotype:
apoptosis.  Positive for PAX5, CD15, and CD30
 Negative for other B-cell markers, T-cell markers, and CD45.
 The accumulation of reactive cells in classical HL occurs in  Occurs with equal frequency in male and female.
response to cytokines, chemokines, and other factors secreted  Has the propensity to involve the lower cervical, supraclavicular,
by the RS cells. and mediastinal lymph nodes.
 Prognosis is excellent.

2.) Mixed Cellularity Type


 Constitutes about 20-25% of the cases.
 Lymph nodes are diffusely effaced by a heterogeneous cellular
infiltrate (T-cell, eosinophil, plasma cells, and macrophages).
 RS cells are mononuclear variants, usually plentiful.
 70% are infected with EBV.
 Immunophenotype is same with Nodular Sclerosis.
 More common in males, more likely associated with older age.
 Accompanied by systemic symptoms such as night sweats, and
weight loss, but overall prognosis is good.

3.) Lymphocyte-Rich Type


 An uncommon form in which reactive lymphocyte make up the
vast majority of the cellular infiltrate.
 Lymph nodes are usually effaced, but vague nodularity to
presence of residual B-cell follicles.
 The attracted cells produce factors that support the growth and  There is presence of mononuclear variants of RS cells.
survival of the tumor cells and modify the reactive cell response.  Classic immunophenotype.
 RS cells are aneuploid and possess direrse clonal chromosomal  40% is associated with EBV.
aberrations.  Very good prognonsis.
 Copy number gains in the REL proto-oncogenes on chromosome
2p are common and may contribute to increased NF-kB activity. 4.) Lymphocyte Depletion Type
 Least common form, 5% of cases.
Morphology  Characterized by paucity of lymphocytes and a reactive
 RS cells are large cells (45µm in diameter) with multiple nuclei or abundance of RS cells or their pleomorphic variants.
a single nucleus with multiple lobes, each with large inclusion-  RS cells are 90% infected with EBV.
like nucleolus about the size of the small lymphocyte.  Occurs predominantly in adults, HIV+ patients, and non-
 Mononuclear variants contain a single nucleus with large industrialized countries.
inclusion-like nucleolus.  Overall outcome is somewhat less favorable than the others.
 Lacunar cells have more delicate, folded, or multilobate nuclei
and abundant pale cytoplasm, often disrupted during cutting 5.) Lymphocyte Predominance Type
sections.  Uncommon, Non-classical variant accounts for 5% of cases.
 In classic forms, RS undergo peculiar form of cell death in which  Nodes are effaced by a nodular infiltrate of small lymphocytes
the cells shrink and become pyknotic, a process described as admixed with variable number of macrophages.
mummification.  Classic RS cells are usually difficult to find, but Lymphocytic and
 Lymphohistiocytic variants (L&H) with polypoid nuclei, Histiocytic variants are common having a multilobed nucleus
inconspicuous nucleoli, and moderately abundant cytoplasm are resembling a popcorn kernel (popcorn cell).
of lymphocyte predominance type.

Page 14 of 21
 Eosinophils or plasma cells are usually absent. Acute Myeloid Leukemia (AML)
 L&H variants express B-cell markers typical of germinal center B  Caused by acquired oncogenic mutation that impedes
cells, such as CD20 and BCL6 and are usually negative of CD15 differentiation, leading to accumulation of immature myeloid
and CD30. blasts in the marrow.
 Replacement of marrow with blasts produces marrow failure
Clinical Features and complications leading to anemia, thrombocytopenia, and
neutropenia.

Classification
 Divided into 4 classification
 First classification includes forms of AML with particular genetic
aberrations.
 AML arising after a myelodysplastic disorder.
 Therapy related AMLs
 Lastly, a fourth ‘wastebasket’ includes AMLs lacking any of these
features.
 They are classified based on degree of differentiation and
lineage of the leukemic blasts.

 Most commonly present as painless lymphadenopathy. Pathogenesis


 Nodular sclerosis and lymphocyte predominance type have stage  Recurrent genetic aberration disrupts genes encoding
I-II disease and are usually free from systemic manifestations. transcription factors that are acquired for normal myeloid
 Patients with disseminated disease (Type III, IV) or mixed- differentiation.
cellularity or lymphocyte depletion type have constitutional  Two most common:
symptoms such as fever, weight loss, and night sweats.  T(8;21) disrupts RUNX1
 Anergy resulting from depressed cell-mediated immunity is seen.  Inv(16) disrupts CBFB
 Cure rate with Stages I and IIA have 90%.  Both create chimeric genes encoding fusion proteins that
interfere with the functions and block the maturation of myeloid
Myeloid Neoplasms cells.
 Common feature of this heterogeneous group of neoplasm is an
origin from hematopoietic progenitor cells. Morphology
 Primarily involve the marrow and to a lesser degree the  Diagnosis of AML is based on the presence of at least 20% of
secondary hematopietic organs (spleen, liver, lymph nodes). myeloid blasts in the bone marrow.
 Three broad categories:  Several blasts are recognized.
1.) Acute Myeloid Leukemia – accumulation of immature blast  They have delicate nuclear chromatin, 2-4 nucleoli, and more
forms in the bone marrow suppresses normal hematopoiesis. voluminous cytoplasm than lymphoblasts.
2.) Myelodysplastic Syndromes – defective maturation, gives  Cytoplasm often contains fine, peroxidase positive azurophilic
rise to ineffective hematopoiesis, leading to cytopenias. granules.
3.) Myeloproliferative disorder – increased production of one or  Auer Rods are distinctive needle-like azurophilic granules which
more cell types. are present in most cases.
 Monoblasts have folded or lobulated nuclei, lack auer rods, and
Pathogenesis are non-specific esterase positive.
 Normal hematopoiesis is tuned by homeostatic feedback  Some blasts show megakaryocytic differentiation, accompanied
mechanisms involving cytokines and growth factors. by marrow fibrosis caused by release of fibrogenic cytokines.
 These are deranged in marrow involvement which escape from  Occasionally, blasts are entirely absent from the blood,
normal homeostatic control and suppress the function of the aleukemic leukemia. Bone marrow examination is essential.
residual normal stem cells.
 Influenced by: Immunophenotype
 Position of the transformed cell in the hierarchy  Diagnosis of AML is confirmed by performing stains for myeloid-
 Effect of transforming events on differentiation. specific antigens.

Page 15 of 21
Clinical Features  Neutrophils contain decreased number of secondary granules,
 Present within few weeks or months at the onset of symptoms toxic granulates, and Dohle bodies.
related to anemia, neutropenia, and thrombocytopenia.  Pseudo-Pelger-Huet cells are neutrophils with 2 lobes,
 Most notably fatigue, fever, and spontaneous mucosal and commonly observed.
cutaneous bleeding.  Pawn Ball Megakaryocyte contains single nuclear lobes with
 Infections are frequent, particularly the oral cavity, skin, lungs, multiple separate nuclei are seen.
kidney, bladder, and colon caused by opportunistic pathogens.  Blast cells may be increased but make up less than 20% of
 Tumors with monocytic differentiation infiltrate the skin overall marrow cellularity.
(Leukemia cutis) and the gingiva.
 AML occasionally presents as a localized soft-tissue mass known Clinical Features
variously as myeloblastoma, granulocytic sarcoma, or chloroma.  Primary MDS is predominantly a disease of adults, usually
discovered incidentally; If symptomatic, presents as weakness,
Prognosis infections, and hemorrhage due to pancytopenia.
 Generally a difficult disease to treat.  Primary MDS is divided into 8 categories.
 60% active complete remission with chemotherapy, but only 15-  Treatment is fairly limited.
30% remain free of the disease for 5 years.  In younger individuals, allogeneic hematopoietic stem cell
 Treatment with all-trans retinoic acid and arsenic salts in AML transplant offers hope for reconstitution of normal
with t(15;17) have best prognosis, 80% cure rate. hematopoiesis.
 AML with t(8;12) or inv(6) have good prognosis with  In older patients, MDS is treated with support antibiotic and
conventional chemotherapy. blood product transfusions.
 Thalidomide-like drugs, and DNA methylation inhibitors
Myelodysplastic Syndromes (MDS) improve effectiveness of hematopoiesis.
 MDS refers to a group of clonal stem cell disorders characterized
by maturation defects that are associated with ineffective Myeloproliferative Disorders
hematopoiesis and a high risk of transformation to AML.  Common pathogenic feature is the presence of mutated,
 Bone marrow is partly or wholly replaced by the clonal progeny constitutively activated tyrosine kinases or other acquired
of neoplastic multipotent cells that retains the capacity to aberrations that lead to growth factor independence.
differentiate but does so in an ineffective and disordered fashion  Most common consequence is an increase in the production of
 These abnormal cells stay within the marrow, thus patients one or more mature blood elements.
experience cytopenias.  Most of this disorder arises from multipotent myeloid
 MDS can be Primary (idiopathic) or Secondary (exposure to progenitors.
genotoxic drugs or radiation therapy).  Common features:
 Increased proliferative drive.
Pathogenesis  Homing of neoplastic stem cells to secondary hematopoietic
 Poorly understood. organs (extramedullary hematopoiesis).
 There are number of recurrently mutated genes and can be  Marrow fibrosis and peripheral cytopenias
lumped into three major functional categories:  Variable transformation to acute leukemia
 Epigenetic Factors
 RNA Splicing Factors 1.) Chronic Myelogenous Leukemia (CML)
 Transcription Factors  Distinguished from others by presence of chimeric BCR-ABL gene
 10% of MDS have loss of function mutations in TP53 (tumor derived from portions of BCR from chromosome 22, and BL on
suppressor genes). chromosome 9.
 Philadelphia Chromosome.
Morphology  Cell of origin is a pluripotent hematopoietic stem cell.
 Marrow is usually hypercellular, it is sometimes normocellular or
rarely hypocellular. Pathogenesis
 Most characteristic finding is dysplastic differentiation affecting  BCR-ABL contains dimerization domain that self-associates,
erythroid, granulocytic, monocytic, and megakaryocytic lineage. leading to activation of ABL tyrosine kinase moiety.
 Within erythroid series common abnormalities include ring  ABL kinase in turn phosphorylates proteins that induce signaling
sideroblasts, erythroblasts with iron-laden mitochondria. through the same pro-growth and pro-survival pathways turned
 Megaloblastoid Maturation resembling that seen in Vit. B12 and on by normal hematopoietic pathways.
Vit. B9 deficiency.  BCR-ABL preferentially drives proliferation of granulocytic and
 Nuclear Budding Abnormalities, recognazied as nuclei with megakaryocytic progenitors, causing abnormal release of
misshapen often polyploid outlines. immature granulocytic forms to the blood.

Page 16 of 21
Morphology
 Marrow is hypercellular, but some residual fat is present.
 Increase in red cell progenitors is subtle and usually
accompanied by an increase in granulocytic precursors and
megakaryocytes as well.
 Moderate to marked increase in reticulin fibers is seen.
 Mild organomegaly is common caused by congestion.
 Peripheral blood contains increased number of basophils and
abnormally large platelets.
 Spent phase – late in the course, PCV is characterize by
extensive marrow fibrosis that displaces hematopoietic cells.

Clinical Features
 PCV is uncommon and appears insidiously.
 Most symptoms are related to increased red cell mass and
hematocrit.
 Usually there is also an increased total blood volume
Morphology  Patients are plethoric and cyanotic due to stagnation and
 Marrow is markedly hypercellular. deoxygenation of blood in the peripheral vessels.
 Elevated proportion of eosniophils and basophils.  Intense pruritus and peptic ulceration may occur due to release
 Megakaryocytes are also increased, usually include small, of histamine from basophils.
dysplastic forms.  Abnormal blood flow and platelet function may lead to an
 Erythroid progenitors are also present in mildly decreased increased risk of bleeding and thrombotic episode.
numbers.  Hgb concentration ranges from 14-18, and hematocrit is usually
 Presence of scattered macrophages with abundant wrinkled, 60% or more.
green-blue cytoplasm so-called sea-blue histiocytes  Chronic bleeding leads to iron deficiency, which can suppress
3 erythropoiesis.
 Blood reveals leukocytosis often exceeding 100,000 cells/mm .
 It consists primarily of neutrophils, band forms, metamyelocytes,  White cell count ranges from 12,000 – 50,000 cells, and platelet
myelocytes, eosinophils, and basophils. count is often greater than 500,000 platelets and exhibit
abnormal morphology such as giant forms.
Clinical Features
th th 3.) Essential Thrombocytosis
 Primarily a disease of adults, peak incidence at 5 to 6 decade.
 Onset is insidious.  Associated with activation point mutations in JAK2 or MPL (a
tyrosine kinase normally activated by thrombopoietin).
 Mild to moderate anemia and hypermetabolism which can lead
to fatigability, weakness, weight loss, and anorexia.  ET manifests with elevated platelet and is separated from PCV by
 Splenomegaly can cause acute left upper quadrant pain due to the absence of polycythemia, and primarily myelofibrosis by the
splenic infarction. absence of marrow fibrosis.
 Constitutive JAK2 and MPL renders progenitors thrombopoietin-
2.) Polycythemia Vera independent and leads to hyperproliferation.
 Strongly associated with activating point mutations in the  Bone marrow cellularity is usually only mildly increased but
tyrosine kinase JAK2. megakaryocytes are markedly increased with abnormal forms.
 PCV is characterized by increased marrow production of red  Erythromelalgia, a throbbing and burning sensation of the hands
cells, granulocytes, platelet, but its increased RBC production is and feet caused by occlusion of small arterioles by platelet
the one responsible for the most clinical symptoms. aggregates is seen.

Pathogenesis 4.) Primary Myelofibrosis


 Development of obliterative marrow fibrosis
 JAK2 participates in JAK/STAT pathway, which lies downstram of
multiple hematopoietic growth factor receptors, including the  Replacement of marrow tissue by fibrous tissue reduces bone
erythropoietin receptor. marrow hematopoiesis, leading to cytopenias and extensive
extramedullary hematopoiesis.
 PCV cells have decreased requirements for erythropoietin and
other factors due to constitutive JAK2 signaling.  The appearance is identical to the spent phase that occurs in
other myeloproliferative disorders.
 97% is associated with valine to phenylalanine substitution in
JAK2 at residue 617.  Chief pathologic feature is the extensive deposition of collagen
 The proliferative drive is less compared to CML. in the marrow by the non-neoplastic fibroblasts.

Page 17 of 21
 The fibrosis displaces hematopoietic elements from the marrow. SPLEEN
 Two fibrinogenic factors have been implicated:  Filter for the blood and a site for immune responses to blood-
 PDGF borne antigens
 TGF-β  Functions of the spleen that impact disease states:
 Phagocytosis of blood cells and particulate matter – splenic
Morphology macrophages are responsible for “pitting red cells”, the
 Marrow is often hypercellular. process where Heinz bodies and Howell-Jolly bodies are
 Erythroid and Granulocytic appear normal but Megakaryocytes excised and for the removal of particles such as bacteria in the
are large, dysplastic, and abnormally clustered. blood
 Bone marrow becomes more hypocellular and diffusely fibrotic.  Antibody production – important site for production of
 Cloud-like clusters of atypical megakaryocytes are usually seen. antibodies against microbial polysaccharides and
 Osteosclerosis or the change of the marrow in to bone may be autoantiboides
seen very late in the course.  Hematopoiesis – for compensatory extramedullary
 Extensive extramedullary hematopoiesis in the spleen. hematopoiesis
 In the blood, premature relase of nucleated erythroid and early  Sequestration of formed blood elements – can induce
granulocyte progenitors (leukoeryhtrobalstosis) is seen. thrombocytopenia and leukopenia
 Dacrocytes (Teardrop-shaped cells) were probably damaged  Splenic insufficiency due to splenectomy or autoinfarction has
during the birthing process of the fibrotic marrow are also seen. increased susceptibility of sepsis induced by encapsulated
bacteria such as pneumococci, meningococci, Haemophilus
Clinical Features influenza
 Less common, usually occurs in 60 years older patients.
 It comes to attention due to progressive anemia and Splenomegaly
splenomegaly.  When sufficiently enlarged, can cause a dragging sensation in the
 Nonspecific symptoms are also present. LUQ and through pressure on the stomach, discomfort after
 Labs show a moderate to severe normochromic, normocytic eating
anemia accompanied by leukoerythroblastosis.  Hypersplenism – characterized by anemia, leukopenia,
 WBC is usually normal or reduced; platelet is usually normal or thrombocytopenia alone or in combination
elevated, blood findings are usually non-specific.
 Harder to treat than PCV or ET. Disorders associated with splenomegaly
Infections Nonspecific splenitis of various blood-
Langerhans Cell Hisitocytosis borne infections, Infectious
mononucleosis, Tuberculosis, Typhoid
 A designation for a variety of proliferative disorders of dendritic
fever, Brucellosis, Cytomegalovirus,
cells or macrophages.
Syphilis, Malaria, Histoplasmosis,
 Some are malignant (e.g. Lymphomas), and some are benign Toxoplasmosis, Kala-azar,
(e.g. Reactive Proliferations). Trypanosomiasis, Schistosomiasis,
 The origin and nature has been controversial leading to a Leishmaniasis, Echinococcosis
discussion whether it is better considered as a neoplasm or a Congestive States Cirrhosis of the liver
reactive process. Related to Portal Portal or splenic vein thrombosis
Hypertension Cardiac failure
 The Langerhans cells have abundant, often vacuolated cytoplasm
and vesicular nuclei containing linear grooves or folds. Lymphohematogenous Hodgkin lymphoma
Disorders Non-Hodgkin lymphomas and
 Presence of Birbeck granules is characteristic, these are lymphocytic leukemias
pentalaminar tubules, often with dilated terminal end, producing Multiple myeloma
a tennis-racket like appearance. Myeloproliferative disorders
 Can present as several clinicopathologic entities: Hemolytic anemias
 Multifocal Multisystem LCH (Letterer-Siwe Disease) Immunologic- Rheumatoid arthritis
 Unifocal and Multifocal Unisystem LCH (Eosinophilic Inflammatory Systemic lupus erythematosus
granuloma) Conditions
 Pulmonary LCH Storage Diseases Gaucher disease
Niemann-Pick disease
 One factor contributing to neoplastic Langerhans cells is the Mucopolysaccharidoses
aberrant expression of chemokine receptors.
Miscellaneous Disorders Amyloidosis
Primary neoplasms and cysts
Secondary neoplasms

Page 18 of 21
A. Nonspecific Acute Splenitis Neoplasms
 Cause by microbiologic agents and cytokines released as part of  Myeloid and lymphoid tumors often cause splenomegaly
immune response  Benign fibromas, osteomas, chondromas, lymphangiomas and
hemangiomas may occur in the spleen
Morphology
 Enlarged up to 200-400gm and soft Congenital Anomalies
 Major feature is acute congestion of the red pulp, may encroach  Complete absence is rare and may be related to situs inversus and
and efface lymphoid follicles cardiac malformation
 Neutrophils, plasma cells and eosinophils are present in white and  Hypoplasia is a more common finding
red pulp  Accessory spleen (spleniculi) – common, small, spherical
 White follicles may undergo necrosis when causative agent is structures found within the abdominal cavity
haemolytic streptococcus
 Abscess may occur Rupture
 Precipitated by blunt trauma
B. Congestive Splenomegaly  Predisposing conditions include infectious mononucleosis,
 Caused by chronic venous outflow obstruction due to intra or malaria, typhoid fever and lymphoid neoplasm
extra hepatic disorder that impinge the portal or splenic veins  Cause intraperitoneal hemorrhage and treated by prompt
 Systemic, or central venous congestion is encountered in cardiac splenectomy
decompensation involving the the right side of the heart
 Cirrhosis of the liver is the main cause of congestive splenomegaly THYMUS
 Spontaneous portal vein thrombosis or pylephlebitis can also
cause congestive Splenomegaly Developmental Disorders
 Thymic hypoplasia or aplasia – seen in DiGeorge syndrome,
Morphology marked by severe defects in cell mediated immunity and
 Enlargement up to 1000-5000gm, firm and capsule is thickened abnormalities of parathyroid development
and fibrous
 Thymic cysts – uncommon lesions, spherical or arborizing, lined by
 Red pulp is congested and becomes fibrotic and cellular stratified to columnar epithelium and not clinically significant
 Deposition of collagen in basement membrane
 Excessive destruction due to prolonged exposure to macrophages Thymic Hyperplasia
 Most frequently encountered in myasthenia gravis
 Also seen in Grave’s disease, SLE, scleroderma, rheumatoid
arthritis and other autoimmune disorders

Thymomas
 Restricted to tumors of thymic epithelial cells
 Contain benign immature T cells (thymocytes)
 Three histological subtypes:
 Benign and noninvasive
 Benign and invasive
 Malignant (thymic carcinoma)
 Usually occurs in >40 years old
 Males and females are equally affected
C. Splenic Infarcts  Most arise in the the anterior superior mediastinum, but may
 Common lesions caused by occlusion of the major splenic artery arise in the neck, thyroid, pulmonary hilus or elsewhere, typically
or its branches uncommon in posterior mediastinum
 One of the most frequent site for emboli lodge
 Often caused by emboli arising from the heart

Morphology
 Bland infarcts are pale, wedge-shaped and subcapsular in location
 Overlying capsule is covered with fibrin
 Development of suppurative necrosis
 Large depressed scars develop in healing

Page 19 of 21
Morphology
 Lobulated, firm, gray-white masses up to 15-20cm in size
 Area of cystic necrosis and calcification
 Noninvasive thymomas
 Composed of medullary-type epithelial cells or a mixtyre of
medullary and cortical cells
 Sparse infiltrates of thymocytes
 Invasive thymomas
 Much more like to metastasise
 Epithelial cells are most commonly of the cortical variety with
abundant cytoplasm and rounded vesicular nuclei mixed with
numerous thymocytes
 Penetrate through the capsule into surrounding structures
 Thymic carcinoma
 Most are squamous cell carcinoma, next variant is
lymphoepithelioma-like carcinoma
 Composed of sheets of cells with indistinct borders that bears
a close histologic resemblance to nasopharyngeal carcinoma

Clinical Features
 Present with symptoms stemming from impingement on
mediastinal structures
 Detected in the course of evaluating patients with myasthenia
gravis
 Other immune disorders include hypogammaglobinemia, pure red
cell aplasia, Graves’ disease, pernicious anemia, dermatomyositis-
polymyositis and Cushing syndrome.

“Ñuhor līr
gūrēnna.”
(I will take what’s
mine)

Page 20 of 21
Page 21 of 21

You might also like