The pathologist's H&E is like the clinician's H&P (history and physical)—basic examinations that are performed on every specimen or patient to form the cornerstone of diagnosis. However, the pathologist is no longer limited to H&E: a wide variety of special studies are available to evaluate pathologic processes, from simple histochemical stains to global gene expression patterns. Pathologists are now clinical cell biologists. Familiarity with the types of special studies available is important as the initial processing of the gross specimen may limit the types of studies that can be performed.
HISTOCHEMISTRY
Almost all histochemical stains are suitable for use on formalin-fixed tissues. Common stains and their uses are listed in Table 7-1 . However, numerous other types of stains and modifications are used and pathologists must be aware of individual laboratory practices.
Table 7-1.
Histochemical stains
| STAIN | COMPONENTS STAINED | POSSIBLE USES AND COMMENTS |
|---|---|---|
| AFOG (acid fuchsin orange G; modified Masson's trichrome) |
|
Evaluation of renal biopsies |
| Alcian blue |
|
Sometimes used to identify mucosubstances in mesotheliomas or intestinal metaplasia; affected by pH; hyaluronidase digestion can be used to identify hyaluronic acid |
| Alcian blue–PAS |
|
Demonstrates both acid and neutral mucins |
| Alcian yellow |
|
Identification of H. pylori in gastric biopsies |
| Acid-fast bacilli stains (Fite–Faraco, Ziehl–Neelsen, Kinyoun) |
|
Identification of mycobacteria; modifications are used to demonstrate M. leprae or Nocardia; tissues fixed in Carnoy's cannot be used, and B5 is suboptimal; slides must be examined under oil |
| Alizarin red S | Calcium:orange red, polarizes | Identifies calcium in tissues |
| Bile | Bile:dark green on a yellow background | Identification of bile |
| Bodian's |
|
Neural tumors, identification of axons |
| Chloroacetate esterase (CAE; Leder) |
|
Evaluation of leukemias; identification of mast cells; cannot be used for tissue fixed in Zenker's or B5 |
| Congo red |
|
Detection of amyloid; immunoperoxidase studies can be used to identify specific types; overstaining can result in false positives |
| Dieterle |
|
Infectious lesions; melanin, chromatin, formalin pigment, and foreign material may also stain |
| Diff-Quik® (a modified Giemsa stain) |
|
Evaluation of chronic gastritis |
| Elastic stains (Verhoeff–van Gieson) (= ET) |
|
Identification of arteries and veins, vasculitis, invasion of lung tumors into visceral pleura, abnormal elastic fibers in elastofibromas |
| Fibrin (see phosphotungistic acid–hematoxylin [Mallory's PTAH]) | To demonstrate fibrin in renal biopsies | |
| Fontana–Masson | Melanin, argentaffin granules, chromaffin granules, some lipofuscin:black Nuclei:red | Identification of melanin in melanomas and secretory granules in neuroendocrine tumors; use of this stain has largely been replaced by immunohistochemistry |
| Giemsa (May–Grünwald) |
|
Lymphoproliferative disorders (good nuclear and cytoplasmic detail);identification of bacteria, rickettsias, and Toxoplasma gondii |
| Gram (Brown–Hopps, Brown–Brenn) |
|
Identification of bacteria, some cases of actinomycetes, Nocardia, coccidioidomycosis, blastomycosis, cryptococcosis, aspergillosis, rhinosporidiosis, and amebiasis |
| Grimelius |
|
Evaluation of neuroendocrine tumors (largely replaced by the use of immunohistochemistry for chromogranin) |
| Hematoxylin and eosin (H&E) |
|
Standard stain for the routine evaluation of tissues |
| Iron (colloidal iron) |
|
Bone marrow (iron stores, myelodysplasias), liver (hemochromatosis);chromophobe renal cell carcinomas are positive |
| Melanin bleach | Removes melanin from tissue, usually for IHC; melanin can be difficult to distinguish from IHC positivity | |
| Methyl green–pyronin Y |
|
Plasma cell lesions (largely replaced by immunohistochemistry);does not work well on tissues decalcified with formic acid |
| Mucicarmine (Mayer) |
|
Identification of adenocarcinomas, identification of cryptococcus |
| Oil red O |
|
Requires frozen sections (lipids are dissolved by most fixatives or during processing);tissue fixed in formalin can be used if tissue is frozen |
| Periodic acid–Schiff (PAS) |
|
Classification of tumors with glycogen (e.g., Ewing's/PNET, rhabdomyosarcoma, renal cell carcinoma), glomerular diseases (BM), identification of adenocarcinomas (mucin), fungal diseases (especially in argentophilic areas: neutrophils and debris), spironolactone bodies in adrenal adenomas treated with this drug |
| Periodic acid–Schiff with diastase digestion (PAS-D) | As above, except glycogen has been digested and will not be stained | Identification of glycogen in tumors; identification of fungus in glycogen-rich tissue (e.g., skin);PAS-D resistant deposits in liver are present in α1-antitrypsin deficiency |
| Phosphotungistic acid–hematoxylin (Mallory's PTAH) |
|
Identification of neural lesions; skeletal muscle differentiation (Zenker's fixative is preferred); this stain has been replaced by IHC for muscle markers |
| Reticular fibers (Gomori, Gordon and Sweets, Snook) (RETIC) |
|
Bone marrow (myelophthisis), liver (fibrosis, veno-occlusive disease), carcinoma versus sarcoma (reticular network);largely replaced by IHC |
| Silver stain (Grocott methenamine–silver nitrate—GMS) (GMS or MMS) |
|
Evaluation of infectious diseases;bacteria will also stain black |
| Steiner |
|
Evaluation of infectious diseases |
| Toluidine blue |
|
Mast cell diseases, chronic cystitis |
| Trichrome (Gomori's trichrome, Masson) (= TRI) |
|
Mature collagen, type 1:dark blue |
| Von Kossa calcium |
|
Demonstration of phosphate and carbonate radicals with calcium in tissues, identification of malakoplakia (Michaelis–Gutmann bodies) |
| Warthin–Starry |
|
Infectious lesions |
| Wright's |
|
Blood smears |
The WebPath section of the University of Utah site (http://medlib.med.utah.edu) has useful descriptions of special stains and illustrative photographs.
IMMUNOPEROXIDASE STUDIES
The development of methods to detect antigens on tissue sections with antibodies was a major advance in surgical pathology. Immunohistochemical (IHC) studies are most frequently used for the following purposes:
-
•
Classification of tumors (e.g., carcinoma versus lymphoma, B-cell versus T-cell lymphoma)
-
•
Identification of in situ lesions versus invasion (e.g., myoepithelial markers in breast cancers, basal cell markers in prostate)
-
•
Prognostic factors (e.g., Ki-67 in glioblastomas)
-
•
Predictive factors to guide specific therapy (e.g., c-kit, estrogen and progesterone receptors, HER2/neu)
-
•
Identification of extracellular material (e.g., β2-microglobulin amyloid)
-
•
Identification of infectious agents (e.g., cytomegalovirus).
Use of immunohistochemistry
A differential diagnosis is generated after examination of the H&E stained slides. Immunohistochemistry is then used to gain evidence for or against diagnostic possibilities. “Trolling” cases through an immunohistochemistry laboratory by ordering numerous antibody studies without a clear reason in mind is more likely to lead to misguided diagnosis due to aberrant immunoreactivity than to provide an unexpected correct diagnosis.
A very useful website has been developed by Dr. Dennis M. Frisman (http://www.immunoquery.com): it tabulates published literature on the immunoreactivity profiles for numerous tumors. There is also a comprehensive list of the included references with web links.
Panels
There are no absolute rules for immunoreactivity in cells and tissues. Aberrant positive immunoreactivity (or absence of immunoreactivity) is occasionally observed for all antibodies, either due to biologic variability (e.g., occasional keratin-positive melanomas) or technical factors (impure antibodies, cross-reaction with other antigens, failure to preserve antigenicity). Thus, immunohistochemical markers are used most effectively as panels of markers, with interpretation based on an immunohistochemical profile.
Slides for immunohistochemistry
Tissue is often dislodged from normal glass slides during the treatments required for IHC. Slides must be coated (e.g., with glue, poly-l-lysine, gelatin, albumin) or special commercial slides must be used. If slides are being prepared by another laboratory, the type of glass slide to be used must be specified.
Factors affecting immunogenicity
Numerous variables can affect antigenicity. The most common are described below. Each laboratory must optimize its procedures for each antibody used. Studies on tissues or slides not prepared in the routine fashion for a laboratory must be interpreted with caution.
Type of fixative.
Some fixatives destroy some antigens (e.g., Bouin's diminishes ER immunoreactivity, keratins are not well preserved in B5).1 Most studies are based on formalin-fixed tissue. Results cannot be assumed to be equivalent for other fixatives.
Length of time of fixation in formalin.
Protein cross-linking and antigenicity generally decrease with fixation times over 24 hours. To some extent, this effect can be reversed using antigen-retrieval methods.
Prior decalcification in hydrochloric acid.
This decreases the antigenicity of some epitopes (predominantly nuclear) but not others (predominantly cytoplasmic).2 Decalcifying agents using EDTA do not alter immunogenicity.
Decreased: ER, PR, Ki-67, p53, Ber-EP4 (tumor cells).
Not affected: calcitonin, chromogranin, GCDFP-15, HMB-45, thyroglobulin, S100, PSA, keratins (CK 20, CAM 5.2, AE1/AE3), others.
Length of time since the glass slide was cut.3, 4, 5, 6
The immunoreactivity of the majority of antigens declines over days to weeks and may be lost completely at 1 month.3, 4, 5, 6 The loss may be due to oxidation of amino acids with exposure of tissue to air, as the immunogenicity of tissue deeper in the block can be preserved for many years. Antigen-retrieval methods do not completely restore the antigenicity of old slides. Coating slides with paraffin, storing the slides in a nitrogen desiccator, and/or storing at lower temperatures can partially preserve antigenicity. However, studies should be performed on newly cut slides, if possible.
Antigen-retrieval procedures.
These include proteolysis, heating (microwave, steam), and special incubation fluids. To some extent these methods reverse the effects of formalin fixation. Variable effects are observed for different antibodies.
Type of antibody (polyclonal versus monoclonal versus mixture of different monoclonals), epitope detected.
Very different results can be obtained with different antibodies to the same protein or different commercial sources of the same antibody.
Incubation time, incubation temperature, dilution of antibody.
Methods of signal amplification.
Temperature of baking the slide.
Controls
Controls are essential for the appropriate interpretation of immunohistochemical studies and to ensure that all steps of this complicated procedure have been adequately performed.
Positive controls.
Tissues known to be immunoreactive should be included each time an antibody is used for a test case. Internal positive controls should always be evaluated when present as they control not only for the technique used but also for the antigenicity of the tissue under investigation. Table 7-30 (see pp. 103) lists normal cells that are generally immunoreactive for each antibody. Some laboratories have used vimentin as a control for immunogenicity as almost all tissue should demonstrate positivity.7 Given the wide and nonspecific distribution of vimentin, smooth muscle α-actin may be more useful in this context as pericytes, vascular smooth muscle, and myoepithelial cells present in most tissues are immunoreactive.
Table 7-30.
Antibodies for immunohistochemistry
| General markers | |||||
|---|---|---|---|---|---|
| NAME (ALTERNATE NAME) | ANTIGEN (LOCATION) | NORMAL CELLS AND TISSUES | TUMORS | USES | COMMENTS |
| Alpha fetoprotein*(AFP, arfetoprotein) | Glycoprotein present in fetal liver (cytoplasm, granular) | Fetal liver, regenerating liver cells | HCC (but not the fibrolamellar variant), hepatoblastomas, yolk sac tumors, embryonal carcinoma (but less commonly) | HCC (+/−) versus other cell types (however, AFP is rarely present in other carcinomas such as breast and ovary) Yolk sac tumors (+) versus other germ cell tumors (−/+). | Correlates with extracellular hyaline eosinophilic globules in yolk sac tumors |
| Alpha-1-antitrypsin(AAT, α,-AT) | Glycoprotein inhibiting proteolytic enzymes produced in the liver (cytoplasm) | Histiocytes, reticulum cells, mast cells, Paneth cells, salivary gland | HCC, germ cell tumors, true histiocytic neoplasms, colon and lung carcinoma, others | Accumulates in liver cells in AAT deficiency | Not specific for tumor type. CD68 is somewhat more specific for macrophages |
| Alpha smooth muscle actin*(SMA, SM-ACT) | Smooth muscle isoform of actin (cytoplasm) | Smooth muscle, myoepithelial cells, blood vessel walls, pericytes, some stromal cells of intestine, testis, and ovary, myofibroblasts in desmoplastic stroma Not in striated muscle or myocardium | Smooth muscle tumors, myofibroblastic tumors, PEComas, glomus tumors, KS, some spindle cell carcinomas (e.g., with features of myoepithelial cells) | Identification of smooth muscle differentiation (muscle or myofibroblasts) in tumors Sclerosing lesions (myoepithelial cells present) versus invasive carcinoma, in the breast | Good marker for myoepithelial cells of the breast but also positive in myofibroblasts in stroma. p63 is only positive in myoepithelial cells |
| AMACR*(P504S, alpha-methylacyl- CoA racemase) | Mitochondrial and peroxisomal enzyme involved in the metabolism of branched-chain fatty acid and bile acid intermediates (cytoplasm) | Not present in normal tissues | Colorectal carcinoma (92%), colonic adenomas (75%), prostate carcinoma (83%), PIN (64%), breast cancer (44%), ovarian carcinoma, TCC, lung carcinoma, RCC, lymphoma, melanoma | Can be combined with p63 to distinguish prostate carcinoma (AMACR +, p63 absent in basal cells) from benign mimics (AMACR -, p63 present in basal cells) | |
| Androgen receptor(AR) | Mediates the function of androgens (nucleus) | Prostate, skin, oral mucosa | Osteosarcoma, prostatic carcinoma, breast carcinoma, ovarian carcinomas, others | ||
| B72.3(Tumor-associated glycoprotein 72, TAG-72, CA 72-4) | Oncofetal glycoprotein, may be a precursor of the MN blood group system, sialosyl-Tn antigen (cytoplasm, membrane) | Not present in most benign adult epithelial cells (may be present in secretory endometrium), apocrine metaplasia, and fetal GI tract | Adenocarcinomas (esp. ovary, colon, breast) | Adenocarcinoma (+ >90%) versus mesothelioma (5%) or mesothelial cells (−) | Other markers are more useful for this purpose |
| bcl-2* | Protein involved in inhibition of apoptosis (membrane, cytoplasm) | Medullary lymphocytes and epithelial cells of the normal thymus, mantle and T zone small lymphocytes | Synovial sarcoma, solitary fibrous tumor, myofibroblastic tumors, schwannoma, neurofibroma, granular cell tumor, GIST, KS, melanoma Small lymphocytic lymphoma/CLL, mantle cell lymphoma, follicular lymphoma, marginal zone lymphoma (MALT), some large B-cell lymphoma |
Synovial sarcoma (+/−) versus mesothelioma (−) Thymic carcinomas strongly express bcl-2 compared to thymomas Small lymphocytic lymphoma, mantle cell lymphoma, and marginal zone lymphoma (MALT) (+) vs reactive follicles (−) |
The bcl-2 gene is involved in the t(14;18) found in follicular lymphomas |
| Ber-EP4(Epithelial specific antigen (ESA), Ep-CAM) | Glycoprotein (membrane) | All epithelial cells except superficial layers of epidermis | Most carcinomas | Adenocarcinoma (+; strong and diffuse in 60-100%) versus mesothelioma (− or focal in 26%) | Other markers are better for distinguishing adenocarcinoma from mesothelioma |
| Beta-amyloid(6F/3D) | Amyloid present in Alzheimer's disease (AD) and in cerebral amyloid angiopathy (extracellular) | None | Senile plaque core in AD, amyloid cores, neuritic plaques, neurofibrillary tangles | Diagnosis of AD, other diseases | Found in AD, Lewy body dementia, Down's syndrome, hereditary cerebral amyloidosis (Dutch type) |
| Beta-eatenin | Component of the adherens junction that binds to e-cadherin and functions in cell adhesion and anchoring the cytoskeleton; signaling molecule of the Wnt/wingless pathway (membrane, cytoplasm) | Urothelium, breast epithelium, colon, esophagus, stomach, thyroid | TCC, colonic adenocarcinomas and adenomas, breast carcinoma, esophageal squamous cell carcinoma, head and neck squamous cell carcinomas, gastric carcinoma, ovarian carcinoma, thyroid carcinoma, prostate carcinoma, HCC, brain neoplasms | Aberrant nuclear expression in solid-pseudopapillary tumors of the pancreas (95%) and pancreatoblastomas (78%) | |
| Beta-2 micro-globulin | Immunoglobulin associated protein (extracellular deposits of amyloid) | Plasma cells | Identification of amyloid in patients on dialysis | Amyloid tends to accumulate around joints and in the GI tract | |
| BG8 | Lewis blood group y antigen (cytoplasm) | Red blood cells, endothelial cells | Adenocarcinomas (95%), rare mesotheliomas (about 5%) | Other markers are better for distinguishing adenocarcinoma from mesothelioma | |
| Blood group antigens | A, B, and H antigens (membrane) | Epithelial cells and red blood cells, endothelial cells | Lost or abnormally expressed in many carcinomas | Can be helpful to identify potentially misidentified specimens | |
| CAI25*(OC125) | Mucin-like glycoprotein, antibody to ovarian carcinoma antigen (luminal surface) | Epithelial cells, mesothelial cells | Adenocarcinomas of ovary, breast, lung (bronchioloalveolar), and others (rarely colon), TCC, the uterus, squamous cell carcinoma, seminal vesicle carcinoma, anaplastic lymphoma | Seminal vesicle carcinoma (+) versus prostate carcinoma (−) | Used as a serum marker for monitoring ovarian cancer |
| CA19-9(Carbohydrate antigen 19-9) | Antigen of sialyl Lewisa - containing glycoprotein; antibody to colon carcinoma (cytoplasm) | Epithelial cells of breast, colon, kidney, liver, lung, pancreas, salivary gland, others | Adenocarcinomas of GI tract, pancreas, ovary, lung, and bladder, rare in mesotheliomas Chronic pancreatitis | Used as a serum marker for monitoring gastrointestinal and pancreatic carcinomas | |
| Calcitonin* | Peptide hormone produced by C cells (cytoplasm and extracellular amyloid) | C cells of the thyroid | Medullary carcinoma of the thyroid (within tumor cells and in amyloid) | ID of C-cell hyperplasia ID of medullary thyroid carcinoma |
Used as a serum marker for medullary carcinoma |
| Caldesmon*(h-caldesmon) | Actin and calmodulin binding protein in smooth muscle (cytoplasm) | Vascular and visceral smooth muscle cells, some myoepithelial cells of the breast | Smooth muscle tumors, PEComa, GIST | Smooth muscle tumors (+) vs myofibroblastic lesions (−) or endometrial stromal tumors (−) | |
| Calponin(CALP)* | Protein that binds to calmodulin, F-actin, and tropomyosin to regulate smooth muscle contraction (cytoplasm) | Vascular and visceral smooth muscle cells, myoepithelial cells of the breast, periacinar and periductal myoepithelial cells of the salivary gland | Myoepithelioma, some smooth muscle tumors, myofibroblastic lesions | May be helpful to identify myoepithelial cells in breast lesions | SMA is a better marker of myofibroblasts |
| Calretinin* | Intracellular calcium-binding protein of the troponin C superfamily with an EF-hand domain (cytoplasm, nucleus) | Subsets of neurons, pineal cells, germinal epithelium of ovary, mesothelial cells, keratinocytes, breast, sweat glands, neuro-endocrine cells, thymus | Epithelial mesotheliomas (less + in sarcomatoid type), adenomatoid tumor, some lung squamous cell carcinomas, rare adeno-carcinomas, mesenchymal tumors (e.g., synovial sarcoma), granular cell tumor, Leydig cell tumor, granulosa cell tumor | Epithelial mesotheliomas (>90%) versus adeno- carcinoma (<10%) | Useful marker in that it is positive in mesothelioma and usually negative in carcinomas |
| Carcinoembryonic antigen*(CEA, CD66e) | Glycoproteins with immuno- globulin-like regions found in fetal tissues (cytoplasm) | Fetal tissues | Adenocarcinomas (liver, colon, pancreas, bile duct, and lung more than breast, liver, ovary), TCC, medullary carcinoma of the thyroid Usually absent in RCC, prostate carcinoma, and papillary or follicular thyroid carcinomas |
Adenocarcinoma (+) versus mesothelioma (−) HCC: polyclonal CEA has a canalicular pattern |
Different reactivity patterns occur with different antibodies and with polyclonal versus monoclonal antibodies |
| CD5*(Leu 1) | Transmembrane glycoprotein (membrane) | T cells and B-cell subsets (mantle zone) | Thymic carcinoma, adeno- carcinomas, mesothelioma (cytoplasmic). T-cell leukemias and lymphomas, aberrantly expressed in low-grade B-cell lymphomas (CLL or mantle cell lymphoma) |
Thymic carcinoma (+/−) versus thymoma (−). Thymic carcinoma (+/−) versus metastatic squamous carcinoma (−) Classification of low-grade B-cell lymphomas Evaluation of T-cell lymphomas (this marker is frequently lost) |
|
| CD10*(CALLA [common acute leukemia antigen], J5) | Cell surface metallo- endopeptidase that inactivates peptides (membrane) | Precursor B cells, granulocytes, rare cells in reactive follicles, myoepithelial cells of breast, bile canaliculi, fibroblasts, brush border of kidney and gut | Endometrial stromal sarcoma, RCC (clear cell and papillary types), HCC, TCC, rhabdomyosarcoma, pancreatic carcinoma, schwannoma, melanoma Precursor lymphoblastic lymphoma/leukemia, follicular lymphoma, Burkitt's lymphoma, CML, angioimmunoblastic lymphoma |
Myoepithelial cell marker in breast Endometrial stromal sarcoma (+) versus leiomyosarcoma (−/+) (but caldesmon is preferred for this purpose) Evaluation of low-grade lymphomas Evaluation of leukemias |
Not specific for nonlymphoid neoplasms |
| CD15*(LeuM1) | 3-fucosyl-N-acetyllactosamine, X-hapten—CHO moiety linked to cell membrane protein (membrane and cytoplasm) | Granulocytes, monocytes | Adenocarcinomas CMV-infected cells RS cells (not LP HD) in a membranous and Golgi pattern, some large T-cell lymphomas, MF, some leukemias | Adenocarcinomas (+) versus mesotheliomas (−) Evaluation of HD | |
| CD30*(Ber-H2, Ki-1) | Single chain transmembrane glycoprotein homologous to the nerve growth factor super-family (cytoplasm, membrane, and Golgi) | Activated B and T cells, some plasma cells, immunoblasts, interdigitating cells, histiocytes, follicular center cells, decidualized endometrium, reactive mesothelial cells, most other tissues negative | Embryonal carcinoma, some vascular tumors (not KS), some mesotheliomas Anaplastic large cell (CD30+) lymphomas, mediastinal large B-cell lymphoma, primary effusion lymphoma, HD (but not LP HD), some other B-and T-cell lymphomas, EBV-transformed B cells |
Evaluation of anaplastic large cell (CD30+) lymphomas Evaluation of HD (RS cells are positive except in LP HD) Evaluation of peripheral T-cell lymphoma (large cells may be positive) |
|
| CD31*(PECAM-I, platelet- endothelial cell adhesion molecule) | Transmembrane glycoprotein functioning in cell adhesion (cytoplasm, membrane) | Endothelial cells, platelets, megakaryocytes, plasma cells, histiocytes, other hematopoietic cells | Vascular tumors (>80% of angiosarcomas), KS, histiocytic neoplasms, PEComa, very rarely other tumors | ID of endothelial differentiation in tumors Evaluation of angiogenesis |
Most sensitive and specific marker for endothelial cells |
| CD34*(HPCA-1, hematopoietic progenitor cell, class 1, QBEnd10) | Single chain transmembrane glycoprotein, leukocyte differentiation antigen (cytoplasm, membrane) | Hematopoietic progenitor cells (decreases with maturation), endothelial cells, fixed connective tissue cells (e.g., in skin), fibroblasts | Acute leukemia, sarcomas of vascular origin, KS, epithelioid sarcoma, GIST, DFSP, solitary fibrous tumor, neurofibroma, schwannoma, spindle cell lipoma | Identification of endothelial or fibroblastic differentiation in tumors Evaluation of angiogenesis Evaluation of the number of blasts in bone marrow in acute leukemia Solitary fibrous tumor (+) versus sarcomatoid mesothelioma (−) DFSP (+) versus dermatofibroma (−) |
Not specific but can be useful in context with other features |
| CD44v3(CD44 variant 3, H-CAM) | Transmembrane glycoprotein that mediates cell adhesion (membrane) | Many, including myometrium | Many, including endometrial carcinomas | Possibly helpful to distinguish cellular leiomyoma (+) from endometrial stromal sarcoma (−) | Many splice variants of CD44 are present in normal and malignant cells |
| CD57*(Leu 7, HNK-1) | Lymphocyte antigen that cross reacts with a myelin- associated glycoprotein (membrane) | T-cell subsets, NK cells, myelinized nerves, neuroendocrine cells, prostate, pancreatic islets, adrenal medulla | Nerve sheath tumors (occasional), leiomyosarcoma, synovial sarcoma, rhabdomyosarcoma, neuroblastoma, gliomas, neuroendocrine carcinomas, neurofibromas some prostate carcinomas Angioimmunoblastic lymphoma, T gamma lymphoproliferative disorder (large granular cell lymphocytic leukemia) |
ID of neuroendocrine differentiation in tumors ID of angioimmunoblastic T-cell lymphoma Evaluation of NK neoplasms |
Not very specific for solid tumors |
| CD63(NKI/C3, melanoma- associated antigen, ME491) | Member of the tetraspanin or transmembrane 4 superfamily (TM4SF) found on lysosomes (cytoplasm or membrane) | Melanocytes, mast cells, histiocytes, salivary gland cells, sweat gland cells, pancreatic cells, islets of Langerhans, prostatic cells, Paneth cells, peribronchial glands, pituitary | Nevi, melanomas, carcinoids, medullary carcinomas of the thyroid, some adenocarcinomas | Cellular neurothekoma (NKI/C3 + and S100 -) versus melanocytic lesions (NKI/C3 and S100 +) ID of melanocytic lesions |
May be negative in desmoplastic melanomas |
| CD68*(KP1, CD68-PGM1, Mac-M) | Intracellular glycoprotein associated with lysosomes (cytoplasm, membrane) | Macrophages, monocytes, neutrophils, basophils, large lymphocytes, Kupffer cells, mast cells, osteoclasts | Neurofibroma, schwannoma, MPNST, granular cell tumors, PEComa, melanomas, atypical fibroxanthoma, RCC Some lymphomas, histiocytic sarcomas, APML, Langerhans proliferative disorders |
Best general marker for macrophages, although not specific to this cell type | The antibody PG-M1 does not react with granulocytes Not very specific for solid tumors |
| CD99*(MIC-2, 12E7, Ewing's sarcoma marker, E2 antigen, HuLy-m6, FMC 29, O13 [different epitope]) |
MIC2 gene product— glycoproteins (p30 and p32) involved in rosette formation with erythrocytes (membrane) Membrane immunoreactivity is more specific than cytoplasmic |
Cortical thymocytes, T lymphocytes, granulosa cells of ovary, pancreatic islet cells, Sertoli cells, some endothelial cells, urothelium, ependymal cells, squamous cells | PNET/Ewing's sarcoma, chondroblastoma, mesenchymal chondrosarcoma, synovial sarcoma, solitary fibrous tumors, GIST, some alveolar rhabdomyosarcomas, desmoplastic small cell tumors, small cell carcinomas, granulosa cell tumors, yolk sac components of germ cell tumors, Sertoli-Leydig cell tumors, atypical fibroxanthoma, meningioma B- and T-cell precursor lymphoblastic lymphoma/leukemia |
Thymic carcinomas (lymphocytes +) versus other carcinomas. ID of PNET/Ewing's sarcoma (immunoreactivity should be clearly membranous in the majority of the cells) Evaluation of lymphoblastic lymphoma/leukemia |
O13 is the most commonly used antibody Immunoreactivity is highly dependent upon the antigen retrieval system used |
| CDI 17*(c-kit, stem cell factor receptor) | Transmembrane tyrosine kinase receptor (ligand is stem cell factor)— apoptosis is inhibited when the ligand is bound (cytoplasm, membrane) | Mast cells, interstitial cells of Cajal (ICC—pacemaker cells of the GI tract found throughout the muscle layers and in the myenteric plexus), epidermal melanocytes, mononuclear bone marrow cells (4%), Leydig cells, early spermatogenic cells, trophoblast, breast epithelium | GIST (>95%), seminomas (>70%), intratubular germ cell neoplasia, mature teratomas (>70%), papillary RCC (cytoplasmic— associated with mutations), chromophobe RCC (membrane —not associated with mutations), some melanomas (focal), mast cell tumors, some carcinomas, some brain tumors, some PNET/Ewing's sarcoma, some angiosarcomas AML (>50%), CML in myeloid blast crisis |
ID of GIST (+) versus leiomyomas (−) and schwannomas (−) ID of seminomas ID of mast cells (mastocytosis) |
Mast cells are an excellent internal control CD117 positivity does not correlate with mutations and/or oncoprotein activity in tumors not known to have activating mutations and is, in general, not of clinical or therapeutic significance in this setting (e.g., to detect tumors likely to respond to therapy directed against the protein, e.g. Gleevec). |
| CDI4I(Thrombomodulin, TM) | Transmembrane glycoprotein, receptor for thrombin (cytoplasm [epithelial cells], membrane [mesothelial cells]) | Endothelium, platelets, monocytes, synovial cells, syncytiotrophoblast, mesothelial cells, dermal keratinocytes, islet cells, peripheral nerves | Mesotheliomas, TCC, KS, squamous cell carcinomas, choriocarcinomas, rarely adenocarcinomas, benign and malignant vascular tumors | Mesothelioma (+ 80%) versus adenocarcinoma (+ 10%) (but variable results have been reported in other studies) | Other markers are better for distinguishing adenocarcinoma from mesothelioma |
| CD146*(melanoma cell adherin molecule, MELCAM, MCAM, MN-4, MUCI8, A32 antigen, S-Endo-I) | Membrane cell adhesion glycoprotein of the Ig gene super-family (membrane) | Implantation site intermediate trophoblast, myofibroblasts, endothelium, pericytes, Schwann cells, ganglion cells, smooth muscle, cerebellar cortex, breast luminal and myoepithelial cells, external root sheath of hair follicle, subcapsular epithelium of thymus, follicular dendritic cells, basal cells of bronchus and parathyroid, subpopulations of activated T cells | Melanoma, angiosarcoma, KS, leiomyosarcoma, placental site trophoblastic tumor, choriocarcinoma May be focally positive in squamous cell carcinoma and small cell carcinoma of the lung, mucoepidermoid carcinoma, breast carcinoma, some leukemias, neuroblastoma |
ID of placental site trophoblastic tumors | |
| CDX2* (CDX-88) | Homeobox nuclear transcription factor specific for the intestinal tract that regulates MUCI expression (nucleus) | Small intestine, colon, and endocrine pancreas | Colon carcinomas (usually strong and diffuse), small intestine carcinomas, mucinous ovarian carcinomas, bladder adenocarcinomas, some gastric, esophageal, pancreatic, and bile duct carcinomas HCC, breast, lung, and head and neck carcinomas are usually negative |
ID of colon carcinomas and other carcinomas of the gastrointestinal tract However, other carcinomas (e.g., mucinous ovarian carcinoma) can also be positive |
|
| Chromogranin A* | Acidic glycoprotein in neurosecretory granules (cytoplasm, granular) | Islet cells of pancreas, bronchial Kulchitsky cells, parathyroid, adrenal medulla, anterior pituitary, C cells of thyroid | Pheochromocytoma, carcinoids (not rectal), small cell carcinoma, neuroblastoma, some breast and prostatic carcinomas, Merkel cell tumors, islet cell tumors, medullary carcinoma of the thyroid, parathyroid lesions, Brenner tumor | ID of neuroendocrine differentiation in tumors Not present in pituitary prolactinomas Pheochromocytoma (+) versus adrenal cortical carcinoma (−) Parathyroid (+) versus thyroid (−) |
Most specific marker of neuroendocrine differentiation Also can be detected in serum Bouin's solution or B5 fixation may increase immunogenicity |
| Claudin-1(CLDN1) | Protein component of the tight junction complex (membrane—not cytoplasmic) | Epithelial cells, perineurial cells, some endothelial cells (venules) | Perineurioma, synovial sarcoma (epithelioid areas, lower in spindle cell areas) carcinomas Some perineurial cells may be present in neurofibromas and schwannomas |
Perineurioma (+) versus DFSP (−), fibromatosis (−), low-grade fibromyxoid sarcoma (−) | |
| Collagen IV | Major constituent of basement membranes (basement membrane) | Mesangial cells within glomeruli, basement membranes, basal lamina of capillaries | Tumors with external lamina (schwannomas, smooth muscle tumors) | Absence or loss may be associated with stromal invasion by carcinomas | |
| Desmin* | Intermediate filament in muscle (cytoplasm) | All striated muscle (Z bands) and many smooth muscle cells, myofibroblasts, smooth muscle of some blood vessels | Rhabdomyosarcoma (80% +), leiomyosarcoma (50-70% +), PEComa, desmoplastic small round cell tumors (usually dot-like), some myofibroblastic tumors, endometrial stromal sarcoma | ID of muscle differentiation in tumors | |
| DPC4*(homozygously deleted in pancreatic carcinoma, locus 4, Smad4) | Transcriptional regulator interacting with the TGF- beta signaling pathway (nucleus) | Normal tissues | Expressed in most carcinomas Lost in 31% of Pan IN-3, 55% of pancreatic carcinomas, and 22% of stage IV colon carcinomas |
Mucinous ovarian carcinoma (+) versus metastatic pancreatic carcinoma (− in 55%) | Mutated in familial juvenile polyposis in 25-60% of cases |
| E-cadherin | Transmembrane cell adhesion molecule that binds to catenins for cell polarization, glandular differentiation, and stratification (membrane) | Epithelial cells | Most carcinomas—may be lost in poorly differentiated carcinomas Not present in LCIS and invasive lobular carcinoma of breast or gastric signet ring cell carcinomas |
Ductal (+) versus lobular (−) lesions of the breast | Diagnostic importance in the breast has not been established |
| EGFR(Epidermal growth factor receptor, HER1) | Transmembrane protein receptor of the type 1 growth factor family with tyrosine kinase activity (membrane positivity scored, cytoplasmic positivity is not scored) | Many types of epithelium, skin eccrine and sebaceous glands, mesenchymal cells, perineurium. The strongest membrane positivity is present in hepatocytes, bile ducts, basilar squamous cells, pancreatic ducts, breast epithelium, lung alveolar lining cells, mesothelial cells, prostate epithelium, endometrial glands and stroma | Adenocarcinomas (esp. colon), squamous cell carcinomas, TCC, neural tumors, sarcomas | Expression is increased in tumors of higher grade and poorer prognosis Colon carcinomas (80-90% positive): response to cetuximab does not appear to be related to IHC score (see Table 7-29) | Patients with colon carcinomas expressing EGFR are eligible for trials of targeted therapy (cetuximab). |
| Epithelial membrane antigen*(EMA, MUC1, HMFG, DF3, CA 15-3, CA 27.29, PEM, many others) | Episialin, glycoprotein found in human milk fat globule membranes (cytoplasm [more common in malignant cells], membrane [more common in benign cells]) | Epithelial cells, perineurial cells, meningeal cells, plasma cells, usually negative in non-neoplastic mesothelial cells | Carcinomas, mesotheliomas (thick membrane pattern), some sarcomas (synovial sarcoma, epithelioid sarcoma, leiomyosarcoma, some osteosarcomas), adenomatoid tumor, chordoma, perineurioma, neurofibroma, meningioma, desmoplastic small round cell tumor, Sertoli cell tumor Some anaplastic large cell lymphomas (CD30 +), plasma cell neoplasms |
ID of epithelial differentiation in tumors; however, keratin is more specific for this purpose Synovial sarcoma (typically focal positivity) versus other sarcomas |
There are over 50 monoclonal antibodies recognizing different glycosylation patterns in normal tissues and tumors16 |
| Epstein-Barr virus* | |||||
| EBV-encoded nonpolyadenylated early RNAs(EBERS) | RNA produced by EBV (nucleus) | EBV-infected B cells | All EBV-related tumors | Most sensitive marker for EBV | Detected by in situ hybridization for RNA on paraffin sections |
| LMP-I | Latent membrane protein (membrane) | EBV-infected B cells | Nasopharyngeal carcinomas, RS cells (not LP HD), transplant lymphomas, AIDS related lymphomas, endemic Burkitt's lymphoma (rare in sporadic cases) | Evaluation of EBV related neoplasms | |
| EBNA 2(nuclear antigen 2) | Nuclear protein (Nucleus) | EBV-infected B cells | Transplant-related lymphomas, AIDS-related lymphomas. Not present in Burkitt's lymphoma, nasopharyngeal carcinomas, or HD | Evaluation of transplant- and AIDS-related lymphomas | |
| Estrogen receptor*(ER, 1D5, H222, others) | Steroid binding protein (nucleus) | Breast epithelial cells (not myoepithelial cells), epithelial and myometrial cells of the uterus | Breast carcinomas (>70%), gynecologic carcinomas, some skin appendage tumors, rare in other carcinomas, present in some meningiomas, smooth muscle tumors, some melanomas, some thyroid tumors, desmoid tumors, myofibroblastomas of breast, vulvovaginal stromal tumor | Prognosis and prediction of response to hormonal therapy of breast cancer Only nuclear positivity is scored ID of metastatic breast cancer |
Antibodies recognize different epitopes and have varying sensitivities in formalin-fixed tissue. Antigenicity may be diminished after decalcification or exposure to heat during surgery |
| Factor VI ll-related antigen*(VWF, FVIII:RAg, von Willebrand's factor) | Glycoprotein involved in coagulation, part of FVIII complex (cytoplasm) | Endothelial cells, megakary- ocytes, platelets, and mast cells, endocardium | Vascular tumors (often absent in angiosarcomas) Not present in KS, PEComa Megakaryocytic AML (M7) is positive |
ID of endothelial differentiation in tumors (specific but not very sensitive) Evaluation of angiogenesis Evaluation of M7 (megakaryocytic) leukemias |
May not detect smaller blood vessels (see CD 31 and 34). Present in Weibel-Palade bodies. Not a sensitive marker for vascular neoplasms |
| Factor Xllla(Factor XIII subunit A) | Transglutaminase involved in the coagulation pathway (cytoplasm) | Fibroblasts, dendritic reticulum cells in reactive follicles, dermal dendrocytes, liver, placenta, platelets, megakaryocytes, monocytes, macrophages | Fibroblastic neoplasms, dermatofibroma | Not very specific | |
| Fascin | Actin binding protein thought to be involved in the formation of micro-filament bundles (cytoplasm) | Interdigitating reticulum cells in lymph nodes, dendritic cells of lymph node, thymus, spleen and peripheral blood, histiocytes, smooth muscle, endothelial cells, squamous mucosal cells, lining cells of splenic sinuses | RS cells and their variants (but not LP HD), rare non HD lymphomas Reticulum cell tumors Some sarcomas Some high-grade breast carcinomas |
Not very specific | |
| Fibronectin | Glycoproteins found in BMs and extracellular matrix, bind to integrins (extracellular) | Stroma of many tumors | |||
| Fli-I*(Friend leukemia integrin-site 1) | Transcription factor (ETS family)—translocation in Ewing's can result in an EWS-Fli-1 fusion protein (nucleus) |
Endothelial cells (hemangioblasts, angioblasts), small lymphocytes | Ewing's sarcoma/PNET, vascular tumors (including KS), Merkel cell carcinoma, melanoma Can also be weakly present in lymphomas, synovial sarcoma, some carcinomas |
ID of vascular tumors (unlike other vascular markers, Fli-1 is nuclear) ID of Ewing's sarcoma/PNET |
Reactivity can be variable with high background and may be difficult to interpret |
| Galectin-3*(Gal-3) | Lectin with anti-apoptosis function (galactoside- binding protein) (nucleus, cytoplasm, membrane, extracellular matrix) | Many epithelial cells, lymphocytes, mesenchymal cells, macrophages, activated endothelial cells | Many carcinomas, adenomas, lymphomas, soft tissue tumors | Thyroid carcinomas (papillary and to a lesser extent follicular) show higher expression than benign lesions In some carcinomas, expression is diminished in higher-grade lesions |
|
| Glial fibrillary acidic protein(GFAP) | Intermediate filament (cytoplasm) | Normal and reactive astrocytes, developing and reactive ependymal cells, developing oligodendrocytes, choroid plexus, Schwann cells, enteric glial cells, pituitary cells, chondrocytes | Tumors of astrocytes, ependymal cells, and oligodendrocytes, MPNST, myoepitheliomas (salivary glands and soft tissue), sweat gland tumors, Merkel cell carcinomas, chordomas | ID of neural differentiation in tumors (30% of MPNSTs are +) Neuroblastomas are negative, schwannomas may be focally + Merkel cell carcinoma (+) versus small cell carcinoma (−) (but CK20 is a better marker for this purpose) ID of myoepithelial neoplasms |
|
| GLUT-1(glucose transporter 1) | Component of trans- membrane glucose transport (membrane) | Erythrocytes, perineurium, blood vessels, tropho- blasts, renal tubules, germinal center cells | TCC, lung carcinoma, squamous cell carcinoma, adenocarcinomas of colon, lung, bile ducts, kidney, ovary, pancreas, stomach, and endometrium, germ cell tumors | Not very specific | |
| Gross cystic disease fluid protein-15*(GCDFP, CDP, BR-2, BRST-2) | Protein found in breast fluid (cytoplasm) | Apocrine sweat glands, apocrine metaplasia of the breast | Breast carcinomas (60%), sweat gland carcinomas, some salivary gland tumors, some prostate carcinomas | ID of apocrine differentiation in tumors ID of breast metastases (however, only positive in about 60%) |
|
| HepPar-1*(hepatocyte paraffin-1, HP!) | Mitochondrial protein (cytoplasm, coarsely granular) | Liver | HCC, some cases of gastric adenocarcinoma, esophageal adenocarcinoma, others negative or rarely positive | HCC (80-95%) versus metastatic carcinomas to the liver | |
| HBME-1* | Antigen to microvilli on mesothelioma cells (membrane and cytoplasm) | Mesothelial cells, epithelial cells | Mesotheliomas (epithelial type—thick, membrane staining), adenocarcinomas, chordomas, chondrosarcomas | Positivity higher in thyroid carcinomas than in adenomas May be absent in thyroid carcinomas with Hürthle cell features |
Not a specific marker for mesotheliomas |
| HER-2/neu(c-erbB2) | Growth factor receptor (tyrosine kinase) homologous to epidermal growth factor receptor (membrane, some cytoplasm) | Absent or rare in normal cells | Breast carcinomas (20-30%), Paget's disease of nipple (>90%), less frequently in other carcinomas (ovary, uterus, GI, pancreas), some synovial sarcomas | Poor prognostic factor in breast cancer Positivity used to select patients for treatment with Herceptin (scored from 0 to 3+) (see Chapter 15) |
Only membrane positivity is scored Gene amplification (detected by FISH) correlates with strong complete membrane immunoreactivity in >90% of cases |
| HHF-35*(Muscle-specific actin, MSA, muscle common actin, EM ACT) | Alpha and gamma smooth muscle actins, recognizes a common epitope of alpha skeletal, cardiac, and smooth muscle (cytoplasm) | Smooth, striated, and cardiac muscle, smooth muscle of blood vessels, pericytes, myoepithelial cells, myofibroblasts | Numerous tumors including tumors of muscle, glomus tumor, PEComa, GIST, DFSP, dermatofibroma, myofibroblastic tumors, spindle cell carcinomas, salivary gland tumors, mesothelioma, others | ID of muscle differentiation in tumors | Sensitive but not specific. Present in tumors not of muscle origin |
| HHV8* | Latent nuclear antigen of human herpesvirus type 8 (nucleus) | Absent in normal tissue | KS (endothelial cells and some perivascular cells) Primary effusion lymphoma (PEL), AIDS-associated multicentric Castleman's disease |
Evaluation of KS and PEL | |
| HMB-45*(E-MEL, melanoma- specific antigen) | Oligosaccharide side-chain of a melanosomal antigen, gp100/pmel17 (cytoplasm) | Fetal melanocytes and some normal adult superficial melanocytes, retinal pigment epithelium | Melanoma (epithelioid but not spindle cell or desmoplastic type), clear cell sarcoma, PEComa, tumors associated with tuberous sclerosis, melanotic schwannoma, others | ID of metastatic melanoma. Melanophages can also be positive Melan-A may be more specific ID of PEComa |
NKI-betab detects the same protein Tissues fixed in B5 may have high background staining |
| hMLHI(human mutS homologue 2)and hMSH2(human mutL homologue 1) | Proteins involved in mismatch repair of DNA (these genes account for 95% of HNPCC) (nucleus) | Most normal tissues May be lost in areas of chronic inflammation |
Expression (or non- expression) is not specific for tumor type | Absence is associated with germline mutations in HNPCC patients and with gene silencing by methylation in 15% of sporadic colon carcinomas —correlated with characteristic clinical, pathologic, and treatment response features IHC will not detect the 5% of patients with mutations in other genes or rare patients with mutated gene products that are immunoreactive | Other assays for microsatellite instability utilize PCR (90% sensitive for microsatellite instability (MSI) |
| Hormones (ER and PR are listed separately) | Insulin, gastrin, glucagon, somatostatin, calcitonin, ACTH, FSH, LH, PRL, TSH, others (cytoplasm) | Hormone-producing cells | Hormone-producing tumors | ID of hormone products in tumors | May not correlate well with serum levels of the same markers May not correlate with response to hormonal therapies (e.g., ER in tumors other than breast and tamoxifen) |
| Human chorionic gonadotropin*(hCG, β-hCG) | Beta chain of the hormone (cytoplasm) | Syncytiotrophoblasts | Choriocarcinoma, giant cells in seminomas, placental site tumors (weak) | ID of trophoblastic differentiation in tumors | |
| Human placental lactogen*(HPL, hPL) | Hormone (cytoplasm) | Trophoblast | Choriocarcinoma (may be weak), complete moles (strong), partial moles (weak), some lung and stomach carcinomas | ID of trophoblastic differentiation in tumors | |
| Inhibin*: alpha subunit | Hormone produced by ovarian granulosa cells and prostate, inhibits FSH production (cytoplasm) | Ovarian granulosa cells, Sertoli cells, pregnancy luteomas, ovarian follicles, syncytiotrophoblast, adrenal cortex, hepatocytes | Granulosa cell tumors, juvenile granulosa cell tumors, Sertoli and Leydig cell tumors, ovarian stromal cells around other tumors, hydatidiform moles, choriocarcinoma, thecofibroma, adrenal cortical tumor, granular cell tumor | ID of sex cord stromal differentiation in ovarian tumors Distinguishes adrenal cortical tumors (>70% +) versus HCC (<5% +) and RCC (<5% +) |
|
| Keratins* | Intermediate filaments (cytoplasm) | Epithelial cells | Carcinomas, mesotheliomas, desmoplastic small round cell tumors (dot-like pattern), thymomas, chordomas, synovial sarcomas, epithelioid sarcoma, leiomyosarcoma, trophoblastic tumors, some other sarcomas, rarely melanomas | ID of poorly differentiated carcinomas Site of origin of carcinomas |
|
| AE1/AE3* | Two monoclonal antibodies. AE1 detects 10, 15, 16, and 19. AE3 detects 1 to 8. (Cytoplasm) | Most carcinomas. The only common carcinomas that are frequently negative are HCC (70% negative) and RCC, clear cell type (20% negative) Epithelioid hemangio-endothelioma, epithelioid sarcoma, synovial sarcoma, mesothelioma, adenomatoid tumor |
ID of epithelial differentiation in tumors. HCC (−/+) versus cholangio- carcinoma and metastatic carcinomas (+) | Good broad-spectrum keratin | |
| CAM 5.2* | 8, 18 (cytoplasm) | Simple and glandular epithelium | Most carcinomas including those usually negative for CK7 and 20: HCC, prostatic carcinoma, thymic carcinoma, gastric carcinoma, RCC small cell carcinoma Carcinoid tumor, thymoma, germ cell tumors, mesothelioma, dendritic cells Synovial sarcoma, epithelioid sarcoma Many squamous cell carcinomas are negative |
ID of carcinomas that may be negative for CK7 and CK20 Paget's disease (+) versus squamous cell carcinoma (−) Positivity for dendritic cells in lymph nodes and elsewhere may be confused for micrometastases |
May be positive when other keratins are negative |
| Keratin 5/6* | 5/6 (cytoplasm) | Basal cells, stratum spinosum of epidermis, mesothelial cells | Squamous cell carcinomas, TCC, mesotheliomas, squamous metaplasia in adenocarcinomas, thymic carcinoma | Less frequently present in non-squamous cell carcinomas | Has limited use in routine practice |
| Keratin 7* | 7 (cytoplasm) | Simple epithelia, respiratory epithelium, transitional epithelium, endothelial cells of small veins and lymphatics Not present in squamous epithelium |
Most adenocarcinomas of glandular epithelial origin, TCC, mesothelioma, neuroendocrine neoplasms Not Merkel cell carcinoma or colon carcinoma Rare in clear cell RCC (but present in other variants), prostate carcinoma, HCC, lung small cell carcinoma, thymoma, carcinoid Not present in squamous cell carcinomas of the skin, but may be present in squamous cell carcinomas arising from non-keratinizing epithelium (e.g., cervical carcinoma) |
The combination of CK7 and CK20 is used to distinguish carcinomas arising at different sites (see Table 7-3, Table 7-4, Table 7-5, Table 7-6, Table 7-7) | |
| Keratin 20* | 20 (cytoplasm) | Gastric foveolar epithelium, intestinal villi and crypt epithelium, Merkel cells, taste buds, umbrella cells of urothelium, subsets of epithelial cells Not present in non- epithelial cells |
Colon carcinoma, Merkel cell carcinoma, TCC, adenocarcinoma of the bladder, pancreatic carcinoma, cholangio- carcinoma, mucinous ovarian carcinoma, esophageal adenocarcinoma | Merkel cell carcinomas CK20 positive, whereas most similar tumors are negative ID of metastatic colon carcinomas (the pattern of CK7-, CK20 + is most frequently seen in this carcinoma, but can rarely be seen in other types) |
|
| PAN-K*(MNF-116) | Broad spectrum detection of keratins including 5, 6, 8, 17, and 18 (cytoplasm) | Simple and squamous epithelial cells | Detection of keratin in all carcinomas, including poorly differentiated carcinomas (esp. spindle cell squamous cell carcinomas) May be more sensitive than AE1/AE3 for carcinomas with myoepithelial (“basal”) features due to inclusion of the “basal” keratin CK17 |
||
| 34βE12*(903) | High-molecular-weight keratins including 1,5, 10, 14 (cytoplasm) | Complex epithelia, basal cells, myoepithelial cells | TCC, cholangiocarcinoma, squamous cell carcinoma, non-mucinous bronchiolo- alveolar lung carcinoma, RCC (papillary and chromophobe types), mesothelioma, papillary thyroid carcinoma, thymic carcinoma, lympho-epithelial carcinoma | TCC (+) versus prostate carcinoma (−) or RCC (−) Prostate carcinoma (no basal cells) versus benign lesions (with some + basal cells present). Can be combined with p63 for this use |
|
| Ki-67*(MIB-1) | Protein found during the entire cell cycle but not in G0 (nucleus) | Any cycling cell | Any cycling tumor | Used as a prognostic marker for some tumors Detects number of cycling cells in Burkitt's lymphoma and large B-cell lymphoma Aberrant membrane and cytoplasmic immuno-reactivity is present in trabecular hyalinizing adenoma of the thyroid and sclerosing hemangioma of the lung |
MIB-1 recognizes an epitope preserved in formalin- fixed tissue |
| Laminin | Component of basement membranes (basement membrane) | Basement membranes | Nerve sheath tumors, smooth muscle tumors | Loss associated with stromal invasion by carcinomas Present in microglandular adenosis of the breast |
|
| Lysozyme(Ly) | Muramidase (mucolytic enzyme) (cytoplasm) | Circulating monocytes, some tissue macrophages, granulocytes, salivary gland, lacrimal gland, stomach and colon epithelial cells (inflamed or regenerative), apocrine glands, Paneth cells, some other epithelial cells | Salivary gland tumors, stomach and colon carcinomas AML with monocytic differentiation | Marker for histiocytes but not specific. May mark activated phagocytic macrophages Evaluation of myeloid leukemias |
Not specific for identification of solid tumors |
| MAC 387(L1 antigen, calprotectin, calgranulin, cystic fibrosis antigen) | Three polypeptide chains released with activation or death of neutrophils (cytoplasm) | Neutrophils, monocytes, some tissue macrophages, eosinophils, squamous mucosa, reactive skin, synovial lining cells | Lung carcinomas (not small cell or carcinoid), squamous cell carcinomas Histiocytic neoplasms (but not Langerhans cells) |
Marker for macrophages (but not as specific as CD68) | Belongs to the S100 protein family Cells can passively take up antigen resulting in false positive results |
| Melan-A or MART.!*(melanoma antigen recognized by T cells), A103) | Melanocyte differentiation antigen (cytoplasm) Melan-A (clone A103) and MART-1 are two different antibodies |
Melanocytes of skin, uvea, and retina Melan-A is also positive in adrenal cortex, granulosa and theca cells of the ovary, Leydig cells |
Melanomas (but <50% of spindle cell or desmoplastic melanomas), PEComas Melan-A is also positive in adrenocortical tumors, Leydig cell tumor, granulosa cell tumor |
ID of melanomas. Melan-A is not positive in melano- phages and may be more specific for the detection of micrometastases in lymph nodes Melan-A distinguishes adrenocortical tumors (−) (>50% +) versus HCC and RCC (−) |
More sensitive than HMB45 Peptides are used for melanoma immunotherapy Melan-A has a broader spectrum of immunoreactivity than MART-1 |
| Myf-4*(MRF4, myogenin) | Human homologue of myogenin— muscle regulatory protein (nucleus) | Striated muscle | Rhabdomyosarcoma | ID of skeletal muscle differentiation in tumors | Better than MyoD1 |
| MyoDI | Nuclear phosphoprotein, role in myogenic regulation (nucleus) | Developing muscle tissues (myoblasts), adult muscle is negative | Rhabdomyosarcoma (esp. poorly differentiated tumors), mixed mullerian tumors | ID of skeletal muscle differentiation in tumors | Background positivity is often high, making interpretation difficult |
| Myoglobin | Oxygen binding protein (cytoplasm) | Striated muscle (including cardiac muscle), not smooth muscle | Tumors of striated muscle (rhabdomyosarcoma + 50%), but often negative in poorly differentiated tumors | ID of skeletal muscle differentiation in tumors | More specific but less sensitive than actin and desmin |
| Myosin Heavy Chain (fast)(SM-MHC, Fast myosin) | Contractile protein with 2 heavy and 4 light chains and many isoforms (cytoplasm) | Visceral and vascular smooth muscle, myoepithelial cells of the breast Striated muscle: type 2 fibers | Tumors with myoepithelial cells Rhabdomyosarcoma (some) |
Marker for myoepithelial cells in the breast—may have less positivity in vascular endothelial cells and myofibroblasts ID of skeletal muscle differentiation in tumors |
Antibodies to different isoforms will detect different types of muscle fibers |
| NEU N | (Neuronal nuclei) | Neuronal cells including cerebellum, cerebral cortex, peripheral ganglion cells | |||
| Neurofilaments(70 + 200 kD, NFP) | Intermediate filaments with three subunits (cytoplasm) | Neuronal cells, adrenal medulla | Tumors of neuronal origin or with neuronal differentiation, neuroblastoma, medulloblastoma, retinoblastoma, Ewing's/PNET, esthesioneuroblastoma, Merkel cell carcinoma, some endocrine tumors (carcinoids, pheochromocytomas) | ID of neuronal differentiation in tumors ID of Merkel cell carcinomas |
|
| Neuron-specific enolase*(NSE—do not confuse with the enzyme non-specific esterase) | Gamma-gamma enolase isoenzyme (cytoplasm) | Neuroectodermal and neuroendocrine cells, more weakly striated and smooth muscle, megakaryocytes, T cells, some platelets, neurons, pituitary cells, hepatocytes | Neuroectodermal and neuroendocrine tumors, melanomas (including desmoplastic melanomas), many breast carcinomas, germ cell tumors, alveolar soft part sarcoma | ID of neuronal or neuroendocrine differentiation in tumors | Lacks specificity |
| p16(MTS1, CDKN2) | Binds to and inhibits the cyclin-dependent kinases cdk4 and cdk6 (cytoplasm and nuclear) | Absent | Cervical squamous cell carcinomas and adeno- carcinomas (both in situ and invasive), endocervical carcinoma, endometrial carcinoma Some basaloid squamous cell carcinomas of the tonsil in young patients that are associated with HPV16 |
Evaluation of cervical lesions Possible use predicting tonsillar site for metastatic squamous cell carcinoma of the head and neck | Overexpression is due to HPV-induced cell cycle dysregulation |
| p27klpl | A cyclin-dependent kinase inhibitor that regulates progression from G1 to S phase | Proliferating cells | |||
| p53*(Multiple antibodies to wild-type and mutant forms) | Tumor suppressor gene product—probably most frequently mutated gene in malignancy (nucleus) | Overexpression uncommon or absent in normal cells or benign tumors | Many malignant tumors, but not specific for malignancy | Overexpression may be used as a prognostic factor | Different antibodies recognize different wild type and mutant forms of the protein and will give different results |
| p57(kip2, p57KIP2) | Cyclin-dependent kinase inhibitor (CDKI) acting to inhibit cell proliferation, paternally imprinted (nucleus) | Cytotrophoblast, intermediate trophoblast, villous stromal cells, decidual stromal cells, absent in syncytiotrophoblast | Diploid complete moles show absent or low expression in cytotrophoblast and villous stromal cells (may be present in villous intermediate trophoblast and decidual stromal cells), partial moles and hydropic abortions have normal expression | ||
| p63* | Protein with at least six major isotypes, including deltaNp63, member of the p53 family (nucleus) | Proliferating basal cells of cervix, urothelium, prostate, and myoepithelial cells of breast, basal squamous cells, squamous metaplasia | Squamous cell carcinomas, TCC, adenomyo- epithelioma, adenoid cystic carcinoma, nasopharyngeal carcinoma, “basal type” breast carcinomas, papillary carcinoma of the thyroid, others | ID of myoepithelial cells in sclerosing breast lesions Diagnosis of prostatic carcinoma by showing absence of basal cells (more sensitive when combined with 34βE12). Basaloid squamous lung cancer (+) versus small cell (−). ID of metastatic poorly differentiated squamous cell carcinomas |
Easier to interpret than SMA as myofibroblasts are negative |
| Placental alkaline phosphatase*(PLAP) | Alkaline phosphatase secreted by trophoblast (cytoplasm) | Placenta (trophoblast) | Germ cell tumors (but not spermatocytic seminoma), intratubular germ cell neoplasia, partial moles, some carcinomas of breast, ovary, lung, stomach, and pancreas, some rhabdomyosarcomas (esp. alveolar type) | Absence of immunoreactivity makes a germ cell tumor unlikely. However, spermatocytic seminomas and immature teratomas are negative ID of intratubular germ cell neoplasia |
|
| Progesterone receptor*(PR, PgR) | Steroid binding protein (nuclear) | Normal breast epithelial cells, endometrial cells, many smooth muscle cells, breast lobular stroma | Breast carcinomas, gynecologic carcinomas, some skin adnexal tumors, secretory meningiomas, endometrial stromal sarcomas, some leiomyomas, myofibro-blastic tumors, rarely other tumors | Prognosis and treatment of breast cancer ID of metastatic breast cancer |
|
| Prealbumin(Transthyretin, TTR) | Plasma transport protein for retinol and thyroxine (cytoplasm) | Pancreatic islet cells, choroid plexus, retinal pigment epithelium, liver | Pancreatic islet cell tumors, carcinoid tumors, choroid plexus papillomas, choroid plexus carcinomas (may be focal or absent) | ID of choroid plexus neoplasms Evaluation of some forms of amyloidosis |
Major subunit protein in some forms of inherited systemic amyloidosis |
| Prostate-specific antigen*(PSA) | Member of kallikrein family of serine protease isolated from human seminal plasma (cytoplasm) | Normal prostatic epithelium, urachal remnants, endometrium, transitional cells of bladder | Prostatic carcinomas, some breast carcinomas | ID of prostatic carcinomas (may be lost in some poorly differentiated carcinomas). Seminal vesicle carcinomas are negative | More specific than PAP Used as a serum screening test for prostate cancer |
| Prostate acid phosphatase*(PrAP, PAP) | Isoenzyme of acid phosphatase (cytoplasm) | Normal prostatic epithelium, periurethral glands, anal glands, macrophages | Prostatic carcinomas, TCC, rectal carcinoids | ID of prostatic carcinomas (may be lost in some poorly differentiated carcinomas) | |
| RCC*(Renal cell carcinoma marker, gp200) | Glycoprotein on surface of renal tubules, breast epithelial cells, epididymis (cytoplasm, membrane) | Renal tubules, breast, epididymis | Clear cell and papillary RCC, breast carcinoma, embryonal carcinoma | Clear cell and papillary RCC (+) versus chromophobe carcinoma (−/+) and oncocytoma (−) | |
| RET*(Rearranged during transfection) | RET proto-oncogene. Surface glycoprotein of the receptor tyrosine kinase family (cytoplasm) | Neurons, embryonic kidney | Papillary thyroid carcinomas (78%), follicular variant of papillary carcinoma (63%), Hürthle cell carcinoma (57%), insular carcinoma (50%), medullary carcinoma, not present in follicular carcinomas or benign lesions Neuroblastoma (+), pheochromocytoma (+) |
Evaluation of thyroid tumors | Germline mutations occur in MEN2A and 2B (10q11.2), familial medullary thyroid carcinoma, and some cases of Hirschsprung's disease |
| S100 protein* | Calcium binding protein isolated from the CNS (member of the EF hand family) (nucleus and cytoplasm) | Glial and Schwann cells, melanocytes, chondrocytes, adipocytes, myoepithelial cells, Langerhans cells, macrophages, reticulum cells of lymph nodes, eccrine glands, others | Melanoma (including spindle cell and desmoplastic types), clear cell sarcoma, schwannoma, chordoma, ependymoma, astroglioma, Langerhans proliferative disorders, some carcinomas (e.g., breast, ovary endometrial, thyroid), granular cell tumor, histiocytic sarcoma, myoepithelioma | ID of melanoma (if negative, melanoma is highly unlikely) ID of Langerhans proliferative disorders Sustentacular cells in pheochromocytomas (loss may be poor prognostic factor) ID of neural tumors ID of cellular schwannomas (more strongly and diffuselypositive than in MPNST) |
Langerhans cells and macrophages in tumors may be misinterpreted as positivity in the tumor itself S100 is very soluble and may be eluted from frozen tissues |
| Synaptophysin* | Transmembrane glycoprotein found in presynaptic vesicles (cytoplasm) | Neuroectodermal and neuroendocrine cells, neurons | Medulloblastoma, neuroblastoma, pheochromocytoma, paragangliomas, carcinoids, small cell carcinoma, medullary carcinoma of the thyroid, neural neoplasms, pancreatic islet cell tumors | ID of neuroendocrine differentiation in tumors ID of neuronal differentiation in CNS tumors |
|
| Synuclein-I | Neuron-specific protein associated with synaptosomes (Lewy bodies) | Brain | Present in Lewy bodies (Lewy body dementia and Parkinson's disease) | ||
| Tau | Microtubule-associated protein (cytoplasm, extracellular) | Normal neuronal cell bodies and dendrites, neuropil, glial cells | Abnormal amounts in Alzheimer's disease in neurofibrillary tangles and senile plaques | Evaluation of Alzheimer's disease, Pick's disease, supranuclear palsy corticobasal degeneration, others | |
| Thyroglobulin* | Glycoprotein produced by thyroid follicular cells (cytoplasm) | Thyroid follicles | Thyroid carcinomas (papillary, follicular, and Hürthle cell types, rarely present in medullary carcinomas) | ID of metastatic thyroid carcinoma Loss may be a poor prognostic factor |
Thyroglobulin can diffuse into metastatic tumors to the thyroid |
| TTF-I*(Thyroid transcription factor 1) | Transcription factor for thyroglobulin, thyroid peroxidase, Clara cell secretory protein, and surfactant proteins (nucleus; aberrant cytoplasm positivity in HCC) | Thyroid, lung, and some brain tissues | Thyroid carcinomas (including medullary carcinoma; may be negative in anaplastic carcinoma), lung adenocarcinomas (75%, but lower in mucinous bronchioloalveolar carcinomas), small cell carcinoma of lung (>90%), HCC (cytoplasmic), absent or focal in most other adenocarcinomas | Mesothelioma (−) versus adenocarcinoma (+/−) Lung adenocarcinoma (+/−) versus metastatic breast carcinoma (−) Small cell carcinoma of lung (+) versus metastasis from other sites (−), but some extrapulmonary small cell carcinomas can also be + HCC (cytoplasmic 71%), rare in other tumor types |
The detection of cytoplasmic TTF-1 may depend on the specific antibody used and the antigen-retrieval method |
| Ulex(Ulex europaeus 1 lectin, UEA 1) | Lectin, fucose residues on blood group H (cytoplasm) | Endothelial cells | Vascular tumors, some carcinomas | Evaluation of angiogenesis | Not very specific |
| Vimentin* | Intermediate filament (cytoplasm) | Mesenchymal cells, fibroblasts, endothelial cells, chondrocytes, histiocytes, lymphocytes, many glial cells, myoepithelial cells, smooth muscle | All mesenchymal tumors, neural tumors, melanomas, meningiomas, chordoma, Leydig cell tumor, granulosa cell tumor, Sertoli cell tumor, adrenal cortical adenoma May be co-expressed with keratin in carcinomas of endometrium, thyroid, kidney (clear cell), adrenal cortex, lung, salivary gland, ovary, and liver |
May be poor prognostic factor if co-expressed with keratin or GFAP | Can be used as an internal control for immunogenicity Not a specific marker for tumor type or line of differentiation |
| WTI*(Wilms' tumor 1 protein) | Zinc finger transcription factor (cytoplasm, nucleus) | Sertoli cells, decidual cells of uterus, granulosa cells of ovary, blood vessels, myelocytic cells | Wilms' tumors (epithelial and blastemal components), epithelial mesotheliomas (nuclei—80-90%), acute leukemia (nuclei), adenocarcinomas (cytoplasmic; esp. breast, ovary), desmoplastic small cell tumors (nuclear and cytoplasmic), rhabdomyosarcoma | Mesothelioma (+, nuclear) versus adenocarcinoma (adenocarcinoma usually negative for nuclear positivity except for ovarian)—monoclonal antibody used Desmoplastic small cell tumors—use polyclonal antibody |
The gene is located on 11p13 and is inactivated in 5-10% of sporadic Wilms' tumors and nearly 100% of Denys-Drash syndrome patients Antibodies detect epitopes at different ends of the protein and may give different results. Not very specific |
| Hematopathology markers | |||||
|---|---|---|---|---|---|
| NAME (ALTERNATE NAME) | ANTIGEN (LOCATION) | NORMAL CELLS AND TISSUES | TUMORS | USES | COMMENTS |
| ALK Protein*(Anaplastic lymphoma kinase, ALK-l, p80) | The ALK gene (2p23) is translocated to part of the nucleophosmin (NPM) gene (5q35) to form the fusion protein p80 and is overexpressed (cytoplasm, nucleus) | Nervous system | Anaplastic (CD30+) large cell lymphomas— approximately one third have t(2;5)(p23; q35) Some inflammatory myofibroblastic tumors | ID of anaplastic large cell lymphomas | The pattern of immunoreactivity varies with the translocation present |
| Alpha-1- antichymotrypsin(ACH) | Serine protease inhibitor (cytoplasm) | Histiocytes, granulocytes, others | Histiocytic tumors, many adenocarcinomas, melanomas, many sarcomas | Marker for histiocytes but CD68 is more specific | Not specific for tumor type |
| Alpha-1-antitrypsin(AAT, α,-AT) | Glycoprotein synthesized in the liver that inhibits proteolytic enzymes (esp. elastase) (cytoplasm) | Histiocytes, reticulum cells, mast cells, Paneth cells, salivary gland | HCC, germ cell tumors, histiocytic neoplasms, colon and lung carcinomas, others | Accumulates in liver cells in AAT deficiency | Not specific for tumor type CD68 is a more specific marker for macrophages |
| bcl-2* | Protein involved in inhibition of apoptosis (membrane, cytoplasm) | Medullary lymphocytes and epithelial cells of the normal thymus, mantle and T zone small lymphocytes | CLL, mantle cell lymphoma, follicular lymphoma, marginal zone lymphoma Synovial sarcoma, other soft tissue tumors |
Follicular center cell lymphomas (+) versus reactive follicles (−). Hyperplastic marginal zones of the spleen, abdominal lymph nodes, and ilial lymphoid tissue are + Malignant thymomas may have greater reactivity than other thymomas Synovial sarcoma is more frequently positive compared to mesothelioma |
Involved in the t(14;18) found in 90% of FCC lymphomas Not specific for ID of solid tumors |
| Bcl-6* | Proto-oncogene—Kruppel- type zinc finger protein with homology to transcription factors (nucleus) | Normal germinal center B cells | Follicular lymphomas, diffuse large B-cell lymphomas, Burkitt lymphoma, mediastinal large B-cell lymphoma, LP HD Not present in B-CLL, hairy cell leukemia, mantle cell lymphoma, and marginal zone lymphomas |
Evaluation of B-cell lymphomas | Involved in gene rearrangements at 3q27 in lymphomas |
| Blood group antigens | A, B, and H antigens (membrane) | Epithelial cells, endothelial cells, erythrocytes | Abnormally expressed or lost in many carcinomas | Sometimes helpful in identifying specimens | |
| BOB. I*(B-cell Oct-binding protein 1) | Coactivator that interacts with Oct transcription factors in B cells (cytoplasm) | B cells (including plasma cells) | B-cell lymphomas and leukemias, Reed- Sternberg cells in LP HD, usually absent in other HD types | Evaluation of HD | BOB.1 and Oct2 are necessary (but not sufficient) for Ig expression |
| BSAP*(β-ce//specific activator protein) | Transcription factor encoded by the Pax-5 gene that regulates B-lineage specific genes | B cells | All B-cell neoplasms and HD | Not reliable in Zenker's fixed tissue | |
| CD la*(T6) | Membrane glycoprotein (membrane) | Cortical thymocytes (immature T cells), Langerhans cells, dendritic cells | Langerhans proliferative disorders, lymphoblastic lymphoma | Evaluation of Langerhans proliferative disorders Evaluation of lymphoblastic lymphoma |
|
| CD2*(TE, Tí 1, rT3, Leu 5a + b, LFA-2) | Glycoprotein mediating adhesion of activated T cells and thymocytes with antigen-presenting cells and target cells, functions in E rosette formation (membrane) | T cells, NK cells, cortical thymocytes | T-cell neoplasms, may be aberrantly lost in peripheral T- cell neoplasms | Pan T cell marker | |
| CD3*(T3) | C3 antigen (five polypeptide chains) (membrane, cytoplasm) | T cells, cortical thymocytes | T-cell neoplasms, may be aberrantly lost in peripheral T-cell neoplasms Anaplastic large cell lymphoma is often negative |
Best pan T cell marker | In paraffin sections, NK cells may also be positive |
| CD4*(TH, T4, Leu 3) | Transmembrane glycoprotein, HIV receptor (membrane) | T helper/inducer cells, macrophages, Langerhans cells | MF, other T-cell neoplasms | Evaluation of MF Evaluation of T-cell neoplasms |
|
| CD5*(Leu 1) | Transmembrane glycoprotein (membrane) | T cells and B-cell subsets (mantle zone) | T-cell leukemias and lymphomas, aberrantly expressed in low grade B-cell lymphomas (CLL or mantle cell lymphoma) Thymic carcinoma, adeno-carcinomas, mesothelioma (cytoplasmic) |
Classification of low-grade B-cell lymphomas Evaluation of T-cell lymphomas (this marker is frequently lost) Thymic carcinoma (∼40%) thymoma (<10%) versus pulmonary squamous cell carcinoma (<5%) |
|
| CD7*(Leu 9) | Membrane-bound glycoprotein (membrane) | Precursor T cells, T-cell subsets, NK cells, thymocytes | T-cell lymphomas and leukemias | Frequently (50%) lost in T-cell lymphomas versus reactive T cells (+) Evaluation of T-cell leukemias |
|
| CD8*(T8, Leu 2) | Two glycoprotein chains (membrane) | T-cell subsets, NK cells, T cytotoxic/suppressor cells | T-cell lymphomas and leukemias | Evaluation of MF and T-cell lymphomas (this marker is frequently lost) | |
| CD10*(CALLA [common acute leukemia antigen], J5, neprilysin) | Cell surface metallo- endopeptidase that inactivates peptides (membrane) | Precursor B cells, granulocytes, rare cells in reactive follicles, myoepithelial cells of breast, bile canaliculi, fibroblasts, brush border of kidney and gut | Follicular lymphomas, pre- B-ALL, Burkitt's lymphoma, CML, angioimmunoblastic lymphoma RCC (clear cell and papillary), HCC, rhabdomyosarcoma, endometrial stromal sarcoma |
Evaluation of follicular center cell lymphomas Evaluation of leukemias Myoepithelial cell marker in breast Endometrial stromal sarcoma (+) versus leiomyosar-coma (−) (but caldesmon is preferred for this purpose) |
|
| CD11b(Mac-1) | Cell surface receptor for the C3bi complement fragment (membrane) | Granulocytes, monocytes, macrophages | Myelomonocytic leukemias | ||
| CD11c* | Member of the beta(2) integrin family that mediates adhesion to vascular endothelium, transendothelial migration, chemotaxis, and phagocytosis (membrane) | Myeloid cells, NK cells, dendritic cells, activated lymphoid cells | Hairy cell leukemia, B-cell prolymphocytic leukemia, some B-CLL, marginal zone lymphoma (MALT) | ||
| CDI 3(My 7) | Aminopeptidase-N, a type II integral membrane metalloprotease functioning in cell surface antigen presentation, receptor for coronaviruses (membrane, cytoplasm) | Granulocytes, macrophages, bone marrow stromal cells, osteoclasts, renal tubules, intestinal brush border, cells lining bile duct canaliculi, endothelial cells, fibroblasts, brain cells | AML, CML with blast crisis, some ALL | Classification of leukemias | Requires frozen tissue |
| CD15*(Leu-M1) | 3-fucosyl-N-acetyllactosamine, X-hapten—CHO moiety linked to cell membrane protein (membrane and granular perinuclear) | Granulocytes, monocytes | Reed-Sternberg cells (not LP HD), some large T-cell lymphomas, MF, some leukemias, some epithelial cells (adenocarcinomas), CMV-infected cells | Adenocarcinomas (+) versus mesotheliomas (−) Evaluation of HD |
|
| CD16* | Low-affinity transmembrane Fc receptor for IgG (membrane) | NK cells, granulocytes, activated macrophages, subsets of T cells | Extranodal NK/T-cell lymphoma, some hepatosplenic T-cell lymphomas | ||
| CD19(B4) | B-cell type I integral membrane glycoprotein (membrane) | B cells, follicular dendritic cells, early myelomono- cytic cells | pre-B-ALL and B-cell neoplasms (but not plasma cell lesions) | Good pan B cell marker | Fresh or frozen tissue required |
| CD20*(lib, Bl, Leu 16) | B-cell non-glycosylated phosphoprotein functioning as a receptor during B-cell activation and differentiation (membrane, cytoplasmic) | B cells, monocytes, not plasma cells | B-cell lymphomas, Reed- Sternberg cells in LP HD, not plasmacytomas | Best pan B cell marker. Evaluation of B-cell lymphomas Evaluation of HD |
Under investigation as a target for clinical treatment of B-cell lymphomas L26 is best for formalin- fixed tissue May be preserved in necrotic tissue |
| CD21*(B2) | Type I integral membrane glycoprotein functioning as the receptor for the C3d fragment of complement C3, CR2, receptor for EBV (membrane) | Follicular dendritic cells, mature B cells | Marginal zone (MALT) lymphomas, CLL (B cell), some T cell ALL, follicular dendritic cell tumors | ID of residual follicular structure in LP HD and other diseases Evaluation of low-grade B-cell lymphomas ID of follicular dendritic cell sarcoma |
|
| CD22*(BL-CAM) | Type I integral membrane glycoprotein (membrane, cytoplasm) | B cells, precursor B cells | B-cell neoplasms (but not plasma cell lesions) | Pan B cell marker | |
| CD23* | Membrane glycoprotein, low-affinity IgE receptor (membrane) | Subpopulation of peripheral B cells, follicular dendritic cells | CLL, but usually not mantle zone lymphoma, MALTomas, or follicular lymphomas | Evaluation of low-grade B-cell lymphomas | |
| CD25*(IL-2 receptor) | Interleukin-2 receptor (membrane, cytoplasm) | Subpopulation of T cells, myeloid precursors, oligodendrocytes HTLV-1 transformed T and B cells |
Hairy cell leukemia, adult T-cell lymphoma/leukemia, some T-cell prolympho- cytic leukemia, precursor lymphoblastic lymphoma, and anaplastic large cell lymphoma | Evaluation of cutaneous T-cell lymphomas for potential anti-CD25 therapy | |
| CD30*(Ki-I, BERH2) | Single chain transmembrane glycoprotein, homologous to the nerve growth factor superfamily (cytoplasm, membrane and Golgi) | Activated B and T cells, some plasma cells, immunoblasts, interdigitating cells, histiocytes, follicular center cells, decidualized endometrium, reactive mesothelial cells, most other tissues negative | Anaplastic (CD30+) large cell lymphomas, large B-cell lymphoma, primary effusion lymphoma, mediastinal large B-cell lymphoma, Reed- Sternberg cells (not LP HD), enteropathy T-cell lymphoma, peripheral T-cell lymphoma, EBV-transformed B cells Embryonal carcinoma, vascular tumors (not KS), some mesotheliomas, rarely carcinomas are positive |
Evaluation of anaplastic (CD30+) lymphomas. Evaluation of HD (Reed-Sternberg cells are positive except in LP HD) Evaluation of peripheral T-cell lymphoma (large cells may be positive) |
|
| CD33*(My 9) | Myeloid-specific receptor (sialic acid-binding immunoglobulin-like lectin or Siglec-3) (membrane) | Granulocytes, monocytes | AML | Evaluation of leukemias | Gemtuzumab ozogamicin is a humanized CD33 antibody linked to an antitumor antibiotic calicheaminin for the treatment of AML |
| CD34*(HPCA-1, QBEnd10) | Single chain transmembrane glycoprotein (cytoplasm, membrane) | Lymphoid and myeloid hematopoietic progenitor cells, endothelial cells, some skin cells, myofibroblasts | Acute leukemia Neurofibroma, angiosarcoma, KS, epithelioid hemangio- endothelioma, solitary fibrous tumor, DFSP, epithelioid sarcoma, GIST, myofibroblastic tumors |
ID of endothelial or myofibroblastic differentiation in tumors Evaluation of angiogenesis Evaluation of the number of blasts in bone marrow in acute leukemia. |
Not specific for endothelial cells |
| CD35*(CRI, C3b/C4b R) | Transmembrane protein t that binds complement components C3b and C4b and mediates phagocytosis (membrane) | Erythrocytes, B cells, a subset of T cells, monocytes, neutrophils, eosinophils, glomerular podocytes, follicular dendritic cells | Marginal zone (MALT) lymphoma, follicular dendritic cell tumors | Detects follicular dendritic cells ID of follicular dendritic cell sarcomas |
|
| CD38* | Type II transmembrane glycoprotein with enzymatic action for the formation and hydrolysis of cADPR (membrane) | Immature B and T lymphocytes, thymocytes, mitogen-activated T cells, Ig-secreting plasma cells, monocytes, NK cells, erythroid and myeloid progenitors, brain cells | Acute leukemias, plasma cell lesions Neurofibrillary tangles in Alzheimer's disease |
ID of plasma cell lesions | Immunoreactivity may be a poor prognostic marker for patients with CLL |
| CD43*(Leu 22, L60) | Cell surface glycoprotein (membrane) | T cells, macrophages, granulocytes | AML (chloromas), T-cell neoplasms, aberrant expression in some low grade B-cell neoplasms (e.g. mantle cell lymphoma, SLL/CLL, marginal zone lymphoma), some MALT lymphomas | Evaluation of T-cell lymphomas and leukemias. Evaluation of low-grade B-cell lymphomas |
Less specific than UCHL-1 for T cells |
| CD45, Leukocyte common antigen*(LCA, CLA) Note: CLA also refers to a different antigen, HECA-452 | Five or more membrane glycoproteins (membrane, cytoplasm) | Lymphocytes, leukocytes, histiocytes, not plasma cells, erythrocytes, platelets | Non-Hodgkin's lymphomas, some anaplastic (CD30+) large cell lymphomas, Reed-Sternberg cells in LP HD (but not other types) | ID of poorly differentiated neoplasms as lymphomas. However, some anaplastic lymphomas and plasmacytomas may be negative | Preserved in necrotic tissue Best general marker for hematologic neoplasms |
| CD45RA(DPB) | Restricted form of leukocyte common antigen (membrane, cytoplasm) | B cells, monocytes, some T cells | B-cell neoplasms, hairy cells (not specific) | Pan B cell marker that can be used in Zenker's fixed tissue | Not completely specific— other B-cell markers are preferred |
| CD45RO(UCHL-1) | Isoform of CD45 (leukocyte common antigen) (membrane, cytoplasm) | T cells, granulocytes, monocytes | T-cell neoplasms, histiocytic sarcoma | Good pan T cell marker (CD3 is more specific) | |
| CD56*(NCAM) | Neural cell adhesion molecule—cell surface glycoprotein (membrane) | Neurons, astrocytes, Schwann cells, NK cells, subset of activated T cells | Some T/NK cell lymphomas, plasmacytomas Neuroblastoma |
Evaluation of panniculitis- like T-cell lymphoma (both CD56+ and CD56-) and T/NK lymphomas | |
| CD57*(Leu 7, HNK-I) | Lymphocyte antigen that cross-reacts with a myelin-associated glycoprotein (membrane) | T-cell subsets, NK cells, myelinized nerves, neuroendocrine cells, prostate, pancreatic islets, adrenal medulla | Angioimmunoblastic T-cell lymphoma Nerve sheath tumors (occasional), leiomyosar- coma, synovial sarcoma, rhabdomyosarcoma, neuroblastoma, gliomas, neuroendocrine carcinomas, neuro-fibromas, some prostate carcinomas |
ID of T gamma lympho- proliferative disorder (large granular cell lymphocytic leukemia) ID of neuroendocrine differentiation in tumors Evaluation of NK neoplasms |
Not very specific for solid tumors |
| CD61(GPIIIa, platelet glycoprotein Ilia) | Glycoprotein, receptor for fibrinogen, fibronectin, von Willebrand factor, and vitronectin (cytoplasm) | Megakaryocytes, platelets | Megakaryocytic leukemias | ID of megakaryocytic differentiation | |
| CD68*(KPI, CD68-PGMI, Mac-M) | Intracellular glycoprotein associated with lysosomes (cytoplasm, membrane) | Macrophages, monocytes, neutrophils, basophils, large lymphocytes, Kupffer cells, mast cells, osteoclasts | Some lymphomas, histiocytic sarcomas, APML, Langerhans proliferative disorders Neurofibroma, schwannoma, MPNST, granular cell tumors, PEComa, melanomas, atypical fibroxanthoma, RCC |
Best general marker for macrophages, although not specific to this cell type | The antibody PG-M1 does not react with granulocytes |
| CD74(LN2) | Subunit of MHC II-associated invariant chain (membrane) | B cells, monocytes, histiocytes | B-cell neoplasms, hairy cell leukemia, plasma cell lesions | Pan B cell marker | |
| CDw75*(LNI) | Sialylated glycoconjugate present in surface Ig- positive B cells (membrane, mytoplasm) | Mature B cells, T-cell subsets, fetal colon, epithelial cells | Reed-Sternberg cells of LP HD (not other types), follicular lymphomas Colon carcinomas (50%), gastric carcinomas |
Evaluation of HD | |
| CD77(BLA.36, PK antigen) | Globotriaosylceramide, glycolipic membrane from Burkitt's lymphoma cell line (cytoplasm, membrane) | Tonsillar B cells, dendritic reticulum cells, sinuslining cells, macrophages, endothelial cell, epithelial cells | HD, Burkitt's lymphoma, rarely other B- and T-cell lymphomas | Evaluation of RS cells | |
| CD79a(mb-1 protein) | Heterodimer of mb-1 (CD79a) and B29 (CD79b) polypeptides, B-cell antigen receptor (membrane) | B cells, plasma cells | Precursor B-cell ALL, B-cell lymphomas, plasma cell lesions, but not primary effusion lymphoma | Evaluation of B-cell neoplasms (may be the only B-cell marker present) | |
| CD79b* | See above (membrane) | Absent from CLL, hairy cell leukemia | |||
| CD95*(Fas) | Transmembrane glycoprotein member of the nerve growth factor receptor/tumor necrosis factor superfamily—mediates apoptosis (membrane) | Activated T and B cells, epithelial cells | Panniculitis-like T-cell lymphoma (if CD56+) | ||
| CD99*(MIC-2, 12E7, Ewing's sarcoma marker, E2 antigen, HuLy-m6, FMC 29, O13 [different epitope]) |
MIC2 gene product— glycoproteins (p30 and p32) involved in rosette formation with erythrocytes (membrane) Membrane immunoreactivity is more specific than cytoplasmic |
Cortical thymocytes, T lymphocytes, granulosa cells of ovary, pancreatic islet cells, Sertoli cells, some endothelial cells, urothelium, ependymal cells, squamous cells | B- and T-cell precursor lymphoblastic lymphoma/leukemia PNET/Ewing's sarcoma, chondroblastoma, synovial sarcoma, solitary fibrous tumors, GIST, some alveolar rhabdomyosar-comas, desmoplastic small cell tumors, small cell carcinomas, granulosa cell tumors, yolk sac components of germ cell tumors, Sertoli-Leydig cell tumors, atypical fibroxanthoma, meningioma |
Evaluation of lymphoblastic lymphoma/leukemia Thymic carcinomas (lymphocytes +) versus other carcinomas. ID of PNET/Ewing's sarcoma (immunoreactivity should be clearly membranous in the majority of the cells) |
O13 is the most commonly used antibody Immunoreactivity is highly dependent upon the antigen retrieval system used |
| CD103* | Mucosal integrin αEβ7 with specificity for e-cadherin (cytoplasm) | T cells | Enteropathy-type T-cell lymphoma, hairy cell leukemia | Requires frozen tissue or cell suspension | |
| CDI 17*(c-kit, stem cell factor receptor) | Transmembrane tyrosine kinase receptor (ligand is stem cell factor)— apoptosis is inhibited when the ligand is bound (cytoplasm, membrane) | Mast cells, interstitial cells of Cajal (ICC—pacemaker cells of the GI tract found throughout the muscle layers and in the myemteroc plexus), epidermal melanocytes, mononuclear bone marrow cells (4%), Leydig cells, early spermatogenic cells, trophoblast, breast epithelium | GIST (>95%), seminomas (>70%), intratubular germ cell neoplasia, mature teratomas (>70%), some melanomas (focal), mast cell tumors, some carcinomas, some brain tumors, some PNET/Ewing's sarcoma, some angiosarcomas AML (>50%), CML in myeloid blast crisis |
ID of GIST (+) versus leiomyomas (−) and schwannomas (−) ID of seminomas ID of mast cells (mastocytosis) |
Mast cells are an excellent internal control CD117 positivity does not correlate with mutations and/or oncoprotein activity in tumors not known to have activating mutations and is, in general, not of clinical or therapeutic significance in this setting (e.g., to detect tumors likely to respond to therapy directed against the protein, e.g., Gleevec) |
| CDI 38*(Syndecan-1) | Transmembrane heparin sulphate glycoprotein that interacts with extracellular matrix and growth factors (membrane) | Pre-B cells, immature B cells, Ig-producing plasma cells, basolateral surface of epithelial cells, vascular smooth muscle, endothelium, neural cells | Plasma cell lesions, primary effusion lymphoma, plasma cell component of other B cell lymphomas Squamous cell carcinomas, other carcinomas |
ID of plasma cells and their neoplasms Expression may be diminished or lost in poorly differentiated carcinomas |
|
| CD207(Langerin) | Langerhans cell specific C-type lectin (cytoplasm) | Langerhans cells of epidermis and epithelia | Langerhans cell histiocytosis | Induces formation of Birbeck granules | |
| Clusterin*(Apolipoprotein J, complement lysis inhibitor, gp80, SGP-2, SP40, TRPM2, T64, ApoJ) | Multifunctional protein involved in lipid transport, complement regulation, immune regulation, cell adhesion, other functions (membrane, cytoplasm, nucleus) | Many tissues | Anaplastic large cell lymphoma (Golgi pattern) Alzheimer's disease—present in amyloid plaques and cerebrovascular deposits Many types of carcinomas |
||
| Cyclin Dl*(PRAD1, bcl-1) | Cyclin-regulating cyclin- dependent kinases during G1 in the cell cycle, phosphorylates and inactivates the retinoblastoma tumor suppressor protein (nucleus) | Cycling cells (however, lymphocytes usually express only cyclins D2 and D3) | Mantle cell lymphoma Breast cancer (esp. lobular carcinomas and other ER positive carcinomas), esophageal cancer, bladder cancer, lung cancer, HCC, colon carcinoma, pancreatic carcinoma, head and neck squamous cell carcinomas, pituitary tumors, sarcomas Parathyroid adenomas (inversion involving cyclin D1 gene and the parathormone receptor) |
ID of mantle cell lymphoma | Involved in t(11;14)(q13;q32) translocation in mantle cell lymphoma |
| DBA.44*(HCL) | B-cell antigen (cytoplasm, membrane) | Mantle zone B cells, some immunoblasts | Hairy cell leukemia (>95%), B-cell lymphomas (30%) | Evaluation of hairy cell leukemia | |
| Epithelial membrane antigen*(EMA, MUC1, HMFG, DF3, CA 15-3, CA 27.29, PEM, many others) | Episialin, glycoprotein found in human milk fat globule membranes (cytoplasm [more common in malignant cells], membrane [more common in benign cells]) | Epithelial cells, perineurial cells, meningeal cells, plasma cells, usually negative in mesothelial cells, monocytes | Some anaplastic large cell lymphomas (CD30+), plasma cell neoplasms, malignant histiocytosis, erythroleukemia, AML (M4 and M5), LP HD Carcinomas, mesotheliomas, some sarcomas (synovial sarcoma, epithelioid sarcoma), adenomatoid tumor, chordomas, peri-neurioma, neurofibroma, meningiomas, desmoplastic small round cell tumor, Sertoli cell tumor |
ID of epithelial differentiation in tumors—however, keratin is more specific for this purpose. Beware of EMA in some large cell lymphomas Synovial sarcoma typically shows focal positivity |
There are over 50 monoclonal antibodies recognizing different glycosylation patterns in normal tissues and tumors16 |
| Epstein-Barr virus EBV-encoded nonpolyadenylated early RNAs(EBERS) | RNA produced by EBV (nucleus) | EBV-infected B cells | All EBV-related tumors | Most sensitive marker for EBV | Detected by in situ hybridization for RNA on paraffin sections |
| LMP-I* | Latent membrane protein (membrane) | EBV-infected B cells | Nasopharyngeal carcinomas, Reed-Sternberg cells (not LP HD), transplant lymphomas, AIDS-related lymphomas, endemic Burkitt's lymphoma (rare in sporadic cases) | Evaluation of EBV-related neoplasms | |
| EBNA 2(nuclear antigen 2) | Nuclear protein (nucleus) | EBV-infected B cells | Transplant-related lymphomas, AIDS-related lymphomas. Not present in Burkitt's lymphoma or nasopharyngeal carcinomas | Evaluation of transplant- and AIDS-related lymphomas | |
| Fascin | Actin bundling protein regulated by phospho- rylation (cytoplasm) | Interdigitating reticulin cells from the T-cell zones, dendritic cells, reticular network, histiocytes, smooth muscle, endothelium, squamous cells, splenic sinuses | Reed-Sternberg cells (but not in LP HD) High-grade breast carcinomas |
ID of Reed-Sternberg cells in classical HD Fascin positivity has also been reported in ana-plastic large cell lymphoma |
|
| FMC7 | Antigen on subgroups of mature B cells, epitope of CD20 (cytoplasm) | B cells | B-cell lymphomas | Not expressed by CLL | Pan B cell marker Epitope of CD20 but reactivity low in cells with low cholesterol |
| Glycophorin A(GPA) | A glycosylated erythrocyte membrane protein (membrane) | Erythroid elements at all stages | Erythroleukemia | ID of erythroid elements (normal and neoplastic) | |
| Granzyme B* | Neutral serine proteases stored in granules in cytotoxic T cells and in NK cells involved in target cell apoptosis by exocytosis (cytoplasm) | Cytotoxic T cells and NK cells | Some T-cell lymphomas, Reed-Sternberg cells of some cases of EBV- positive HD | ||
| Heavy immunoglobulin chains*(G, A, M, D) | Heavy chain of immunoglobulins (Cytoplasm [plasma cells], membrane [lymphocytes]) | Plasma cells (G>A>M>D) | Plasma cell tumors (monotypic expression of usually G or A), mantle zone lymphomas and WDLL/CLL may co-express M and D, lympho-plasmacytic lymphoma (M) | ID of monoclonal populations of plasma or plasmacytoid cells | |
| HECA-452*(Endothelial cell antigen, cutaneous lymphocyte- associated antigen, CLA) | Cell surface glycoprotein (membrane) | T cells, more common in cutaneous T cells | Mycosis fungoides and other cutaneous T-cell lymphomas | Note: CLA is also used to refer to CD45 | |
| Hemoglobin(Hb) | Hemoglobin (cytoplasm) | Erythroid cells | Some leukemias | Marker for erythroid cells | |
| HHV8* | Latent nuclear antigen of human herpesvirus type 8 (nucleus) | Absent in normal tissue | Primary effusion lymphoma (PEL), AIDS-associated multicentric Castleman's disease Kaposi's sarcoma (endothelial cells and some perivascular cells) |
Evaluation of Kaposi's sarcoma and primary effusion lymphoma | |
| HLA-DR | Major histocompatibility complex Class II gene | B lymphocytes, macrophages, Langerhans cells, dendritic cells, activated T cells, some endothelial and epithelial cells | Leukemic myoblasts | Not very specific for cell type. | |
| Light immunoglobulin chains*(lambda [L], kappa [K]) | Light chain of immunoglobulins (cytoplasm) | Plasma cells (normally K>L), B cells | Plasma cell tumors, B-cell lymphomas | ID of monoclonal populations of plasma c ells and B cells ID of some types of amyloid |
May require frozen tissue for assessment of B lymphoid cells Excellent Ig preservation in plasma cells in B5 or Zenker's fixed tissue |
| Lysozyme(Ly) | Muramidase (cytoplasm) | Circulating monocytes, some tissue macrophages, granulocytes, salivary gland, lacrimal gland, stomach and colon epithelial cells (inflamed or regenerative), apocrine glands, some other epithelial cells | AML with monocytic differentiation, salivary gland tumors, stomach and colon carcinomas. | Marker for histiocytes but not specific. May mark activated phagocytic macrophages Evaluation of myeloid leukemias Strongly positive in monocytoid leukemias |
Not specific for solid tumor identification |
| Mast cell tryptase | Serine protease (cytoplasm) | Mast cells | Mast cell neoplasms | ID of mast cell differentiation | |
| Myeloperoxidase*(MPO) | Enzyme in primary granules of myeloid cells (cytoplasm) | Myeloid cells, monocytes | AML, chloromas | Classification of leukemias | Can be used with tissue fixed in Zenker's fixative |
| Oct2*(Octomer transcription factor) | Transcription factor of the POU homeo-domain family binding to the Ig gene octomer sites regulating B-specific genes (nucleus) | B cells | B-cell lymphomas and leukemias Reed-Sternberg cells in LP HD (but not other types) | Evaluation of HD | Interacts with the transcriptional coactivator BOB.1 BOB.1 and Oct are necessary (but not sufficient) for Ig expression |
| Perform* | Pore-forming protein in cytoplasmic granules of cytotoxic T cells (cytoplasm) | NK cells, large granular lymphocytes, gamma/delta T cells | NK cell lymphomas, anaplastic large cell lymphoma | Evaluation of T-cell lymphomas | |
| TCR*(T-cell antigen receptor, JOVI 1) | Two polypeptide chains (alpha and beta) | Peripheral T cells | Many T-cell lymphomas | Evaluation of T-cell lymphomas | Alpha/beta and gamma/delta T cell receptors can be evaluated in frozen tissue |
| Terminal deoxytransferase*(TdT) | Enzyme that catalyzes addition of nucleotides to ss DNA (nucleus) | Immature T and B cells | Lymphoblastic lymphoma/ALL | Lymphoblastic lymphoma (+) versus Burkitt lymphoma (−) | |
| TIA-I(T-cell intracellular antigen) | A cytolytic granule associated protein expressed in some CD8+ T cells (cytoplasm) | T cells, mast cells, polymorphonuclear leukocytes, eosinophils | Many T-cell lymphomas | Evaluation of T-cell lymphomas | |
| traf-l*(Tumor necrosis factor receptor-associated factor) | Membrane-bound proteins that activate the nuclear factor-(kappa)B (NF-(kappa)B) transcription factor resulting in cell proliferation (cCytoplasm) | Hodgkin's lymphoma | May interact with LMP1 | ||
Abbreviations: AD, Alzheimer's disease;AIDS, acquired immunodeficiency syndrome;ALL, acute lymphocytic leukemia;AML, acute myelogenous leukemia;APML, acute promyelogenous leukemia; BM, basement membrane; CML, chronic myelogenous leukemia; CMV, cytomegalovirus; DFSP, dermatofibrosarcoma protuberans; EBV, Epstein-Barr virus; FISH, fluorescence in situ hybridization; GIST, gastrointestinal stromal tumor; HCC: hepatocellular carcinoma; HD, Hodgkin's disease; HNPCC, hereditary non-polyposis colorectal cancer; ID, identification; KS, Kaposi's sarcoma; LP HD, lymphocyte predominant Hodgkin's disease; MF, mycosis fungoides; MPNST, malignant peripheral nerve sheath tumor; NK, natural killer; PIN, prostatic intraepithelial neoplasia; PNET, primitive neuroectodermal tumor; RCC, renal cell carcinoma; RS, Reed-Sternberg cells;TCC, transitional cell carcinoma.
Notes: NAME: The most common name used to refer to the marker; see also Box 7-1. The name may refer to the antigen, a CD number, or a specific antibody raised to the antigen. In some cases more than one name is commonly used. Antibodies with asterisks appear in the Tables. Most CD numbers correspond to a specific gene product. However, some correspond to antigens from post-translational modifications. For example, CD15 (LeuM1) is a carbohydrate side chain linked to a protein.
ALTERNATE NAME: This list includes abbreviations, antibody names (sometimes recognizing different epitopes), or other terms for the marker.
ANTIGEN: The antigen recognized by the antibody.
LOCATION: The normal location of the antigen. In some cases, only certain locations of the antigen are considered a positive result (e.g., nuclear immunoreactivity for estrogen receptor, membrane immunoreactivity for HER-2/neu).
NORMAL CELLS AND TISSUES: The presence of the marker in normal cells and tissues. These cells serve as important internal positive controls. Abnormal positive immunoreactivity is also an important control for the specificity of the study.
TUMORS: The tumors in which immunoreactivity is typically expected. Refer to the Tables for additional information.
USES: The most common uses for the marker. Different pathologists and institutions often have preferences for the use of certain markers.
COMMENTS: Additional comments regarding the marker.
Additional information on CD antigens can be found at http://www.ncbi.nlm.nih.gov/prow/guide/45277084.htm
Examples of internal controls are:
-
•
S100: Normal nerves, melanocytes and Langerhans' cells in epidermis, cartilage, some myoepithelial cells, skin adnexa
-
•
Estrogen and progesterone receptors: Normal luminal cells in ducts and lobules of the breast
-
•
CD31, FVIII: Vascular endothelium
-
•
c-kit: Mast cells
-
•
Smooth muscle α-actin: Blood vessel walls, myoepithelial cells in the breast
-
•
Vimentin: Blood vessels, stromal cells
-
•
High-molecular-weight keratin: Squamous epithelium
-
•
Low-molecular-weight keratin: Glandular epithelium
-
•
CD15: polymorphonuclear leukocytes.
Negative controls.
The primary antibody is replaced with non-immune animal serum diluted to the same concentration as the primary antibody for a negative control. No positive reaction should be present. If multiple primary antibodies that are reactive with different target antigens are used, then they may serve as negative controls for each other. Although the best negative control would be to use antibody preabsorbed against the target antigen, this is rarely practical in a diagnostic laboratory. Diagnostic slides should also be evaluated for internal negative controls. Aberrant immunoreactivity of tissues that should not be immunoreactive indicates that the immunoreactivity is nonspecific and should not be used for interpretation.
Evaluation of immunoperoxidase studies
The following features must be taken into consideration when evaluating immunoperoxidase studies:
Examples:
Nuclear: ER, TTF-1, P63, Myf4, Ki-67 (MIB-1)
Membranous: EMA, HER2/neu, e-cadherin, EGFR
Cytoplasmic: actin, keratin
Stromal: amyloid (β2-microglobulin, calcitonin, lambda chain)
In rare cases, immunoreactivity in an unusual location is of diagnostic importance:
-
•
TTF-1: Cytoplasmic (instead of nuclear) positivity in hepatocellular carcinomas.
-
•
Ki-67 (MIB-1): Cytoplasmic and membrane (instead of nuclear) positivity in trabecular hyalinizing adenomas of the thyroid and sclerosing hemangiomas of the lung.
-
•
Beta-catenin: Nuclear (instead of cytoplasmic) positivity in solid-pseudopapillary tumors of the pancreas and pancreaticoblastomas. Both nuclear and cytoplasmic positivity is seen in the majority of colon carcinomas. Nuclear positivity is present in approximately 20% of endometrioid endometrial carcinomas.
Identification of immunoreactive cells.
Immunoreactivity of tumor cells must be distinguished from immunoreactivity of normal entrapped cells (e.g., desmin-positive skeletal muscle cells infiltrated by tumor, S100-positive Langerhans' cells in tumors, smooth muscle α-actin-positive blood vessels, etc.). Plasma cells have large amounts of cytoplasmic immunoglobulin and can react nonspecifically with many antibodies.
Intensity of immunoreactivity.
Some weak immunoreactivity may be present as a nonspecific finding. It is important to compare positive cells with control slides and with normally non-immunoreactive cells to determine whether the immunoreactivity is significant.
Number of immunoreactive cells.
In some cases, the number of positive cells may be important as a criterion for positivity or as a prognostic marker (e.g., markers of proliferation such as Ki-67). In other cases, rare weakly positive cells must be distinguished from intermingled normal cells or just nonspecific immunoreactivity.
Criteria for a “positive” result.
Specific criteria for evaluating IHC have been developed for a few antibodies (see Tables 15-3, 15-4, and 7-28 ). However, criteria do not exist for most antibodies or are not universally used by all pathologists. The significance of immunoreactivity varies with the type of lesion, the antibody, and the specific assay. Strong positivity in the majority of cells is easily interpreted as a positive result. As the number of positive cells decreases, and the intensity of immunoreactivity weakens, the lower threshold of a “positive” result becomes more difficult to determine.
Table 7-28.
Scoring of the EGFR (HER 1) test
| SCORE | INTENSITY OF MEMBRANE STAINING | % OF CELLS POSITIVE |
|---|---|---|
| 0 | No staining | 0 |
| 1+ (Positive) | Weak | >1% |
| 2+ (Positive) | Moderate | >1% |
| 3+ (Positive) | Strong | >1% |
The EGFR pharmDx(tm) assay has been approved by the FDA to select patients with colorectal carcinoma for treatment with a monoclonal antibody to EGFR (cetuximab or Erbitux). This test has not been shown to be superior to other comparable tests.
Unlike HER2/neu, the mechanism of overexpression of EGFR does not appear to be gene overexpression.
Immunoreactivity can be membrane or cytoplasmic. Only membrane immunoreactivity is scored, but can be partial or complete.
In clinical trials, 75% to 85% of colorectal carcinomas have been positive (1+ to 3+). Patients with positive results treated with cetuximab alone or in combination with other agents have shown clinical responses (11% to 23%). Patients with carcinomas with scores of 0 for EGFR were not treated.
No correlation has been found between the degree of tumor response and the percentage of EGFR-positive cells or the intensity of staining.
Note: Many normal cells are also positive for EGFR (notably hepatocytes and basal squamous cells).
Time.
Alkaline phosphatase chromogens (red color) fade over time. DAB (brown color) is more permanent. This is not a problem in evaluating current pathology specimens. However, if immunoperoxidase slides are reviewed after a period of time, some chromogens may have faded and once-positive results may appear to be negative.
Location of immunoreactivity (Fig. 7-1 ). Antigens are present in specific sites. Some antigens may be present in more than one location or be extracellular.
Figure 7-1.

Location of immunoreactivity.
Artifacts. Nonspecific positivity should be suspected when immunoreactivity is present in atypical locations:
Background: Suspect nonspecific positivity if normal cells or stroma are positive. This can occur with suboptimal performance of the assay or suboptimal antibodies.
Edge artifact: Antibodies can pool at edges or in holes in tissue. True positivity should also be present in the center of the tissue.
Necrosis or crushing of cells: Nonspecific positivity can be seen in disrupted cells. Although keratin is generally reliable in necrotic tumors, other markers generally should not be interpreted.
Inappropriate location (e.g., cytoplasm instead of nucleus): Occasionally ER or PR is present in the cytoplasm instead of the nucleus. This is not interpreted as a positive result.
Common panels for immunohistochemical studies
Table 7-3, Table 7-4, Table 7-5, Table 7-6, Table 7-7, Table 7-8, Table 7-9, Table 7-10, Table 7-11, Table 7-12, Table 7-13, Table 7-14, Table 7-15, Table 7-16, Table 7-17, Table 7-18, Table 7-19, Table 7-20, Table 7-21, Table 7-22, Table 7-23, Table 7-24, Table 7-25, Table 7-26, Table 7-27, Table 7-28, Table 7-29, Table 7-30 include information from the literature as well as the personal experiences of the staff at Brigham and Women's Hospital. Because of the many differences in specific antibodies, laboratory assays, and criteria for considering a result “positive,” results may vary among institutions. The results have been divided into five categories for general markers and four categories for hematopathology markers (Table 7-2 ). Note that “%” refers to the number of tumors reported to be positive, not the number of cells positive within a tumor.
Table 7-3.
Predominantly CK7+/CK20+
| TUMOR | CK7+ CK20+ | CK7+ CK20– | CK7– CK20+ | CK7– CK20– | 34β E12 | CAM 5.2 | CK 5/6 | EMA | BER- EP4 | CEA m | CEA p | TTF-1 | P63 | WT-1 | S100 | CHRO | HEP | OTHER |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Cholangiocarcinoma | High | Low | Low | neg | High | POS | Low | POS | POS | High | POS | neg | Low | rare | ||||
| Transitional cell carcinoma | POS | Low | neg | neg | Mod | POS | High | POS | Mod | Mod | neg | High | neg | neg | neg | neg | ||
| Pancreas | High | Low | Low | neg | POS | Low | POS | POS | High | POS | neg | Mod | neg | neg | Low | DPC4 lost in 55% | ||
| Ovarian mucinous | POS | Low | neg | neg | POS | neg | POS | Mod | Low | neg | Low | |||||||
| Esophageal adenocarcinoma | POS | neg | neg | neg | neg | Low | neg | Mod | ||||||||||
Table 7-4.
CK7−/CK20+
| TUMOR | CK7+ CK20+ | CK7+ CK20– | CK7– CK20+ | CK7– CK20– | 34 β E12 | CAM 5.2 | CK 5/6 | EMA | BER- EP4 | CEA m | CEA p | TTF-1 | P63 | WT-1 | S100 | CHRO | HEP | OTHER |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Merkel cell carcinoma | Rare | neg | High | Low | High | neg | High | POS | POS | neg | Low | High | neg? | NSE High | ||||
| Colon adenocarcinoma | Low | neg | High | Low | neg | POS | neg | High | POS | POS | POS | neg | Low | neg | Low | neg | neg | CDX2 POS |
Table 7-5.
Predominantly CK7+/CK20−
| TUMOR | CK7+ CK20+ | CK7+ CK20– | CK7– CK20+ | CK7– CK20– | 34 β E12 | CAM 5.2 | CK 5/6 | EMA | BER- EP4 | CEA m | CEA p | TTF-1 | P63 | WT-1 | S100 | CHRO | HEP | OTHER |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Acinic cell carcinoma | neg | POS | neg | neg | POS | POS | Mod | Low | POS | Low | ||||||||
| Adenoid cystic carcinoma | neg | POS | neg | neg | POS | High | POS | Mod | POS | Low | POS | Mod | neg | GFAP Low | ||||
| Breast ductal carcinoma | Low | High | neg | neg | nega | POS | Low | POS | High | High | Mod | neg | Lowa | High | Mod | Low | neg | ER/PRb GCDP Mod |
| Breast lobular carcinoma | Low | POS | neg | neg | POS | neg | POS | Mod | Mod | Mod | neg | Low | Low | neg | ER/PRb GCDP Mod E-cadherin neg |
|||
| Brenner tumor | neg | POS | neg | neg | POS | High | Low | neg? | POS | Calretinin Low NSE POS |
||||||||
| Cervical squamous cell carcinoma | neg | High | neg | Low | POS | neg | POS | POS | POS | Low | neg | POS | neg | neg | HPV POS p16 High |
|||
| Choroid plexus | neg | High | neg | Low | POS | Low | neg | Mod | GFAP High | |||||||||
| Chordoma | neg | POS | neg | neg | Mod | POS | POS | neg | neg | neg | POS | neg | GFAP neg | |||||
| Craniopharyngioma | neg | POS | neg | neg | POS | POS | ||||||||||||
| Embryonal carcinoma | neg | POS | neg | neg | neg | POS | Low | Low | Low | neg? | neg | neg | neg | neg | PLAP High CD30 High |
|||
| Endometrial carcinoma | Low | High | neg | neg | POS | Low | POS | POS | Low | Low | neg | neg? | High | neg | neg | Vimentin POS ER High |
||
| Lung: adenocarcinoma | Low | High | neg | Low | Mod | POS | neg | POS | POS | High | High | High | High? | Low | Low | neg | Low | |
| Lung: BALc non-mucinous | neg | POS | neg | neg | POS | POS | High | High | High | Mod | neg | Mod | neg | |||||
| Meningioma: secretory typed | neg | POS | neg | neg | neg | High | POS | POS | POS | Low | neg | PR Mod ER neg |
||||||
| Mesothelioma | neg | High | neg | Low | High | POS | High | High | neg | neg | neg | neg | neg | High | neg | Low | neg | Calretinin High |
| Mixed tumore | neg | POS | neg | neg | POS | POS | Low | Low | neg? | POS | POS | neg | GFAP High SMA POS Calponin POS |
|||||
| Ovarian: endometrioid | neg | POS | neg | neg | POS | Low | POS | POS | Low | Low | neg? | Low | High | Low | neg | ER Mod | ||
| Ovarian:serous carcinoma | neg | POS | neg | neg | POS | Low | POS | POS | neg | neg | neg? | Low | POS | High | neg | ER High Calretinin Low |
||
| Renal cell: papillary and chromophobe | neg | POS | neg | neg | POS | POS | Modf | |||||||||||
| Thyroid:papillary | neg | POS | neg | neg | POS | POS | Mod | High | neg | Mod | POS | High | High | neg | neg | Thy POS Calci neg |
||
| Thyroid:follicular | neg | POS | neg | neg | neg | neg | Mod | neg | Low | POS | Mod | neg | neg | Thy POS Calci neg |
||||
| Thyroid:medullary | neg | POS | neg | neg | neg | neg | neg | POS | Mod | POS | POS | Thy rare Calci POS |
||||||
p63 may be positive in breast “basal like” carcinomas, some spindle cell metaplastic carcinomas, squamous cell carcinomas, and som e papillary carcinomas. These subtypes may also have less typical keratin subsets such as CK14 (detected by 34β E12), CK17 (detected by MNF-116), or CK5/6.
Most well and moderately differentiated ductal carcinomas, and carcinomas of special type (except for medullary) will be positive for hormone receptors. Poorly differentiated carcinomas, metaplastic carcinomas, and medullary carcinomas are usually negative. Well and moderately differentiated lobular carcinomas are almost always positive for ER, and usually positive for PR. Poorly differentiated lobular carcinomas may be negative for these markers.
Non-mucinous bronchiolo-alveolar carcinomas (BAL) have an immunophenotype similar to lung adenocarcinomas. Mucinous BALs are more likely to be CK20 positive (approximately 70% positive) and less likely to be TTF-1 positive (approximately 30% positive).
Secretory meningiomas are frequently positive for CK7 and CEA, whereas other subtypes are usually negative for CK7 and CEA. The majority of all types of meningiomas are positive for PR (including meningiomas in males).
Mixed tumors (pleomorphic adenomas) occur most frequently in the salivary glands, but can also arise in soft tissues (myoepithelial tumors of soft tissue). These tumors have a similar immunophenotype with keratin (AE1/AE3 77%) or PANK (68%) or EMA (63%) present in the majority of tumors and frequent expression of markers associated with myoepithelial cells (e.g., calponin, GFAP, SMA, S100, p63). However, p63 is seen less frequently (23%) as compared to salivary tumors (100%).
Chromophobe renal cell carcinomas may be positive for WT-1. Other types are negative.
Table 7-6.
Predominantly CK7−/CK20−
| TUMOR | CK7+ CK20+ | CK7+ CK20– | CK7– CK20+ | CK7– CK20– | 34 β E12 | CAM 5.2 | CK 5/6 | EMA | BER- EP4 | CEA m | CEA p | TTF-1 | P63 | WT-1 | S100 | CHRO | HEP | OTHER |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Adrenal cortical adenoma | neg | neg | neg | POS | neg | neg | neg | neg | Low | neg | neg | neg | Low | Melan-A103 POS Inhibin POS |
||||
| Carcinoid | neg | Low | Low | High | neg | POS | Low | Low | Mod | Mod | VARa | neg | VARb | POS | Low | |||
| Epithelioid sarcoma | neg | Low | neg | POS | Mod | High | Low (focal) | POS (focal) | Low (foc) | neg | neg | |||||||
| Esophageal squamous cell carcinoma | neg | Low | neg | High | POS | High? | POS | POS | High? | Low? | Low | neg | POS | neg | neg | neg? | ||
| Seminoma | neg | Low | neg | High | neg | Low | Low | neg | neg | neg | neg | neg | neg | PLAP POS CD117 POS |
||||
| Head and neck squamous cell carcinoma | neg | Low | Low | High | POS | neg | POS | POS | neg | neg | POS | neg | neg | |||||
| Hepatocellular carcinoma | Low | Low | neg | High | Low | POS | neg | Low | Low | neg | Highc | Highd (cyt) | Low | neg | neg | High | AFP Mod | |
| Lung:squamous cell carcinoma | neg | Low | Low | High | POS | High | POS | Mod | Low | neg | POS | neg | Low | |||||
| Lung:small cell carcinoma | neg | Low | neg | High | neg | High | neg | POS | POS | Mod | High | POS | rare | neg? | Mod | neg | ||
| Pheo/paraganglioma | Rare | Rare | Rare | POS | neg | neg | neg | neg | neg | High | POS | Inhibin neg Melan-A103 rare |
||||||
| Prostatic carcinoma | neg | neg | Low | High | neg | POS | neg | Low | POS | neg | Mod | neg | neg | neg | neg | Low | neg | PSA POS |
| Renal cell carcinoma: clear cell | neg | Low | neg | High | neg | High | neg | POS | Low | Low | neg | neg | Low | neg? | Low | neg | neg | Vime POS |
| Squamous cell carcinoma | neg | Low | High | POS | Low | POS | POS | neg | Mod | Low | Low | POS | neg | neg | neg | |||
| Thymic carcinoma | POS | POS | POS | Mod | High | Low | neg? | neg | POS | neg | neg | Low | CD5 Mod | |||||
| Thymoma | neg | Low | neg | High | High | High | Mod | Low | neg? | neg | POS | neg | neg | neg? | neg? | CD5 neg | ||
Non-pulmonary carcinoid tumors are negative for TTF-1. Some pulmonary carcinoids may be positive.
Sustentacular cells may be positive for S100 and positivity can vary with site.
CEA has a canalicular pattern in hepatocellular carcinoma, a diffuse cytoplasmic pattern in other carcinomas.
TTF-1 immunoreactivity in hepatocellular carcinoma is cytoplasmic (not nuclear as in lung and thyroid carcinomas). Positivity can vary with the antibody used to detect TTF-1.
Table 7-7.
No dominant CK7/CK20 pattern or pattern unknown
| TUMOR | CK7+ CK20+ | CK7+ CK20– | CK7– CK20+ | CK7– CK20– | 34 β E12 | CAM 5.2 | CK 5/6 | EMA | BER- EP4 | CEA m | CEA p | TTF-1 | P63 | WT-1 | S100 | CHRO | HEP | OTHER |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Gastric adenocarcinoma | Low | Low | Low | Low | neg | POS | neg | High | POS | High | High | neg | Low | neg? | Low | neg | Low | |
| Ameloblastoma/Adamantinomaa | POS | neg | neg | neg? | neg? | |||||||||||||
| Lymphoepithelial carcinomab | POS | High | Mod? | Mod? | POS | negc | ||||||||||||
Approximately 15% of ameloblastomas are positive for CK7.
Approximately 50% of nasopharyngeal carcinomas are positive for CK7. Many cases in Asian and North African patients (less commonly in US patients) are associated with EBV. EBV can be demonstrated by in situ hybridization, PCR, or occasionally by immunohistochemistry. These carcinomas are also positive for broad-spectrum keratins (AE1/AE3 and PANK).
S100-positive dendritic cells are present.
Table 7-8.
Spindle-cell/soft-tissue lesions and sarcomas
| AE1/AE3 | CAM 5.2 | EMA | S100 | HMB-45 | HHF-35 | SMA | DESMIN | H-CALDESMON | CD34 | CD31 | FVIII | c-kit | CD99 | OTHER | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Neural | |||||||||||||||
| Perineurioma | neg | neg | POS | Low | neg | Mod | Low | neg | neg | neg | neg | neg | Mod | CLAUD-1 POSa | |
| Neurofibroma | neg | neg | POS b | POS | neg | neg | neg | neg | High | neg | neg | ||||
| MPNST | Low | Low | Low | Mod | neg | Low | Low | neg | neg | Low | neg | GFAP Mod | |||
| Schwannoma | Low | neg | Negc | POS | neg | neg | neg | neg | Mod | neg | neg | neg | CD68 POS | ||
| Granular cell tumord | neg | neg | neg | POS | neg | neg | neg | neg | neg | CD68 POS Calretinin POS Inhibin POS |
|||||
| Melanoma | rare | rare | neg | POS | Highe | neg | neg | neg | neg | neg | neg | Mod | Low | Melan-A High FLI-1 neg |
|
| Clear cell sarcoma | neg | neg | neg | High | POS | Low | neg | neg | neg | neg | neg | Low | Low | Melan-A Mod | |
| PEComaf | neg | neg | neg | Low | POS | POS | POS | High | Mod | Low | neg | neg | VARg | Melan-A POS | |
| Gastrointestinal stromal tumor | neg | neg | neg | Low | Mod | Low | neg | High | High | neg | POS | POS | |||
| Muscle | |||||||||||||||
| Rhabdomyosarcoma | Low | Low | Low | neg | neg | High | Mod | High | neg | Low | neg | neg | neg | Low | Myf4 POS WT-1 Mod FLI-1 neg |
| Glomus tumor | neg | neg | neg | neg | neg | POS | POS | Low | High | Low | neg | neg | neg | ||
| Leiomyoma or leiomyosarcoma | Low | Low | Mod | neg | neg | POS | POS | High | POS | Low | neg | neg | neg | Low | ER/PR High CD10 Low |
| Endometrial stromal sarcoma | Mod (focal) | Low (focal) | neg | High | Mod | neg | neg | neg | ER/PR High CD10 High | ||||||
| Vascular | |||||||||||||||
| Angiosarcoma | Lowh | Lowh | rare | neg | neg | Low | Low | neg | High | High | High | Low | FLI-1 POS | ||
| Kaposi's sarcoma | neg | neg | neg | neg | neg | POS | neg | POS | neg | Mod | neg | FLI-1 POS | |||
| Epithelioid hemangioen- dothelioma | High | neg | neg | neg | neg | neg | Low | neg | High | High | POS | neg | FLI-1 POS | ||
| “Fibrous” | neg | Low | neg | neg | neg | neg | neg | ||||||||
| Fibrosarcoma | neg | Low | |||||||||||||
| Solitary fibrous tumor | neg | neg | Low | neg | neg | Low | neg | neg | POS | neg | neg | neg | High | ||
| DFSP | neg | neg | neg | neg | neg | High | Low | neg | neg | POS | neg | neg | neg | ||
| Dermatofibroma | neg | neg | neg | neg | High | High | neg i | neg | neg | neg | |||||
| Fibromatosis | neg | neg | neg | Mod | High | High | Mod | neg | neg | neg | neg | ER Low | |||
| Postoperative spindle cell nodule | Mod | Mod | Low | neg | High | High | Mod | neg | neg | neg | |||||
| Myofibroblastic tumors | neg | neg | neg | POS | High | Mod | neg | Mod | neg | ER High PR POS |
|||||
| Atypical fibroxanthoma | neg | neg | neg | neg | neg | Low | neg | Low | CD68 Mod | ||||||
| Other | |||||||||||||||
| Osteosarcoma | neg | neg | Low | Low | Mod | High | neg | neg | Low | ||||||
| Chondrosarcoma | neg | neg | Low | POS | neg | neg | neg | neg | neg | Low | |||||
| Chondroblastoma | neg | neg | neg | POS | Mod | Low | neg | neg? | POS | ||||||
| Mesenchymal chondrosarcoma | neg | neg | neg | POS | neg | rare | Low | POS | My4 neg | ||||||
| Extraskeletal myxoid chondrosarcoma | neg | neg | Low | Low | neg | neg | neg | Low | Low | neg | |||||
| Alveolar soft part sarcoma | neg | neg | Low | neg | Low | Low | Low | Low | neg | neg | Low | myoD1 neg myogenin neg TFE3 POSj |
|||
| Epithelioid sarcoma | POS | POS | POS | neg | neg | Low | Low | neg | Mod | neg | neg | neg | Low | FLI-1 neg | |
| Synovial sarcomak | High | High | High | Mod | neg | neg | Low | neg | neg | neg | neg | neg | neg | High | WT-1 neg |
| Adenomatoid tumor | POS | POS | POS | neg | neg | neg | Ber-EP4 High Calretinin POS |
||||||||
| Mesothelioma sarcomatoid typem | High | POS | Low | POS | High | Low | neg | Low | WT-1n Calretinin Low |
||||||
| Meningioma | nego | nego | High | Low | neg | Low | Low | neg | Low | neg | neg | POS | ER neg PR POS PANK Low |
||
| Carcinoma:spindle cellp | VAR | VAR | VAR | VAR | neg | rare | rare | neg | neg | neg | neg | ||||
lClaudin-1 is positive in glandular areas of synovial sarcoma but less so in spindle cell areas.
Some claudin-1-positive perineurial cells can be present in neurofibromas and schwannomas.
Perineural cells are positive for EMA in neurofibromas.
EMA may be positive in capsule and perineural cells of schwannomas.
Congenital granular cell tumors are positive for CD68 but negative for S100 and NSE.
HMB-45 is less frequently present in spindle cell melanomas and usually negative in classic desmoplastic melanomas. Other markers for melanoma are also less frequently positive in these subsets.
PEComas (perivascular epithelioid cell tumors) include angiomyolipoma, lymphangioleiomyomatosis, clear cell sugar tumor of the lung, clear cell myomelanocytic tumor of ligamentum teres/falciform ligament, and abdominopelvic sarcoma of perivascular epithelioid cells.
Results in the literature are conflicting. Angiomyolipomas are likely not positive for CD117.
Keratin positivity in angiosarcomas is more common in epithelioid types.
Cellular dermatofibroma may show focal desmin immunoreactivity.
Alveolar soft-part sarcomas are characterized by a translocation that fuses the TFE3 transcription factor gene at Xp11 to a novel gene at 17q25 called ASPL. These sarcomas demonstrate nuclear immunoreactivity for TFE3 (as do rare pediatric renal tumors with the same translocation) and this immunoreactivity is not present in other tumors or normal tissues. The characteristic cytoplasmic crystals are composed of monocarboxylate transporter 1 (MCT1) and its chaperone CD147. However, these proteins are found in many other cell types and are not specific for this tumor.
Keratin and EMA positivity are usually only focal in monophasic synovial sarcomas.
The immunohistochemical pattern for epithelioid mesotheliomas is given in Table 7-30.
WT-1 may be positive in a minor epithelioid component of sarcomatoid mesotheliomas, but is generally negative in the spindle cells.
Secretory meningiomas are typically cytokeratin 7 positive (CK20 negative) and also positive for CEA. Other subtypes are generally negative for keratin. However, malignant meningiomas may be positive for keratin.
Squamous cell carcinomas with a spindle cell morphology are generally strongly positive for AE1/AE3 (less commonly for CAM 5.2), EMA, and p63. Spindle cell carcinomas of the breast often express markers expressed by myoepithelial cells such as “basal keratins” (including cytokeratin 14 which is included in the group detected by PANK or MNF-116), smooth muscle α-actin, and p63. Poorly differentiated carcinomas with spindle cell morphology may only show focal positivity for keratins and EMA.
Table 7-9.
Small blue cell tumors
| TUMOR | PANK | CAM 5.2 | CK20 | EMA | S100 | HMB- 45 | NSE | SYN | CHRO | CD99 | SMA | HHF 35 | DES MIN | MYF-4 | LCA | NFP | WT-1a | PASb |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Melanoma | rare | rare | neg | neg | POS | Highc | High | Low | neg | Low | Low | neg | neg | neg | neg | |||
| Esthesioneuroblastoma | Low | Mod | Low | POS | POS | High | Mod | Low | neg | neg? | neg | Mod | ||||||
| Neuroblastoma | neg | neg | neg | Low | Mod | neg | POS | High | High | neg | neg | neg | neg | neg | High | Low | neg | |
| Small cell carcinomad | POS | Mod | neg | POS | neg | neg | High | Mod | Mod | Low | neg | neg | neg | neg | ||||
| Merkel cell carcinomae | POS | POS | POS | POS | neg | neg | High | Mod | High | Low | neg? | neg | Mod | neg | ||||
| Desmoplastic small round cell tumor | POS | POS | neg | POS | Low | neg | High | Low | Low | Mod | Low | Low | POS | neg | neg | POS | POS | |
| Ewing's sarcoma (PNET) | Low | Low | Low | Low | neg | Mod | Low | neg | POSf | neg | LOW | neg | neg | LOW | neg | POS | ||
| Medulloblastoma | neg | neg? | Low | POS | POS | Low | Low | neg | ||||||||||
| Rhabdomyosarcoma | neg | Mod | neg | Low | neg | Mod | neg | neg | Low | Low | POS | POS | POS | neg | Low | Mod | POS | |
| AML | neg | neg | neg | neg | Low | neg | POS? | Mod | High | |||||||||
| Lymphoma | neg | neg | neg | neg | neg | neg | neg | neg | neg | Var | neg | neg | neg | neg | POS | negg | ||
Ewing's sarcoma (PNET), desmoplastic small round cell tumor, rhabdomyosarcoma, neuroblastoma, and ic cytogenetic changes (see Table 7-33). medulloblastoma have EM has some advantages over immunohistochemistry in the evaluation of childhood small round blue cell tumors.11 Initial panel. Keratin, S100, LCA.
Additional studies may be helpful depending on the histologic appearance and the results of the initial studies.
Polyclonal WT-1.
PAS is a histochemical stain for glycogen. A PAS-D stain confirms the presence of glycogen by treatment of the tissue with diasta se, which digests the glycogen and eliminates the positivity. Although used for these tumors in the past, these studies are currently not usually performed.
MART-1 is also frequently positive in melanomas.
Small cell carcinomas of the lung are positive for TTF-1.
Merkel cell carcinomas demonstrate a dot-like perinuclear pattern for most markers.
Significant immunoreactivity is a membrane pattern in the majority of the cells.
Some plasma cell lymphomas may be positive.
Table 7-10.
Myoepithelial markers in breast carcinoma
| MARKER | LOCATION | NORMAL LUMINAL CELLS | MYOEPITHELIAL CELLS | BLOOD VESSELS | MYOFIBROBLASTS | CARCINOMASa | COMMENT |
|---|---|---|---|---|---|---|---|
| p63 | Nucleus | neg | POS | neg | neg | rare | Only nuclear marker Clean background |
| SMA | Cytoplasm | neg | POS | POS | POS | rare | Positive in most myoepithelial cells |
| CD10 | Membrane | neg | POS | neg | POS | rare | |
| SMM-HC | Cytoplasm | neg | POS | POS | High | rare | |
| Calponin | Cytoplasm | neg | POS | POS | Mod | rare | |
-
•Invasive carcinoma versus sclerosing adenosis (frequently involved by DCIS, LCIS, or apocrine metaplasia).
-
•DCIS versus DCIS with microinvasion. Double immunolabeling with p63 (brown nucleus) and cytokeratin (AE1/AE3—red cytoplasm) can be useful to highlight small nests of tumor cells lacking myoepithelial cells.
-
•DCIS versus carcinoma invading as circumscribed tumor nests versus lymphovascular invasion.
S100 protein and cytokeratins (e.g.34β E12) are not recommended for this purpose, as fewer myoepithelial cells are positive and luminal cells are also sometimes positive. p63 is a good general marker for myoepithelial cells and is particularly helpful in cases with prominent myofibroblasts (e.g., sc lerosing lesions) or with blood vessels closely apposed to tumor cells (e.g., papillary fronds in papillary DCIS). In some cases, SMA may be positive in more cells than p63.
Rare carcinomas with myoepithelial features (adenoid cystic carcinomas, some spindle cell carcinomas, some basal-like carcinomas, some carcinomas associated with BRCA1 mutations) can show focal to diffuse positivity for myoepithelial markers.
Table 7-11.
Epidermal lesions of the nipple
| AE1/AE3 | CAM 5.2 OR CK7 | CK20 | EMA | S100 | HMB-45 | GCDFP-15 | CEA p | CEA m | HER2 | ER OR PR | MUCICARMINE STAIN | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Paget's disease of the nipple | POS | POS | neg | POS | Mod | neg | Mod | Mod | Low | POS | Low | High |
| Squamous cell carcinoma | POS | Low | Low | POS | Low | neg | neg | Low | Mod | Low | neg | neg |
| Melanoma | Low | Low | neg | Low | POS | POS | neg | Mod | neg | neg | neg | neg |
Most cases of Paget's disease of the nipple are associated with nd approximately DCIS deeper in the half will also have areas of invasion. Rare cases may be difficult to interpret due to the absence of associated disease in the b reast or if the initial biopsy is shallow. In some cases, Paget cells may take up melanin and may be difficult to distinguish from melanoma. Toker cells in nipple epidermis are CAM 5.2 and CK7 positive but negative for HER2/neu.
Initial panel. CAM 5.2 (or CK7), HER2, and S100 with additional antibody studies based on these findings, if necessary.
Cases of extramammary Paget's disease are more likely to be CK20 positive and less likely to be positive for HER2/neu (less than 40%).
Table 7-12.
Endocervical carcinoma versus endometrial carcinoma
| CK7 | CK20 | VIM | CEA m | CEA p | P16 | HPV (IN SITU) | ER | PR | |
|---|---|---|---|---|---|---|---|---|---|
| Endocervical carcinoma | POS | rare | rare | POS | High | POS (difffuse, strong) | High | Low (focal) | Low |
| Endometrial carcinoma | POS | rare | POS | Lowa | Mod | Low (patchy, weak) | neg | High (diffuse) | High |
27% of cases have some positivity but primarily in squamous areas and only focally in glandular areas.
Table 7-13.
Endometrial stromal sarcoma versus leiomyosarcoma
| CD10 | DESMIN | H-CALDESMON | ER/PR | |
|---|---|---|---|---|
| Endometrial stromal sarcoma | High | Mod | neg | High |
| Leiomyosarcoma | Low | High | POS | High |
Table 7-14.
Primary ovarian carcinoma versus metastatic carcinomas
| CK7 | CK20 | DPC4 (SMAD4) | CDX2 | ER | CEA m | CEA p | |
|---|---|---|---|---|---|---|---|
| Endometrioid ovarian carcinoma | POS | neg | Low | Mod | Mod | Low | |
| Clear cell ovarian carcinoma | POS | neg | Mod? | Mod | neg | ||
| Mucinous ovarian carcinoma | POS (diffuse) | High (patchy) | POS | Mod | Low | Mod | Low |
| Mucinous breast carcinoma | POS | Low | POS | nega | POS | Mod | Low |
| Pancreatic carcinoma | POS | High | Mod | Mod | neg | High | POS |
| Appendiceal carcinoma | Low (patchy) | POS | POS | High | Low | ||
| Mucinous colon carcinoma | Low (patchy) | POS (diffuse) | High | POS | neg | POS | POS |
Breast cancers, in general, are negative for CDX2. Results for mucinous breast carcinomas have not been reported.
Table 7-15.
Ovarian carcinoma versus mesothelioma
| CK7 | CK20 | CK5/6 | CEA m | CEA p | CD15 (LEUM1) | ER | WT-1 | CALRET | BER-EP4 | |
|---|---|---|---|---|---|---|---|---|---|---|
| Peritoneal mesothelioma | High | neg | POS | neg | neg | rare | rare | High | High | neg |
| Ovarian serous carcinoma | POS | Low | neg | neg | neg | Mod | POS | POS | Low | POS |
| Ovarian endometrioid carcinoma | POS | neg | Low | Mod | Low | High | High | Low | POS | |
| Ovarian mucinous carcinoma | POS | High | neg | Mod | Low | Low | neg | Low | ||
Table 7-16.
Trophoblastic lesions
| KERATIN | ALPHA- INHIBIN | HPLa | HCGa | CD146 (MELCAM) | KI-67b | P57c | DNA PLOIDYd | |
|---|---|---|---|---|---|---|---|---|
| Choriocarcinoma | POS | POS | Weak (focal) | Strong (diffuse) | POS | 69% | ||
| Placental site trophoblastic tumor | POS | POS | Mod (greater than hCG) | Focal (less than HhPL) | POS | >14% | ||
| Epithelioid trophoblastic tumor | POS | POS | Focal | Focal | Focal | >14% | ||
| Placental site nodule | POS | POS | Weak (focal) | Focal | Focal | <1% | ||
| Exaggerated placental site | POS (diffuse) | Focal | POS | 0% | ||||
| Partial mole | POS | POS | Weake (diffuse) | Weak (diffuse) | POS | Triploid | ||
| Complete mole | POS | POS | Weake (focal) | Strong (diffuse) | raref | Diploid (paternal) | ||
| Hydropic fetus | POS | POS | POS | Diploid (60%) Triploid (40%) |
||||
Cytokeratin and alpha-inhibin (present in syncytiotrophoblastic cells and some intermediate trophoblastic cells) are not useful for the differential diagnosis of these lesions, but may be helpful if other types of tumors are in the differential diagnosis.
Evaluated in syncytiotrophoblast.
Implantation-site intermediate trophoblastic cells are evaluated for the number of Ki-67-positive cells. CD146 can be used to help identify these cells using a double label technique. Lymphocytes can also be positive for Ki-67 and should not be counted.
p57 is a paternally imprinted gene, expressed from the maternal gene, which shows decreased expression in complete moles, whose DNA is completely derived from paternal DNA.
Ploidy is usually determined by flow cytometry.
Increases with advancing pregnancy.
In complete moles, p57 positivity is present in villous stromal cells and extravillous trophoblast but absent in intermediate trophoblast lining the villi.
Table 7-17.
Tumors of germ cells and sex-cord stromal tumors
| CK7 | CK20 | AE1/AE3 | CAM5.2 | NSE | EMA | PLAPa (mem) | AFP | CD30 (Ki-1, Ber-H2) | CD117 (c-kit) | VIM | hCG | HPL | INHIBIN | MELAN- A103 | OTHER | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Seminoma | Mod | neg | Mod | Lowb | High | neg | POS | neg | Low | POS | Mod | Lowc | neg | neg | neg | |
| Embryonal carcinoma | High | neg | POS | POS | High | Low | High | Low | Highd | neg | Low | Low | neg | neg | neg | |
| Yolk sac tumor | POS | POS | High | neg | Mod | High | Low | Low | neg | neg | neg | neg | ||||
| Choriocarcinoma | POS | POS | Mod | Mod | Mod | neg | neg | neg | POS | POS | POS | neg | ||||
| Intratubular germ cell neoplasia | neg | POS | POS | neg | ||||||||||||
| Spermatocytic seminoma | Mod (focal) | neg | Mod | |||||||||||||
| Leydig cell tumor | Mod | Mod | Mod | Mod | Low | Low | neg | POS | POS | High | ||||||
| Granulosa cell tumor | Mod | Low | Low | Mod | Low | neg | neg | neg | neg | POS | POS | High | WT-1 High HHF35 High S100 Mod |
|||
| Sertoli cell tumor | Mod | Mod | POS | neg | High | POS | ||||||||||
PLAP is expressed in embryonic germ cells, but not in normal spermatogonia, spermatocytes, and spermatids.
CAM5.2 is present as a strong dot-like paranuclear positivity.80% of mediastinal seminomas are positive for CAM5.2 compared to 20% to 30% of testicular seminomas.
hCG may be positive in trophoblasts in seminomas.
Only 35% of embryonal carcinomas metastatic to lymph nodes after chemotherapy are positive for CD30.
Table 7-18.
Adrenal and kidney tumors
| AE1/AE3 | CK7 | CK20 | PANK | CAM5.2 | MUC-1 (EMA) | S100 | CHROM | SYN | MELAN- A103a | INHIBIN | NSE | NFP | AMACR | VIM | OTHER | IRON STAIN | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Adrenal tumorsb | |||||||||||||||||
| Cortical adenoma | neg | neg | neg | Low | Low | neg | neg | neg | POS | POS | High | High | neg | neg | High | TTF-1 neg CD10 neg |
|
| Cortical carcinoma | neg | neg | neg | High | POS | POS | neg | ||||||||||
| Pheo/paraganglioma | neg | neg | neg | neg | neg | Highc | POS | POS | neg | neg | POS | POS | Mod | GFAP mod | |||
| Kidney tumors | |||||||||||||||||
| Renal cell carcinoma: clear cell | High | Low | neg | High | High | High (diff) | Low | neg | neg | neg | neg | Mod | neg | High | p63 neg TTF-1 neg GFAP low RCC POS CD10 POS |
Focal, course | |
| Papillary | POS | High | Low | POS | Mod (mem) | POS | RCC POS CD10 POS |
Focal, coarse | |||||||||
| Chromophobe | High | High | neg | POS | POS (mem) | neg | neg | RCC Mod CD10 neg |
Diff, strong | ||||||||
| Oncocytomad | Mod | High | neg | POS | RCC neg CD10 low |
Focal, weak | |||||||||||
| Transitional cell carcinoma | Mod | POS | High | POS | POS | POS | neg | neg | neg | neg | Low | Low | Low | p63 POS CD10 mod |
|||
Renal cell carcinoma subtypes have typical cytogenetic abnormalities (see Table 7-33).
CD117 (c-kit) has been reported to be positive in almost all papillary renal cell carcinomas (cytoplasmic) and chromophobe carcinomas (membrane) but is not present in clear cell carcinomas.
Mutations in c-kit were only found in papillary carcinomas. diff, diffuse positivity; mem, positivity located on membrane.
Positivity is also present with MART-1.
Clear cell renal cell carcinoma (RCC) metastatic to the adrenal can sometimes be confused with an adrenal cortical tumor (thus, the older term for clear cell carcinoma of “hypernephroma”). RCC has clear cytoplasm (compared to the bubbly cytoplasm of the adrenal cortex) and blood lakes are typically present. Glycogen is present in RCC and absent in adrenal lesions (demonstrated by PAS with and without diastase). Cytokeratin and EMA are useful IHC markers.
Positivity is present in sustentacular cells. These cells may be absent in malignant tumors.
50% of oncocytomas have a punctate/dot-like pattern for CK8 or CK18 which is not seen in RCC. EM may be helpful to distinguish oncocytoma from chromophobe RCC (see Table 7-32).
Table 7-19.
Tumors of bladder, prostatic, or renal origin
| CK7 | CK20 | KERATIN HMW | PSA | PAP | AMACR | CEA m | CEA p | P63 | CA125 | MUCI | |
|---|---|---|---|---|---|---|---|---|---|---|---|
| Prostatic carcinoma | Low | Low | neg | High | POS | POS | neg | Mod | neg | neg | neg |
| Transitional cell carcinoma | POS | High | Mod | neg | neg | Low | Mod | Mod | High | neg | neg |
| Bladder adenocarcinoma | High | High | neg | neg | neg | Mod | High | Low | POS | ||
| Renal cell carcinoma: clear cell | Low | neg | neg | neg | Low | neg | Low | neg | neg | ||
| Rectal adenocarcinoma | Low | POS | neg | neg | neg | POS | POS | neg | POS | ||
| Seminal vesicle carcinoma | High | neg | neg | neg | VAR | POS | High | ||||
Table 7-20.
Prostate carcinoma versus other lesions
| 34β E12 (BASAL CELLS) | P63 (BASAL CELLS) | AMACR (504S) (GLANDULAR CELLS) | |
|---|---|---|---|
| Benign glands | POS | POS | neg |
| PIN | POS | POS | High |
| Invasive carcinoma | neg | neg | POS |
Antibody cocktails. These antibodies can be combined to facilitate the evaluation of small lesions:
34β E12 + p63 labels a greater number of basal cells than either marker alone AMACR + p63 and/or 34β E12 facilitates the identification of small foci of invasive carcinoma.
Table 7-21.
Hepatic tumors
| CK7 | CK20 | AE1/AE3 | CAM 5.2 | KERATIN HMW | CEA m | CEA p | TTF-1 | HEP | AFP | CD10 | CHROM | MUCI | BILE | CIRRHOSIS | HBV | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Hepatocellular carcinoma (HCC) | Low | neg | Low | POS | neg | neg | Higha | Highb (cyt) | High | Mod | Higha | neg | rare | May be present | 65–90% | 50% |
| Hepatoblastoma | Low | POS | Low | Higha | POS | High | Low | absent | rare | |||||||
| HCC:fibrolamellar | Mod ? | neg | POS a | POS | neg ? | neg | May be present | absent | rare | |||||||
| Cholangiocarcinoma | POS | Mod | POS | POS | High | High | POS | neg | neg | neg | neg | 75–100 | neg | rare | rare | |
| Metastatic carcinoid tumor | Low | Low | High | POS | Mod | Mod | Lowc (nuc) | neg | Low | POS | neg | absent | absent | |||
Sinusoids of HCC show diffuse CD34 positivity in 80% to 90% of cases, but this is not seen in normal liver. CD34 positivity can also be seen in focal nodular hyperplasia. Metastatic carcinomas can show diffuse positivity in 20% of cases, but the positive endothelial cells are present throughout the tumor and the cells do not surround nests of tumor cells, as is seen in HCC.
Reticulin stains can be helpful in the evaluation of fine needle aspirates or core needle biopsies of liver lesions. HCC has an a bnormal pattern of absent, decreased, or expanded trabecula, whereas benign lesions show a normal trabecular pattern.
Metastatic carcinomas can usually be distinguished from HCC by frequent expression of CK7, only rare expression of HepPar1, the absence of a bile canalicular pattern for CEA p and CD10, and the absence of cytoplasmic positivity for TTF-1.
Metastatic carcinomas to the liver often cannot be reliably distinguished from cholangiocarcinomas by histologic appearance or immunohistochemical pattern, with the exception of colorectal carcinomas. If the patient has a known primary carcinoma, it is most helpful to compare the two tumors.
cyt = cytoplasmic immunoreactivity; nuc = nuclear immunoreactivity.
Bile canalicular pattern. Other carcinomas have a membrane or cytoplasmic pattern.
TTF-1 is seen in the cytoplasm (unlike the nuclear pattern seen in lung and thyroid carcinomas).
Carcinoids arising at sites other than lung are very unlikely to be positive for TTF-1. Lung carcinoids may be positive and are more likely to express CK7.
Table 7-22.
Thyroid and parathyroid lesions
| KER HMW | CK19 | HBMEa | GALEC TIN-3 | CALCITONIN | SYN | CHRO | RET | P27 | PPAR GAMMA | THY | TTF-1 | S100b | CEA M | CEA P | CD57 | RB PRO TEIN | VIM | OTHER | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Thyroid lesions | |||||||||||||||||||
| Hyperplastic nodule | Mod | Low | Low | neg | POS | neg | POS | POS | Low | POS | |||||||||
| Follicular adenoma | neg | Mod | Low | Low | neg | Mod | neg | neg | POS | Low 10% | POS | POS | Low | Low | POS | POS | |||
| Follicular carcinoma | neg | Mod | Mod | Low | neg | High | neg | neg | POS | Low 30% | POS | POS | Mod | neg | Low | Mod | neg? | POS | |
| Papillary carcinoma: follicular variant | POS | POS | Mod | neg | Low | Mod | Low 10% | POS | POS | High | neg | ||||||||
| Papillary carcinoma | POS | POS | High | POS | neg | High | neg | Low | POS | Low 10% | POS | POS | High | neg | Mod | POS | neg | POS | p63 POS |
| Medullary carcinoma | neg | Mod | POS | POS | POS | neg | Low | POS | POS | Mod | Mod | Mod | Highc | ||||||
| Anaplastic carcinomad | Mod | rare | rare | ||||||||||||||||
| Parathyroid lesions | |||||||||||||||||||
| Parathyroid adenomas and carcinomas | Low | Low | POS | Highe | neg | neg | neg | Low | POS | neg/weak | PTh POS RCC POS Cyclin D1 POS |
||||||||
Thyroid adenomas, follicular carcinomas, papillary carcinomas, and medullary carcinomas are CK7 positive and CK20 negative. Variable immunoreactivity has been reported for CK7 in anaplastic carcinomas.
Metastatic carcinomas to the thyroid will be negative for thyroglobulin, TTF-1 (except for lung carcinomas), and calcitonin.
DDIT3 and ARG2 are new markers that may prove helpful for distinguishing follicular carcinoma (approximately 70% to 80% positive) from adenoma (90% negative).
Tumors with Hürthle cell changes may be negative for HBME.
Hürthle cells (both benign and neoplastic) are positive for S100 (nuclear and cytoplasmic).
Spindle cells may be positive for vimentin.
Anaplastic thyroid carcinomas are frequently negative for TTF-1, thyroglobulin, and CK20.
p27 is low in parathyroid carcinomas.
Table 7-23.
B-cell neoplasms
| B cell markers | |||||||||||||||||
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| CD4S LCA | CDI9 B4 | CD20 L26 | CD22 | CD79a | slg | clg | CDS Leu 1 | CDIO CALLA | CD23 | CD43 Leu 22 | CD34 | bcl-2 | bcl-6 | CDI 38 SYNDECAN | CYCLIN Dl | OTHER | |
| Precursor lymphoblastic lymphoma/leukemia | +/− | + | +/− | +/− | + cyt | +M | +a | +/− | +/− | TdT + CD99 + |
|||||||
| Small lymphocytic lymphoma/CLL | + | + | + wk | + wk | + | +M/D wk | −/+ | + | + | +/− | + | CD11c+ wk CD79b -FMC7 - |
|||||
| Mantle cell lymphoma | + | + | + | + | + | +M/D | − | + | − | − | + | − | + | − | − | + | CyclinD + FMC + |
| Marginal zone lymphoma (MALT) | + | + | + | + | + | + | +/− | − | − | − | +/− | − | + | − | −/+b | − | CD11c +/− CD21 + CD3S+ |
| Follicular lymphoma | + | + | + | + | + | +M | − | − | + | −/+ | −/+ | − | +/− | + | − | − | CDw7S + |
| Burkitt lymphoma and Burkitt-like lymphoma | + | + | + | + | + | +M | +/− | + | +/− | + | TdT- MIB-I 100% EBER in situ in 52% MYCc |
||||||
| Mediastinal large B-cell lymphoma | + | + | + | + | +/− | − | − | − | + | − | − | CD30+/− wk | |||||
| Large B-cell lymphoma | +/− | + | + | + | + | +/− | +/− | −/+ | −/+ | −/+ | − | −/+ | +/− | − | CD30 +/− MIB-I >40% |
||
| Lymphoplasmacytic lymphoma | +/− | + | + | + | + | +M/D | +M/G St | − | − | +/− | −/+b | − | |||||
| Hairy cell leukemia | + | + | + | + | + | + | −/+ | DBA.44+ CD79b-CDI lc+ CDI03+ CD2S+ st FMC7+ | |||||||||
| Primary effusion lymphoma | + | − | − | − | − | − | − | − | + | − | CD30 (Ki-I)+ HHV8+ EBER +/− | ||||||
| Plasmacytoma/myeloma | −/+ | − | −/+ | − | + | − | +G/A St | − | −/+ | − | +/− | − | + | −/+ | CDS6+ CD38 + EMA + | ||
cyt = cytoplasmic immunoreactivity; M, D, G, A, type of heavy Ig chain present; st = strong immunoreactivity; wk = weak immunoreactivity
Lymphoblasts in t(4;l I)(q2l; q23) ALL are CDIO negative and frequently CD24 negative.
Positive in plasma cell component.
- t(8;l4) (heavy chains) 85% of cases
- t(2;8) (kappa light chain)
- t(8;22) (lambda light chain).
Table 7-24.
T-cell neoplasms
| CD4S LCA | TCR | CD2 TE/TI 1 | CD3 T3 | CD43 Leu 22 | CDS Leu 1 | CD7 LEU 9 | CD4 T4 | CD8 T8 | CD2S IL2R | TIA-I | Granzyme B | CDS6 NCAM | CD30 Ki-I | TdT | ALK | OTHER | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Precursor lymphoblastic lymphoma/leukemia | + | − | +/− | + | +/− | +/− | + | +/− | +/− | +/− | − | − | − | − | + | CD34+ CD99+ CD la +/− |
|
| T-cell prolymphocytic leukemia | + | + | + | +wk | + | + | + | +/− | −/+ | +/− | − | − | − | − | − | CD la- | |
| Adult T-cell lymphoma/leukemia | + | + | + | + | + | + | −/+ | + | − | + | − | − | − | +/− | − | ||
| Mycosis fungoides and Sé zary syndrome | TCRβ+ | + | + | + | + | − | + | −/+ | −/+ | −/+ | +/− | − | − | − | − | IHEC A+ | |
| Peripheral T-cell lymphoma, NOS | + | + | +/− | +/− | + | +/− | −/+ | +/− | −/+ | + | +/− | −/+ | + (large cells) | − | |||
| Hepatosplenic | TCRδ + | ||||||||||||||||
| T- cell lymphoma | TCRaβ- | + | + | + | +/− | + | +/− | CDS7-CD16−/+ LMP-I-Perforin - | |||||||||
| Panniculitis-like | − | + | + | + | − | − | ∼ | + | + | + | − | − | − | ||||
| T-cell lymphoma | − | + | + | + | − | − | − | + | + | + | − | − | − | ||||
| CDS6+ | CD3ɛ | CD95+ | |||||||||||||||
| CDS6- | + | − | + | + | −/+ | + | CD9S- | ||||||||||
| CD3ɛ | |||||||||||||||||
| Angioimmunoblastic lymphoma | + | + | + | + | + | + | + | + | −/+ | − | + | + | − | − | − | − | CDI0+/− CDS7+ bcl-6± |
| Enteropathy-type T-cell lymphoma | + | + | + | − | + | − | −/+ | − | +/− | +/− | + (small cells) | + (large cells) | − | − | CD103+ | ||
| Anaplastic large cell lymphoma (Ki-I lymphoma) | +/− | +/− | +/− | −/+ | +/− | −/+ | −/+ | +/− | −/+ | +/− | +/− | +/− | −/+ | + (mem, Golgi) | − | +/−a (cyt, nuc) | Clusterin+b EMA+/−Perforin +/−EBER-BSAP- |
| Extranodal NK/T-cell lymphoma, nasal type | + | − | + | CD3e+ (cyt) | + | − | −/+ | − | − | − | + | + | + | −/+ | − | − | EBER+ CD16+ CD57− |
| Blastic NK-cell lymphoma | − | −/+ | − | +/− | −/+ | +/− | − | + | − | +/− | − | CD33-Myelo- | |||||
cyt = cytoplasmic; nuc = nuclear; wk = weak immunoreactivity.
Only positive in systemic ALCL (subset); negative in primary cutaneous ALCL.
Expressed in all cases of systemic ALCL but less commonly in primary cutaneous ALCL and very rarely in diffuse large B-cell lymphoma, peripheral T cell lymphoma, and NS HD
Table 7-25.
Hodgkin's lymphoma
| CD45 LCA | CD20 L26 | CD3 T3 | CD15 LEUM1 | CD30 Ki–1 | EMA | sIg | CD79a | CDw75 | Oct2 | BOB.1 | BSAP | LMP1 | OTHER | |
|---|---|---|---|---|---|---|---|---|---|---|---|---|---|---|
| Classical Hodgkin lymphoma (HL) | − | −/+ | − | +/− | + | − Rare | − | −/+ | − | − | −/+ | + | +/− | traf–1 + bcl–2 + |
| Nodular sclerosis HL | − | −/+ | − | +/− | + | − Rare | − | −/+ | − | − | −/+ | + | −/+ | |
| Lymphocyte-rich HL | − | −/+ | − | +/− | + | − Rare | − | −/+ | − | − | +/− | + | +/− | |
| Mixed cellularity HL | − | −/+ | − | +/− | + | − Rare | − | −/+ | − | − | −/+ | + | +/− | |
| Lymphocyte-depleted HL | − | −/+ | − | +/− | + | − Rare | − | −/+ | − | − | −/+ | + | + (if HIV +) | |
| Nodular lymphocyte predominant HL | + | + | − | − | −/+ | +/− | + | + wk | +/− | + | + | + | − | bcl-6 + bcl-2 - |
wk = weak.
Table 7-26.
Markers for tumors of unknown origin
| TYPE OF TUMOR | IMMUNOHISTOCHEMICAL MARKER(S) | POTENTIAL TREATMENT AND COMMENTS |
|---|---|---|
| Breast | ER/PR | ER/PR+ tumors can be palliated with hormonal treatment |
| HER-2/neu | HER-2/neu+ carcinomas can be treated with Herceptina | |
| GCDFP-15 | GCDFP-15 is not very sensitive, as many breast carcinomas are negative The most common type of breast carcinoma to present as an occult primary is invasive lobular carcinoma Rare women will present with positive axillary nodes with no known primary. Most of these women will have breast cancer. The prognosis is the same, whether or not the primary is detected |
|
| Carcinoid tumor | Chromogranin | Chromogranin positivity should be strong and diffuse. Focal and/or weak positivity can be seen in many carcinomas Metastatic breast cancer and prostate cancer can closely resemble carcinoid tumor and both can be positive for chromogranin Carcinoid tumors can be palliated with tumor-directed pharmaceuticals |
| Germ cell tumors | PLAP | PLAP is not specific but a germ cell tumor is unlikely if it is negative. Inhibin is more likely to be positive in choriocarcinomas Chemotherapy for possible cure |
| GIST | c-kit (CD117) | Treatment with Gleevecb |
| Lung adenocarcinoma | TTF-1 | 10% to 20% of patients will have specific activating mutations in EGFR (detected by PCR) and these patients may respond well to treatment with gefitinibc |
| Lymphoma | LCA, B- and T-cell markers | Treatment for cure or long-term palliation |
| Prostate | PSA or PrAP | Hormonal therapy effective for palliation |
| Small cell carcinoma | TTF-1 (if of lung origin) | Diagnosis made by H&E appearance Neuroendocrine markers are often positive. p63 is usually negative Chemotherapy for palliation |
| Squamous cell carcinomas | CK5/6, p63 | Not specific, but characteristic. H&E appearance usually sufficient to reveal keratin production or intercellular bridges Radiation therapy often effective |
| p16 or HPV | HPV or p16 is most commonly present in carcinoma of the cervix, but may be seen in carcinomas at other sites (e.g., basaloid carcinomas of the tonsil) Approximately 26-38% of patients with a cervical lymph node metastasis of unknown primary will have an occult tonsillar carcinoma. Complete sampling of the tonsil may be necessary to identify these small carcinomas |
|
| Thyroid: papillary or follicular carcinoma | Thyroglobulin and TTF-1 | Lung carcinomas are also TTF-1 positive, but will be thyroglobulin negative Highly effective treatment for cure with radioactive iodine |
| Thyroid: medullary carcinoma | Calcitonin | Palliative treatment with tumor-directed radionucleotides If familial, important for counseling other family members |
| Trophoblastic tumors | Inhibin | Inhibin is not specific, but a trophoblastic tumor is unlikely if it is negative Chemotherapy for possible cure |
Pathologists frequently receive specimens with metastatic tumors.12 Often, the site of origin is known to the clinician but this information is not provided to the pathologist. A good clinical history is frequently more successful for correct classification than a battery of immunoperoxidase studies. The CK7/CK20 pattern is generally helpful to narrow down the potential site of origin of carcinomas (see Table 7-3, Table 7-4, Table 7-5, Table 7-6, Table 7-7). Additional studies can then be used to identify specific types of carcinoma. The most important tumors to identify are those with specific therapeutic treatments for cure or palliation.
Trastuzumab (Herceptin) is a monoclonal antibody directed against the HER-2/neu receptor.
Imatinib mesylate (STI571, Gleevec™, Glivec™) is a small molecule tyrosine kinase inhibitor used for CML, ALL (Ph+), and GIST. The KIT protein is encoded by the c-KIT proto-oncogene and is a transmembrane receptor protein with tyrosine kinase activity. Mutated proteins may or may not respond to therapy with Imatinib. Mutations that render KIT independent of its ligand, SCF (stem cell factor), have been found in GIST, AML, germ cell tumors and systemic mastocytosis. Wild-type KIT and KIT with mutations in the juxtamembrane domain (the intracellular segment between the transmembrane and tyrosine kinase domains) are found in GISTs and are sensitive to imatinib. Other tumor types are associated with mutations in the enzymatic domain and the altered protein is generally not sensitive to imatinib.
Gefitinib (Iressa) is a tyrosine kinase inhibitor that is effective against a small subset of lung adenocarcinomas with specific activating mutations.
Table 7-27.
Markers for poorly differentiated tumors
| TYPE OF TUMOR | IMMUNOHISTOCHEMICAL MARKER | COMMENTS |
|---|---|---|
| Carcinoma | Broad-spectrum keratins | Some carcinomas may express unusual keratin subtypes. If negative, try other keratin types (e.g., CAM 5.2). The CK7/CK20 pattern may be helpful in determining the likely site of origin |
| AE1/AE3 or PANK (MNF-116) | Some non-carcinomas can have an epithelioid appearance and strongly express keratins (e.g., epithelioid angiosarcoma, epithelioid sarcoma, mesothelioma) | |
| Melanoma | S100 protein | S100 is strongly positive in the vast majority of melanomas |
| Some carcinomas (esp. breast) and sarcomas are also positive for S100 and additional markers may be required | ||
| HMB-45 and MART-1 are expressed by most epithelioid melanomas but may be focal or absent in non-epithelioid melanomas (e.g., spindle cell or desmoplastic melanomas) | ||
| Lymphoma | Leukocyte common antigen (LCA) | Present in almost all non-Hodgkin's lymphomas. |
| May be absent in 30% of anaplastic (Ki-1) large cell lymphomas. These lymphomas are keratin negative but may express EMA. These tumors will be positive for CD30 (K.i-1) and ALK |
Table 7-29.
Differential diagnosis of epithelial mesothelioma and lung adenocarcinoma
| EPITHELIAL MESOTHELIOMA | LUNG ADENOCARCINOMA | |
|---|---|---|
| Immunohistochemistry | ||
| AE1/AE3 keratin | POS (perinuclear)a | POS (membrane)b |
| Calretinin | POS | neg |
| WT1 (clone 6F-H2) | POS (nuclear)c | negd |
| CEA (polyclonal) | neg | Highe |
| LeuM1 (CD15) | neg | High |
| TTF-1 | neg | High |
| Mucins | ||
| Mucicarmine | 3-4% | 60% |
| PAS-D | <3% | 65% |
| Alcian blue | 30% | POS |
| Alcian blue + hyaluronidase | Staining lost | Staining preserved |
| Ultrastructure (EM) | ||
| Microvilli | Elongated, serpiginous, and branched | Short, blunt, rigid appearing |
| Length to diameter ratio | 10 to 16:1 | 4 to 7:1 |
| Cytogenetics | ||
| Deletions of 1p, 3p, 17p, loss of 9 and 22 | Deletions of 3p, highly variable changes | |
Tissue should be obtained for EM and cytogenetics, if possible.
Initial panel. AE1/AE3, calretinin, WT-1 (clone 6F-H2), CEA Leu-M1, and TTF-1 with additional studies ordered in difficult cases.
Other antibodies generally reported as negative in epithelial mesotheliomas and positive in lung adenocarcinomas include the following: MOC-1, B72.3, Ber-EP4, and BG-8. Cytokeratins 5 and 6 are reported to be positive in mesotheliomas and negative in lung carcinomas. However, in our experience, these markers have proven less useful than those listed above. The use of EMA is controversial. Strong membrane positivity is characteristic of epithelial mesothelioma, whereas cytoplasmic positivity is characteristic of adenocarcinomas.
Less is known about the immunophenotype of pure sarcomatoid mesotheliomas. The spindle cells are positive for cytokeratin, but are less frequently positive for the other markers as compared to the epithelioid cells. Tumors that can, on occasion, resemble mesotheliomas are generally negative for cytokeratins, with the notable exceptions of some cases of angiosarcoma, epithelioid hemangioendothelioma, synovial sarcoma, epithelioid sarcoma, and leiomyosarcoma (see Table 7-8).
Keratin immunoreactivity is accentuated around the nucleus and is present in the cytoplasm, without prominent membrane accentuation.
Keratin immunoreactivity is diffusely present in the cytoplasm with membrane accentuation in some cells.
WT1 immunoreactivity is nuclear.
Metastatic adenocarcinomas are generally negative for WT1 except for ovarian serous carcinomas and some renal carcinomas (see Table 7-5).
Most metastatic adenocarcinomas will be positive for CEA but there are some exceptions (see Table 7-5).
Table 7-2.
Evaluation of positivity of immunohistochemical studies
| GENERAL MARKERS | HEMATOPATHOLOGY MARKERS | ||||
|---|---|---|---|---|---|
| CATEGORY | % OF TUMORS | INTERPRETATION | CATEGORY | % OF TUMORS | INTERPRETATION |
| Positive (POS) | >90% | Almost always positive; a negative result would be unusual | + | >90% | Almost always positive |
| High | 60–90% | Most tumors are positive | +/− | >50% | Majority positive |
| Moderate (Mod) | 40–60% | May or may not be positive – usually the least useful type of marker | –/+ | <50% | Minority positive |
| Low | 10–40% | Most tumors are negative | − | <10% | Rarely positive |
| Negative (neg) or rare | <10% | Almost all tumors are negative; a positive result would be unusual | Blank | Results unknown or too few cases to quantify | |
| Blank | Results unknown or too few cases to quantify ? = Results based on very few cases (e.g.,<10) | ||||
The actual markers used to evaluate a case depend upon the differential diagnosis based on the H&E appearance. In some cases, an initial panel that is often used for typical cases has been suggested. Not all markers listed would be used for all cases, and some markers are included to indicate when they would not be useful for distinguishing the tumors listed in the table.
Cytokeratin 7 and Cytokeratin
The combination of these two cytokeratins has been found to be useful to divide carcinomas into four main groups (Ck7+/Ck20+, Ck7+/Ck20–, Ck7–/Ck20+, Ck7–/Ck20–).8, 9, 10
In Table 7-3, Table 7-4, Table 7-5, Table 7-6, Table 7-7, other commonly used antibodies have been included to show differences within each group. The most useful additional antibodies depend on the specific differential diagnosis.
Spindle cell lesions, soft tissue lesions, and sarcomas
See Table 7-8.
Small blue cell tumors
See Table 7-9.
Myoepithelial markers in breast cancer
See Table 7-10.
Epidermal lesions of the nipple
See Table 7-11.
Endocervical carcinoma versus endometrial carcinoma
See Table 7-12.
Endometrial stromal sarcoma versus leiomyosarcoma
See Table 7-13.
Primary ovarian carcinoma versus metastatic carcinomas
See Table 7-14.
Ovarian carcinoma versus mesothelioma
See Table 7-15.
Trophoblastic lesions
See Table 7-16.
Tumors of germ cells and sex-cord stromal tumors
See Table 7-17.
Adrenal and kidney tumors
See Table 7-18.
Tumors of bladder, prostatic, or renal origin
See Table-7.19.
Prostate carcinoma versus other lesions
See Table 7-20.
Hepatic tumors
See Table 7-21.
Thyroid and parathyroid lesions
See Table 7-22.
B-cell neoplasms
See Table 7-23.
T-cell neoplasms
See Table 7-24.
Hodgkin's lymphoma
See Table 7.25.
Metastatic tumors of unknown origin
See Table 7-26.
Poorly differentiated tumors
See Table 7-27.
Estrogen and progesterone receptor evaluation and HER2/neu score
See Chapter 15, pages 240–243.
EGFR (HER1) score
See Table 7-28.
Differential diagnosis of epithelial mesothelioma and lung adenocarcinoma13
See Table 7-29.
Box 7-1.
Alternative names for antigens
| Looking for? | Find it under: |
|---|---|
| 1D5 | Estrogen receptor (G) |
| 6F/3D | Beta-amyloid |
| 12E7 | CD99 (G, H) |
| 34βE12 | Keratins (G) |
| 38.13 | CD77 (H) |
| 70 kD NF | Neurofilaments (G) |
| 200 kD NF | Neurofilaments (G) |
| 903 | Keratins—34βE12 (G) |
| A (blood group antigen) | Blood group antigens (G) |
| A (Ig heavy chain α) | Heavy chain immunoglobulins (H) |
| A32 antigen | CD146 (G) |
| A103 | MELAN-A (G) |
| AAT | Alpha 1-antitrypsin (G, H) |
| ACH | Alpha-1 antichymotrypsin (H) |
| AE1/AE3 | Keratins (G) |
| AFP | Alpha-fetoprotein (G) |
| Alpha 1-antitrypsin | Alpha 1-antitrypsin (G, H) |
| Alpha 1-antichymotrypsin | Alpha 1-antichymotrypsin (H) |
| Alpha 1-fetoprotein | Alpha fetoprotein (G) |
| Alpha fetoprotein | Alpha fetoprotein (G) |
| Alpha-methylacyl-CoA racemase | AMACR (G) |
| Alpha smooth muscle actin | Alpha smooth muscle actin (G) |
| AMACR | AMACR (G) |
| Amyloid | Beta-amyloid (G) |
| Androgen receptor | Androgen receptor (G) |
| Apolipoprotein J | Clusterin (H) |
| AR | Androgen receptor (G) |
| B (blood group antigen) | Blood group antigens (G) |
| B1 | CD20 (H) |
| B2 | CD21 (H) |
| B4 | CD19 (H) |
| B72.3 | B72.3 (G) |
| bcl-1 | Cyclin Dl (H) |
| bcl-2 | bcl-2 (H, G) |
| B-cell specific activator protein | BSAP (H) |
| BER-EP4 | BER-EP4 (G) |
| BERH2 | CD30 (G, H) |
| Beta-amyloid | Beta-amyloid (G) |
| Beta-catenin | Beta-catenin (G) |
| Beta-2 microglobulin | Beta-2 microglobulin (G) |
| BG8 | BG8 (G) |
| β-hCG | Human chorionic gonadotropin (G) |
| BLA.36 | CD77 (H) |
| BL-CAM | CD22 (H) |
| Blood group antigens | Blood group antigens (G, H) |
| BR-2 | Gross cystic disease fluid protein-15 (G) |
| BRST-2 | Gross cystic disease fluid protein-15 (G) |
| C3b/C4bR | CD35 (H) |
| C5b-9 | C5b-9 (G) |
| c-kit | CD117 (G) |
| CA 15-3 | Epithelial membrane antigen (G, H) |
| CA 19-9 | CA 19-9 (G) |
| CA 27.28 | Epithelial membrane antigen (G, H) |
| CA 72-4 | B72.3 (G) |
| CA125 | CA125 (G) |
| CA19-9 | CA19-9 (G) |
| Calcitonin | Calcitonin (G), Hormones (G) |
| Caldesmon | Caldesmon (G) |
| Calgranulin | MAC 387 (G) |
| CALLA | CD10 (G, H) |
| CALP | Calponin (G) |
| Calponin | Calponin (G) |
| Calprotectin | MAC 387 (G) |
| Calretinin | Calretinin (G) |
| CAM5.2 | Keratins (G) |
| Carbohydrate antigen 19-9 | CA19-9 (G) |
| Carcinoembryonic antigen | Carcinoembryonic antigen (G) |
| CDla | CDla (H) |
| CD2 | CD2 (H) |
| CD3 | CD3 (H) |
| CD4 | CD4 (H) |
| CD5 | CD5 (G, H) |
| CD7 | CD7 (H) |
| CD8 | CD8 (H) |
| CD10 | CD10 (G, H) |
| CDllb | CDllb (H) |
| CDllc | CDllc (H) |
| CD13 | CD13 (H) |
| CD15 | CD15 (G, H) |
| CD16 | CD16 (H) |
| CD19 | CD19 (H) |
| CD20 | CD20 (H) |
| CD21 | CD21 (H) |
| CD22 | CD22 (H) |
| CD23 | CD23 (H) |
| CD25 | CD25 (H) |
| CD30 | CD30 (G, H) |
| CD31 | CD31 (G) |
| CD33 | CD33 (H) |
| CD34 | CD34 (G, H) |
| CD35 | CD35 (H) |
| CD38 | CD38 (H) |
| CD43 | CD43 (H) |
| CD44v3 | CD44v3 (G) |
| CD45 | CD45 (H) |
| CD45RA | CD45RA (H) |
| CD45Ro | CD45Ro (H) |
| CD56 | CD56 (H) |
| CD 57 | CD57 (G) |
| CD61 | CD68 (G, H) |
| CD68 | CD68 (G, H) |
| CD74 | CD74 (H) |
| CDw75 | CDw75 (H) |
| CD77 | CD77 (H) |
| CD79a | CD79a (H) |
| CD79b | CD79b (H) |
| CD95 | CD95 (H) |
| CD99 | CD99 (G, H) |
| CD117 | CD117 (G) |
| CD141 | CD141 (G) |
| CDX | CDX (G) |
| CDKN2 | pl6 (G) |
| CDP | Gross cystic disease fluid protein-15 (G) |
| CEA | Carcinoembryonic antigen (G) |
| c-erbB2 | HER-2/neu (G) |
| Chromogranin A | Chromogranin A (G) |
| c-kit | CD117 (G) |
| CLA | CD45 (H) or HECA-452 (H) |
| CLDN1 | Claudin (G) |
| Clusterin | Clusterin (H) |
| Collagen IV | Collagen IV (G) |
| Common acute | CD10 (G, H) |
| leukemia antigen | |
| Complement lysis inhibitor | Clusterin (H) |
| CR1 | CD35 (H) |
| Cyclin D1 | Cyclin Dl (H) |
| Cystic fibrosis antigen | MAC 387 (G) |
| D (Ig heavy chain δ) | Heavy chain immunoglobulins (H) |
| DBA.44 | DBA.44 (H) |
| Desmin | Desmin (G) |
| DF3 | Epithelial membrane antigen (G, H) |
| DPB | CD45RA (H) |
| E2 antigen | CD99 (G, H) |
| EBERS | Epstein-Barr virus (G, H) |
| EBNA | Epstein-Barr virus (G, H) |
| E-cadherin | E-cadherin (G) |
| EGFR | EGFR (G) |
| EM ACT | HHF-35 (G) |
| EMA | Epithelial membrane antigen (G) |
| E-MEL | HMB-45 (G) |
| Endothelial cell antigen | HECA-452 (H) |
| Ep-CAM | BER-EP4 (G) |
| Epidermal growth factor receptor | EGFR (G) |
| Epithelial membrane antigen | Epithelial membrane antigen (G, H) |
| Epithelial specific antigen | BER-EP4 (G) |
| Epstein-Barr virus | Epstein-Barr virus (G, H) |
| ER | Estrogen receptor (G) |
| erbB2 | HER-2/neu (G) |
| ESA | BER-EP4 (G) |
| Estrogen receptor | Estrogen receptor (G) |
| Ewing's sarcoma marker | CD99 (G, H) |
| Factor VIII related antigen | Factor VIII (G) |
| FVIILRAg | Factor VIII (G) |
| Factor XHIa | Factor XHIa (G) |
| Fasein | Fascin (H) |
| Fast myosin | Myosin Heavy Chain (G) |
| Fibronectin | Fibronectin (G) |
| Fli-1 | Fli-1 (G) |
| FMC7 | FMC7 (H) |
| FMC 29 | CD99 (G, H) |
| Friend leukemia integrin-site 1 | Fli-1 (G) |
| FVIILg | Factor VIII (G) |
| G (Ig heavy chain gamma) | Heavy chain immunoglobulins (H) |
| Gal-3 | Galectin-3 (G) |
| Galectin-3 | Galectin-3 (G) |
| Gastrin | Hormones (G) |
| GCDFP | Gross cystic disease fluid protein-15 (G) |
| GFAP | Glial fibrillary acidic protein (G) |
| Glial fibrillary acidic protein | Glial fibrillary acidic protein (G) |
| Glucagon | Hormones (G) |
| Glucose transporter 1 | GLUT-1 (G) |
| GLUT-1 | GLUT-1 (G) |
| GPIIIa | CD61 (H) |
| gp80 | Clusterin (H) |
| gp200 | RCC (G) |
| GPA | Glycophorin A (H) |
| Granzyme B | Granzyme B (H) |
| Gross cystic disease | Gross cystic disease fluid |
| fluid disease-15 | protein-15 (G) |
| H (blood group antigen) | Blood group antigens (G) |
| H222 | Estrogen receptor (G) |
| Hb | Hemoglobin (H) |
| HBME-1 | HBME-1 (G) |
| h-caldesmon | Caldesmon (G) |
| H-CAM | CD44v3 (G) |
| hCG | Human chorionic gonadotropin (G) |
| HCL | DBA.44 (H) |
| HBME-1 | HBME-1 (G) |
| Heavy chain immunoglobulins | Heavy chain immunoglobulins (H) |
| HECA-452 | HECA-452 (H) |
| Hematopoietic progenitor cell, class 1 | CD34 |
| Hemoglobin | Hemoglobin (H) |
| HepPar-1 | HepPar-1 (G) |
| Hepatocyte paraffin-1 | HepPar-1 (G) |
| HER-2/neu | HER-2/neu (G) |
| HHF-35 | HHF-35 (G) |
| HHV8 | HHV8 (H) |
| HLA-DR | HLA-DR (H) |
| HMB-45 | HMB-45 (G) |
| HMFG | Epithelial membrane antigen (G, H) |
| HNK-1 | CD57 (G, H) |
| hMLHl | hMLHl (G) |
| hMSH2 | hMLHl (G) |
| HNK-1 | CD57 (G) |
| HP1 | HepPar-1 (G) |
| HPCA-1 | CD34 (G, H) |
| HPL | Human placental lactogen (G) |
| HuLy-m6 | CD99 (G, H) |
| Human chorionic gonadotropin | Human chorionic gonadotropin (G) |
| Human herpesvirus 8 | HHV8 (G, H) |
| Human mutL homologue 1 | hMLHl (G) |
| Human mutS homologue 2 | hMLHl (G) |
| Human placental lactogen | Human placental lactogen (G) |
| IL-2 receptor | CD25 (H) |
| Inhibin-alpha subunit | Inhibin-alpha subunit (G) |
| Insulin | Hormones (G) |
| J5 | CD10 (G, H) |
| JOVI 1 | TCR (H) |
| K (Ig light chain κ) | Light chain immunoglobulins (H) |
| Keratin 5/6 | Keratins (G) |
| Keratin 7 | Keratins (G) |
| Keratin 20 | Keratins (G) |
| Keratins | Keratins (G) |
| Ki-1 | CD30 (G, H) |
| Ki-67 | Ki-67 (G) |
| kip2 | p57 (G) |
| Kit | CD117 (G) |
| KP-1 | CD68 (G, H) |
| L (Ig light chain lambda) | Light chain immunoglobulins (H) |
| L1antigen | MAC 387 (G) |
| L26 | CD20 (H) |
| L60 | CD43 (H) |
| Laminin | Laminin (G) |
| LCA | CD45 (H) |
| Leu 1 | CD5 (H) |
| Leu 2 | CD8 (H) |
| Leu 3 | CD4 (H) |
| Leu 5a + b | CD2 (H) |
| Leu 7 | CD57 (G, H) |
| Leu 9 | CD7 (H) |
| Leu 16 | CD20 (H) |
| Leu 22 | CD43 (H) |
| Leukocyte common antigen | CD45 (H) |
| LeuMl | CD15 (G, H) |
| Light chain immunoglobulins | Light chain immunoglobulins (H) |
| LFA-2 | CD2 (H) |
| LMP-1 | Epstein-Barr virus (G, H) |
| LN1 | CDw75 (H) |
| LN2 | CD74 (H) |
| Lysozyme | Lysozyme (H, G) |
| M (Ig heavy chain μ) | Heavy chain immunoglobulins (H) |
| Mac-1 | CDllb (H) |
| MAC 387 | MAC 387 (G) |
| Mac-M | CD68 (G, H) |
| MART-1 | MELAN-A (G) |
| Mast cell tryptase | Mast cell tryptase (H) |
| mb-1 | CD79a (H) |
| MCAM | CD146 (G) |
| ME491 | CD63 (G) |
| MELAN-A | MELAN-A (G) |
| Melanoma antigen recognized by T cells | MELAN-A (G) |
| Melanoma-associated antigen | CD63 (G) |
| Melanoma cell adhesion molecule | CD146 (G) |
| Melanoma-specific antigen | HMB-45 (G) |
| MELCAM (or Mel-CAM) | CD146 (G) |
| MIB-1 | Ki-67 (G) |
| MIC-2 | CD99 (G, H) |
| MN-4 | CD146 (G) |
| MNF-116 | Keratin—Pan-K (G) |
| MPO | Myeloperoxidase (H) |
| MRF4 | Myf-4 (G) |
| MSA | HHF-35 (G) |
| MTS1 | pl6 (G) |
| MUC1 | Epithelial membrane antigen (G, H) |
| MUC18 | CD146 (G) |
| Muscle common actin | HHF-35 (G) |
| Muscle-specific actin | HHF-35 (G) |
| My 7 | CD13 (H) |
| My 9 | CD33 (H) |
| Myeloperoxidase | Myeloperoxidase (H) |
| Myf-4 | Myf-4 (G) |
| MyoDl | MyoDl (G) |
| Myogenin | Myf-4 (G) |
| Myoglobin | Myoglobin (G) |
| Myosin Heavy Chain | Myosin Heavy Chain (G) |
| NCAM | CD56 (H) |
| Neprilysin | CD10 (G, H) |
| NEU N | NEU N (G) |
| Neurofilaments | Neurofilaments (G) |
| Neuron-specific enolase | Neuron-specific enolase (G) |
| NFP | Neurofilaments (G) |
| NKI-betab | HMB-45 (G) |
| NKI/C3 | CD63 (G) |
| NSE | Neuron-specific enolase (G) |
| 013 | CD99 (G, H) |
| OC125 | CA125 (G) |
| Oct2 | Oct2 (H) |
| Octomer transcription factor | Oct2 (H) |
| pl6 | pl6 (G) |
| p27kip1 | p27kip1 (G) |
| p53 | p53 (G) |
| p57 | p57 (G) |
| p63 | p63 (G) |
| P504S | AMACR (G) |
| PAN-K | Keratins (G) |
| PAP | Prostate acid phosphatase (G) |
| PECAM-1 | CD31 (G) |
| PEM | Epithelial membrane antigen (G, H) |
| Perform | Perform (H) |
| PGM1 | CD68 (G, H) |
| PgR | Progesterone receptor (G) |
| PK antigen | CD77 (H) |
| Placental alkaline phosphatase | Placental alkaline phosphatase (G) |
| PLAP | Placental alkaline phosphatase (G) |
| Platelet glycoprotein IIP | i CD61 (H) |
| PR | Progesterone receptor (G) |
| PRAD1 | Cyclin Dl (H) |
| PrAP | Prostate acid phosphatase (G) |
| Prealbumin | Prealbumin (G) |
| Progesterone receptor | Progesterone receptor (G) |
| Prostate acid phosphatase | Prostate acid phosphatase (G) |
| Prostate specific antigen | Prostate-specific antigen (G) |
| PSA | Prostate-specific antigen (G) |
| QBEndlO | CD34 (G, H) |
| Renal cell carcinoma marker | RCC (G) |
| ret | ret (G) |
| RCC | RCC (G) |
| rT3 | CD2 (H) |
| S100 | S100 (G) |
| S-Endo-1 | CD146 (G) |
| SGP-2 | Clusterin (H) |
| SMA | Alpha smooth muscle actin (G) |
| SM-ACT | Alpha smooth muscle actin (G) |
| Smad4 | DPC4 (G) |
| SM-MHC | Myosin Heavy Chain (G) |
| Somatostatin | Hormones (G) |
| SP40 | Clusterin (H) |
| Stem cell factor receptor | CD117 (G) |
| Synaptophysin | Synaptophysin (G) |
| Syndecan-1 | CD138 (H) |
| Synuclein-1 | Synuclein-1 (G) |
| T3 | CD3 (H) |
| T4 | CD4 (H) |
| T6 | CDla (H) |
| T8 | CD8 (H) |
| Til | CD2 (H) |
| T64 | Clusterin (H) |
| TAG-72 | B72.3 (G) |
| Tau | Tau (G) |
| T cell antigen receptor | TCR (H) |
| T cell intracellular antigen | TIA-1 (H) |
| TCR | TCR (H) |
| TdT | Terminal deoxytransferase (H) |
| TE | CD2 (H) |
| Terminal | Terminal |
| deoxytransferase | deoxytransferase (H) |
| TH | CD4 (H) |
| Thrombomodulin | CD141 (G) |
| Thyroglobulin | Thyroglobulin (G) |
| Thyroid transcription factor 1 | TTF-1 (G) |
| TIA-1 | TIA-1 (H) |
| TM | CD141 (G) |
| traf-1 | traf-1 (H) |
| Transthyretin | Prealbumin (G) |
| TRPM2 | Clusterin (H) |
| TTF-1 | TTF-1 (G) |
| TTR | Prealbumin (G) |
| Tumor-associated glycoprotein 72 | B72.3 (G) |
| Tumor necrosis factor factorreceptor-associated | traf-1 (H) |
| UCHL-1 | CD45Ro (H) |
| UEA 1 | Ulex (G) |
| Ulex | Ulex (G) |
| Vimentin | Vimentin (G) |
| von Willebrand's factor | Factor VIII (G) |
| VWF | Factor VIII (G) |
| Wilms' tumor 1 protein | WT1 (G) |
| WT1 | WT1 (G) |
G, General markers; H, hematopathology markers.
Results
The results of immunoperoxidase studies are incorporated into the surgical pathology report.14 The following information is included:
-
•
The type of tissue studied: formalin-fixed (or other fixatives) tissue, cryostat sections, cytology preparations, etc.
-
•
The type of immunoagents used, being as specific as possible. For example, do not just list “keratin” but specify the type of keratin (e.g., AE1/AE3).
-
•
The results of the studies in sufficient detail to allow interpretation: for example, the type of cell that is immunoreactive (e.g., tumor versus nontumor), intensity of immunoreactivity (e.g. weak, strong), and/or the number of cells immunoreactive (e.g., focal versus diffuse).
-
•
Integration of the results into the final diagnosis, specifying whether they confirm or support a diagnosis, make one diagnosis more likely than others, or exclude one or more diagnoses.
ELECTRON MICROSCOPY
EM continues to have an important role in surgical pathology.15
Indications for EM studies
-
•
Diagnostic renal biopsies for glomerular disease
-
•
Adenocarcinoma versus mesothelioma (see Table 7-29)
-
•
Difficult to classify tumors (Table 7-31 )
-
•
Nerve (e.g., toxic or drug-induced neuropathy) and muscle biopsies (e.g., inclusion body or nemaline myopathy)
-
•
Bullous skin diseases (e.g., epidermolysis bullosa)
-
•
Ciliary dysmorphology (primary ciliary dyskinesia or Kartagener's syndrome)
-
•
Endomyocardial biopsies (e.g., Adriamycin toxicity, amyloid, nemaline)
-
•
Liver biopsies for microvesicular fat in acute fatty liver of pregnancy
-
•
Small bowel biopsies to look for pathogens (e.g., Whipple's disease)
-
•
Congenital, inherited, and metabolic diseases (e.g., ceroid lipofuscinoses)
-
•
Prion diseases.
Table 7-31.
Electron microscopic features of poorly differentiated tumors
| TUMOR | ULTRASTRUCTURE | ADDITIONAL TESTS | COMMENTS |
|---|---|---|---|
| Carcinoma |
|
|
Other tumors can also be keratin positive and have desmosomes, filaments, and cytokeratin (mesothelioma, meningioma, synovial sarcoma, and epithelioid sarcoma) |
| Melanoma |
|
|
Melanosomes are also seen in clear cell sarcoma, pigmented schwannomas, PEComa, and other rare tumors. Mature forms can be taken up by melanophages, keratinocytes, and carcinomas |
| Lymphoma | No specific features are present. The cells lack cellular junctions and there is a paucity of cytoplasmic organelles | IHC: LCA | LCA may be absent in 30% of anaplastic (ALK1) lymphomas. These tumors can be EMA (+) but are keratin (−) |
| Sarcoma |
|
IHC: May be helpful for identifying specific types | Keratin negative except for synovial sarcoma and epithelioid sarcoma (or rarely in other types) |
Method
Ultrastructural details of tissues are rapidly lost; therefore fresh tissue must be fixed rapidly and well for EM. Tissues are usually fixed in special fixatives for EM to preserve lipids and glycogen (e.g., 2% paraformaldehyde and 2.5% glutaraldehyde in 0.1 M cacodylate buffer, pH 7.4).
-
1.
Place a small fragment of tissue in a drop of fixative on a cutting surface.
-
2.
Cut the tissue into multiple tiny fragments, each no greater than 0.1 cm in any dimension.
-
3.
Place the tissue into the vial of fixative. Shake the vial to make sure all the tissue fragments are covered by fixative.
Note.
If tissue from a small biopsy is found to be nondiagnostic on H&E, any tissue saved for EM should be retrieved for examination by light microscopy.
Results
A separate EM report is usually issued. The results should be incorporated into the final diagnosis.
SNAP-FROZEN TISSUE
Frozen tissue is useful for immunoperoxidase staining (some antibodies only detect antigens in frozen tissue), enzyme studies (muscle biopsies), and to save tissue for DNA or RNA studies.
Indications
Frozen tissue is useful for all specimens in which there is a question of a lymphoproliferative disorder, sarcomas, unusual tumors, and muscle biopsies.
Methods
Small (approximately 0.5 × 0.5 × 0.3 cm3) portions of tissue are placed in a clean specimen container moistened with a small amount of normal saline until they can be frozen.
Protocol for freezing tissue
Equipment needed:
-
•
Flask
-
•
Labeled self-seal bag
-
•
Pyrex beaker
-
•
Liquid nitrogen
-
•
Tin foil
-
•
Isopentane (2-methylbutane).
Technique
-
1.
Fill a flask half full with liquid nitrogen. Always wear protective gloves and a face shield. Dry ice can also be used. The liquid nitrogen is kept in a canister in the Reproductive Endocrinology Laboratory. Gloves and a face shield are stored nearby.
-
2.Work in a safety cabinet and wear surgical gloves and goggles. Label a freezer bag with the following:
-
•Date
-
•Patient's name in full
-
•Histology reference number
-
•Diagnosis/type of tissue.
-
•
-
3.
Place 2 cm (¾ inch) of isopentane (2-methylbutane) into a glass beaker. Lower it gently into the flask containing liquid nitrogen. The isopentane is ready to use when the liquid base is frozen solid (white particles will appear) and the remaining liquid is viscous. Remove the beaker from the flask carefully.
-
4.
Using the forceps, drop small pieces of tissue directly into the isopentane. Freeze the tissue for approximately 30 seconds.
-
5.
Remove the tissue with the forceps and quickly wrap in aluminum foil. Place the tissue in a labeled freezer bag and place in the flask containing the liquid nitrogen.
-
6.
Transfer the specimen bag from the flask to a –20°C freezer.
Table 7-32.
Cells, tumors, and structures with characteristic findings by electron microscopyTUMOR EM FINDINGS CORRELATIONS AND OTHER DIAGNOSTIC TESTS Alveolar soft part sarcoma Rhomboid, rod-shaped, or spiculated crystals in a regular lattice pattern - The characteristic cytoplasmic crystals are composed of monocarboxylate transporter 1 (MCT1) and its chaperone CD147. These proteins are found in many other cell types and are not specific for this tumor
- Cytogenetics: t(X;17) creates a ASPL–TFE3 fusion protein
- IHC: TFE3 positive (as well as rare pediatric renal tumors with the same translocation). Immunoreactivity is not present in other tumors or normal tissues
- Histo: The crystals are PAS with diastase positive
Amyloid - Non-branching. brils, 7.5 to 10 nm in width and up to 1 μm in length
- May be present associated with plasma cell tumors, medullary carcinoma of the thyroid, Alzheimer's disease, or as an isolated finding (primary amyloidosis)
- IHC: Can be used to identify specific types of amyloid (e.g., lambda or kappa chains, β2- microglobulin, calcitonin, tau)
Bronchioloalveolar carcinoma of the lung (BAL) - Lamellar (surfactant) “myelin-like” granules in the supranuclear cytoplasm (typical of type II pneumocytes)
- Clara-like electron-dense granules in supranuclear cytoplasm.
- Intranuclear inclusions comprised of parallel microtubular arrays
- These features can also be seen in metastatic BAL
- Cytogenetics: These carcinomas are less likely to be associated with smoking and have fewer cytogenetic changes
- Bronchioloalveolar carcinomas or adenocarcinomas with features of BAL are more likely to respond to Iressa (38%) as compared to other lung carcinomas (14%) due to specific mutations in EGFR
- Mucinous BAL has intranuclear inclusions but generally lacks the other EM features
Chordoma - Desmosomes, large vacuoles, glycogen, dilated ER, cytoplasmic invaginations, and intermediate filaments
- The physaliphorous (having bubbles or vacuoles) appearance is due to dilated ER, glycogen, and cytoplasmic invaginations
IHC: keratin (corresponds to intermediate filaments), EMA, S100 Clear cell sarcoma Melanosomes in various stages of development Glycogen (resulting in clear cytoplasm) Cytogenetics: t(12;22) EWS–ATF1 fusion protein IHC:S100, HMB-45 Dense core granules - Dense core granules (vesicle bound by a single membrane with a dense center—60 to 300 nm), cytoplasmic organelles involved in regulated exocytosis of cell products
- Examples:
- Pancreatic beta cells (insulin):angular crystalline inclusions
- Pheochromocytoma (epinephrine and norepine- phrine):large, pleomorphic, often clear or only partially filled
- Carcinoid:
- Foregut—small, round
- Midgut—larger, pleomorphic
- Hindgut—mixed
- Found in tumors of neuronal or neuroendocrine origin
- Vesicles are comprised of granins (predominantly chromogranin A, chromogranin B, and secretogranin II) and various peptide hormones and transmitters, ATP, and biogenic amines
- IHC: chromogranin A (most specific). Specific products of tumors can also be detected
- Note: Prostate cancers and breast cancers can also show strong chromogranin positivity and can be mistaken for neuroendocrine tumors, particularly at metastatic sites
Desmoplastic small round cell tumor Numerous desmosomes and tight junctions, numerous cell processes, large number of organelles (mitochondria and RER), microfilaments, small neurosecretory granules - Cytogenetics: t(11;22) EWS–WT1 fusion protein
- IHC: keratin, desmin, WT1, actin, EMA, NSE
Endothelial cells Weibel–Palade bodies (cigar-shaped membrane bound structures filled with tubules in parallel arrays) Intracytoplasmic lumina may be present in normal cells and in epithelioid vascular neoplasms - Weibel–Palade bodies are frequently absent in tumors arising from endothelial cells (e.g., angiosarcomas). IHC markers are more sensitive to detect endothelial derivation
- The membranes are formed by P-selectin and the tubules contain FVIII
- IHC: Vascular markers (CD34, CD31, FVIII)
Ewing's sarcoma (PNET) Homogeneous cell population characterized by the lack of specialized features, large pools of glycogen, no organelles, no extracellular matrix, variable numbers of neurosecretory granules and cell processes - Cytogenetics: t(11;22) EWS–FLI1 fusion protein (and other less common variants)
- IHC: CD99. FLI1 is also present, but is less specific
- Histo: PAS +/− diastase can detect glycogen, but is not currently used for diagnosis
Granular cell tumor Numerous lysosomes (filled with tubular, vesicular, and amorphous material), phagosomes, and granules (correlating with the “granular” cytoplasm), reduplicated basal lamina surrounding groups of cells IHC: S100, inhibin, CD68, calretinin Langerhans cell histiocytosis Birbeck granules (rod or tennis racket shaped) structures of variable length with a central periodically striated lamella - May serve as a reservoir for Langerin (a transmembrane type II Ca2+-dependent lectin) and CD1a in the endosomal recycling compartment
- IHC: CD1a, Langerin, S100
Mast cells Lamellar or scroll-like membrane pattern, granules of variable size IHC: CD117 (c-kit), mast cell tryptase Medullary carcinoma of the thyroid Numerous neurosecretory granules (calcitonin) associated with stromal amyloid (calcitonin) - Cytogenetics: mutations in the RET gene (sporadic and germline)
- IHC: Calcitonin (in tumor cells and amyloid), chromogranin
Merkel cell carcinoma Neurosecretory granules in processes or along cell membranes (subplasmalemmal) IHC: chromogranin, NSE, cytokeratin 20 Mesothelioma Elongated, serpiginous, and branched microvilli (generally 10 to 16 length:1 width) apical without a glycocalyx or actin rootlets - Cytogenetics: Characteristic chromosome deletions and loss of 9 and 22
- IHC: Calretinin, WT1
Neuroblastoma Cellular processes with microtubules (neuropil), dense core granules, Homer–Wright rosettes (the center is comprised of a tangle of cell processes), synaptic vesicles, no glycogen - Cytogenetics: Changes are linked to prognosis
- IHC: chromogranin, NSE, NFP, synaptophysin
Oncocytoma Numerous mitochondria packed in the cytoplasm (correlating with the granular appearance of the cytoplasm). In contrast, chromophobe renal cell carcinoma has fewer mitochondria and more microvesicles - Cytogenetics: Monosomy with loss of X or Y, 11q13.
- Chromophobe carcinomas have different cytogenetic changes
- IHC: RCC is negative in oncocytomas but positive in 45–50% of chromophobe renal cell carcinomas
Perineurioma Long cell processes wrapping around adjacent cells IHC: Claudin-1 (a component of tight junctions), EMA Rhabdoid tumor of the kidney Large paranuclear whorls of intermediate filaments (corresponding to cytokeratin and vimentin) and occasional tonofilaments - Cytogenetics: hSNF5/INI1 deletions and mutations
- IHC: Cytokeratin, vimentin
Rhabdomyosarcoma Parallel thick (12 to 15 cm) and thin (6 to 8 nm) myosin-actin filaments, Z bands, filament ribosomal complexes - Cytogenetics: Characteristic changes in alveolar and embryonal types
- IHC: Muscle markers (HHF-35, desmin, myf4)
Spider cells may be seen in cardiac tumors (clear cytoplasm divided by cytoplasmic processes and cross striations formed by leptofibrils) Schwannoma Basal lamina prominent, often reduplicated. Luse bodies (long spacing collagen, extracellular), myelin figures, long cell processes wrapping around collagen, may rarely have melanosomes (melanotic schwannoma) - Cytogenetics: Deletion of 2q (NF2 inactivation)
- IHC: S100
-
7.
Discard the isopentane in a waste bottle. Liquid nitrogen can be allowed to evaporate.
-
8.
Sterilize the beaker and forceps with 10% formalin.
-
9.
Dispose of contaminated sharps and specimen containers in appropriate impervious biohazard containers. Wash your hands after removal of gloves.
If there is insufficient tissue for snap freezing, a frozen section from the OR Consultation Room may be saved frozen for potential studies. Many cryostats undergo an automatic defrost cycle and tissue left as a block in the cryostat will thaw and refreeze. Thus tissue to be saved frozen should be transferred to a freezer.
Results
The results of immunoperoxidase studies on frozen tissue are usually incorporated into the surgical pathology report.
IMMUNOFLUORESCENCE
Like immunoperoxidase studies, immunofluorescence detects antigens in tissues. However, because amplification of the signal is not used, it is better suited for precise localization of antigen/antibody complexes in tissues or for determining the deposition pattern of immune complexes (e.g., linear versus granular). Thus, it is most useful for the investigation of diseases related to immune complex deposition such as glomerular diseases and bullous diseases of the skin.
Tissue for immunofluorescence may be snap frozen (see instructions above) or stored in special fixatives for IF. If the specimen is not frozen, special care must be taken to ensure that the biopsy is kept moist in a sealed container.
Direct IF uses antibodies to detect antigens in the patient's tissues.
Indirect IF uses control tissues to detect antibodies (e.g., anti-BM) in the patient's serum.
Indications
Biopsies of some skin diseases (e.g., lupus, pemphigus, pemphigoid, and dermatitis herpetiformis), all diagnostic non-transplant renal biopsies, some transplant renal biopsies, and the evaluation of vasculitis in nerve biopsies.
Method
Tissue must be submitted fresh.
Results
The results of the examination are usually incorporated into the surgical pathology report.
Immunofluorescence of skin lesions
SLE (lupus band test).
There is linear or granular staining along the dermal–epidermal junction for multiple immunoreactants (most commonly IgG and less often IgM or C3) in approximately 80% of cases. The specificity increases with the number of positive immunoreactants. Uninvolved sun-exposed skin shows positivity in most patients with active systemic lupus. Uninvolved skin in patients with discoid lupus is usually negative for this test.
Herpes gestationis.
Perilesional skin shows linear basement membrane zone C3 and sometimes IgG.
Dermatitis herpetiformis.
Granular IgA is seen at the tips of dermal papillae of uninvolved skin.
Pemphigus.
IgG and C3 between epidermal cells create a net-like pattern. In pemphigus vulgaris, a split just above the basal cell layer creates a “tombstone” appearance to the row of basal cells at the base of the vesicle. In pemphigus foliaceus and related disorders, the split occurs near the granular cell layer.
Pemphigoid.
Ig and C3 are present along the basement membrane but not between cells. Indirect IF reveals an anti-BM antibody.
MOLECULAR GENETIC PATHOLOGY
Molecular genetic pathology is the newest subspeciality in pathology with board certification. Molecular diagnostics incorporates many types of techniques for the investigation of genetic alterations in cells and viruses (e.g., Southern blotting, PCR analysis, FISH). It has applications in three main areas:
-
1.Inherited diseases:
-
•Identification of inherited diseases (e.g., cystic fibrosis, hemochromatosis, factor V Leiden, prothrombin 20210A, fragile X syndrome)
-
•Identification of genes conferring susceptibility to diseases (e.g., BRCA1).
-
•
-
2.Infectious diseases:
-
•Detection of organisms
-
•Identification of specific organisms
-
•Quantitation of viral infection (e.g., HIV viral load).
-
•
-
3.Cancer
-
•Identification of specific genetic alterations associated with tumors (Table 7-33 )
-
•Identification of clonality in hematolymphoid proliferations (Table 7-34 )
-
•Detection of minimal residual disease after treatment.
-
•
Molecular genetic studies are especially helpful for difficult-to-classify hematolymphoid proliferations because of the frequent and characteristic rearrangements that occur in many of these disorders. Unlike cytogenetics, the cells need not be viable; however, it is preferable that the nucleic acids are relatively intact. Southern blot and RNA based PCR (RT-PCR) assays are best performed on fresh or frozen tissues. Formalin-fixed, paraffin-embedded tissue is amenable to DNA-based PCR assays. Some fixatives (e.g., Bouin's) cause extensive breakage of DNA and may preclude genetic analysis of the tissue.
Table 7-33.
Common cytogenetic and genetic changes in solid tumors of diagnostic or therapeutic significance
| TUMOR TYPE | CHARACTERISTIC CYTOGENETIC CHANGES | GENETIC CHANGES | FREQUENCY | COMMENTS |
|---|---|---|---|---|
| Adenoid cystic carcinomas | 6q translocations and deletions | >50% | ||
| Adrenal cortical carcinomas | 2p16 loss | >90% | This area is close to the region associated with Carney complex type 2 | |
| 17p13 LOH | 85% | These changes are less common in localized tumors (25–35%) but, if present, such tumors are more likely to metastasize. The 11p15 imprinted region is also involved in Beckwith–Wiedemann syndrome |
||
| 11p15 LOH with duplication of the active paternal allele leading to IGF-II overexpression | 85% | |||
| Alveolar soft part sarcoma | der(X)(X;17)(p11;q25) | ASPL–TFE3 fusion | >90% | TFE3 can be detected by IHC. This translocation is also present in rare papillary-like renal tumors in young adults (see “Renal tumors” below) |
| Aneurysmal bone cyst | t(16;17)(q22;p13) | CDH11–USP6 fusion | >50% | |
| Angiomatoid fibrous histiocytoma | t(12;16)(q13;p11) | FUS–ATF fusion | ||
| Breast carcinoma | HER-2/neu amplification | 20–30% | Detected by FISH (gene amplification) or IHC (protein overexpression). Positive carcinomas are more likely to respond to Herceptin | |
| BRCA1 and BRCA2 germline mutations | <5% | Patients are more likely to be young and have multiple carcinomas. BRCA1 carcinomas are frequently high grade, have “medullary” features, and lack ER, PR, HER-2. BRCA2 carcinomas have no specific pathologic features | ||
| Carcinoma of the upper aerodigestive tract in children | t(15;19)(q13;p13.2) | BRD4–NUT fusion | Patients with this translocation have a poor prognosis | |
| Chondromyxoid fibroma | Deletion of 6q | >75% | ||
| Clear cell sarcoma | t(12;22)(q13;q12) | EWS–ATF1 fusion | >75% | |
| Colon carcinoma | hMLH1 and hMSH2 mutations | 15% of sporadic carcinomas | 95% of HNPCC patients have germline mutations in these genes. Absence can be detected by IHC or by PCR assays for microsatellite instability. Mutations are correlated with characteristic clinical, pathologic, and treatment response features | |
| EGFR (HER1) overexpression | 82% of all carcinomas | Approximately 23% of patients treated with cetuximabb and chemotherapy respond. IHC for EGFR may be used to select eligible patients | ||
| APC mutations | 80% of all carcinomas | Also present as a germline mutation in familial adenomatous polyposis syndrome | ||
| LKBI/STKI1 LOH | ∼15% | Germline mutations occur in some cases of Peutz-Jeghers syndrome. Mutations appear to be rare in sporadic colon carcinoma but LOH is observed in some | ||
| DPC4 (Smad4 or MADH4) mutations (I8q2l.l) | 10-20% | Germline mutations occur in some cases of juvenile polyposis syndrome. Mutations in sporadic carcinomas are uncommon | ||
| Desmoplastic small round cell tumor | t(l l;22)(pl 3;q12) | EWS-WTI fusion | >7S% | WTI can be detected by IHC |
| Dermatofibrosarcoma protuberans | t(l7;22)(q2l;q13) resulting in a ring chromosome | COLIAI-PDGFB fusion | >7S% | The same translocation is present in giant cell fibroblastoma, but without formation of a ring chromosome |
| Endometrial stromal tumor | t(7;l7)(plS;q2l) | JAZFI-JJAZI fusion | 30% | |
| Ewing's sarcoma (PNET) | t(l I;22)(q24;q12) | EWS-FLII fusion | >80% | FLI1 can be detected by IHC but is not specific for Ewing's |
| t(2l;22)(q22;q12) | EWS-ERG fusion | 5-10% | ||
| t(2;22)(q33;q12) | EWS-FEV fusion | <5% | ||
| t(7;22)(p22;q12) | EWS-ETVI fusion | <5% | ||
| t(l7;22)(ql2;q12) | EWS-EIAF fusion | <5% | ||
| inv(22)(q I2)(ql2) | EWS-ZSG fusion | <5% | ||
| Extraskeletal myxoid chondrosarcoma | t(9;22)(q22;q12) | EWS-NR4A3 fusion | >75% | |
| t(9;l7)(q22;q11) | TAF2N-NR4A3 fusion | <10% | ||
| t(9;IS)(q22;q2l) | TCF12-NR4A3 fusion | <10% | ||
| Fibromatosis (desmoid) | Trisomies of 8 and 20 | 30% | ||
| Deletion of 5q | APC inactivation | 10% | ||
| Fibromyxoid sarcoma, low grade | t(7;l6)(q33;pl 1.2) | FUS-BBF2H7 fusion | Unknown | |
| Fibrosarcoma, infantile |
t(12;15) (pi 3;q26) |
ETV6-NTRK3 fusion |
>75% | The same translocation is seen in cellular mesoblastic nephroma |
| Trisomies 8, 11, 17,20 | >75% | |||
| Gastrointestinal stromal tumor | Monosomies 14 and 22 | > 75% | ||
| Deletion of 1p | >25% | |||
| KIT or PDGFRA mutation | >90% | CDI 17 (KIT) is detected by IHC and is useful for diagnosis. Gleevecc is effective against tumors with activating mutations in either gene | ||
| Germ cell tumors | Isochromosome I2p | >80-90% | Includes all histologie subtypes | |
| KIT mutations | 25-70% | Seminomas | ||
| Giant cell tumor | Telomeric changes | >50% | ||
| Giant cell tumor, diffuse type (PVNS) | Trisomies 5 and 7 t(1;2) | >25% | ||
| Hepatoblastoma | Trisomies 2q and 20 | >75% | ||
| Hibernoma | 11q13 rearrangement | >50% | ||
| Inflammatory myofibroblastic tumor | 2p23 rearrangement | ALK fusion with multiple partners | 50% | ALK can be detected by IHC in one third of cases |
| Leiomyoma, Uterine | t(12;14)(q15;q24) deletion of 7q | HMGA2 rearrangement | 40% | Uterine leiomyosarcomas have more complex karyotypes |
| Lipoblastoma | 8q12 rearrangement polysomy 8 | PLAGI oncogene | >80% | |
| Lipoma | ||||
| Typical | 12q15 rearrangement 6p21 rearrangement |
HMGA2 rearrangement HMGAI rearrangement |
60% | |
| Spindle cell or pleomorphic | Deletion of 13q or 16q | >75% | ||
| Chondroid | t(11;16)(q13;p12-13) | |||
| Liposarcoma | ||||
| Well-differentiated | Ring form of chrom 12q1,5/giant markers | HMGA2, MDM2 amplification | >75% | |
| Myxoid/round cell | t(12;16)(q13;p11) | FUS-CHOP fusion | >75% | |
| t(12;22)(q13;q12) | EWS-CHOP fusion | <5% | ||
| Pleomorphic | Complex | 90% | ||
| Lung adenocarcinomas that respond to gefitinib (most have features of bronchioloalveolar carcinoma) |
Fewer changes than seen in carcinomas associated with smoking |
EGFR—small deletions or amino acid substitutions |
10-20% of all lung carcinomas |
Mutations predict response to the tyrosine kinase inhibitor gefitinib (Iressa)d |
| 40-80% of lung carcinomas show EGFR overexpression by IHC, but only carcinomas with specific mutations respond to gefitinib | ||||
| Medulloblastoma | Isochromosome 17q | >25% | ||
| Meningioma | Monosomy 22 | 90% | ||
| 1p deletion | 25% | |||
| Mesothelioma | Deletion of 1p | ? BCLIO inactivation | >50% | Cytogenetic changes are less complex than those seen in carcinomas. Cytogenetic analysis of cytologic specimens (e.g., pleural fluid) can be of value if larger biopsies are not available |
| Deletion of 9p | p15, p16, and p19 inactivation | >75% | ||
| Deletion of 22q | NF2 inactivation | >50% | ||
| Deletions of 3p and 6q | >50% | |||
| Mucoepidermoid carcinoma | t(11;19)(q21;p13) | MECTI-MAML2 fusion | >50% | |
| Neuroblastoma | ||||
| Hyperdiploid, no 1p deletion | 40% | Good prognosis | ||
| 1p deletion | 40% | Poor prognosis | ||
| Double minute chromosomes | N-myc amplification | >25% | ||
| Oligodendroglioma | Deletion of 1p36 and 19q13.3 | 50% | Useful for diagnosis and to predict response to radiation and/or chemotherapy | |
| 9p21 deletion | CDKN2A (p16) deletion | Occurs in some anaplastic oligodendrogliomas. Poor prognostic factor | ||
| Osteochondroma | Deletion of 8q | EXTI inactivation | >25% | |
| Osteosarcoma | ||||
| Low grade | Ring chromosomes | >50% | ||
| High grade | Complex | RB and P53 inactivation | >80% | |
| Pheochromocytoma | ||||
| Sporadic (70%) | Losses on 1p | >80% | ||
| Hereditary (30%) | Germline mutations in | >90% of hereditary cases |
Patients are more likely to be young (<50), have multiple tumors, and have a family history of pheochromocytoma, paraganglioma, or medullary carcinoma of the thyroid |
|
| RET, VHL, NFI, SDHB, | ||||
| SDHD, MEN2A, MEN2B | ||||
| Pleomorphic adenoma (salivary) | 8q12 rearrangement | PLAGI fusion genes | >50% | |
| 12q15 rearrangement | HMGIC oncogenes | <20% | ||
| Renal tumors | ||||
| Clear cell carcinoma | Deletion of 3p | >90% | ||
| Papillary carcinoma: adult | Trisomies 3, 7, 12, 16, 17, and 20 | >90% | ||
| KIT mutations | >90% | CD117 (c-kit) present by IHC in cytoplasm and is associated with activating mutations | ||
| “Papillary-like” carcinoma: young adults | t(X;1)(p11.2;q21) | PRCC-TFE3 fusion | The majority of these carcinomas are associated with fusion proteins involving TFE3 or | |
| t(X;1)(p11.2;p34) | TFE3-PSF fusion | |||
| inv(X)(p11.2q12) | TFE3-NonO fusion | |||
| t(X;17)(p11.2;q25.3) | RCCI 7(ASPL)-TFE3 fusion | TFEB. | ||
| t(6;11)(p21.1;q12) | TFEB-Alpha fusion | The ASPL-TFE3 fusion is also present in alveolar soft part sarcoma | ||
| Oncocytoma | −1,-X or -Y | >25% | ||
| 11q13 rearrangement | >25% | |||
| Chromophobe carcinoma | Monosomies 1,2, 3, 6, 10, 13, 17, and 21 | >75% | CD117 (c-kit) is present by IHC on membranes, but activating mutations have not been detected | |
| Retinoblastoma |
13q14 deletion | RBI inactivation | >75% | 40% of cases are due to germline mutations in RBI |
| Isochromosome 6p | 25% | |||
| Rhabdoid tumor of the kidney and | Normal karyotype | hSNF5/INII (22q11.2) deletions and mutations | >90% | Infants and children with both tumors have a germline mutation in INI! (rhabdoid predisposition syndrome) |
| Atypical teratoid/rhabdoid tumor (AT/RT) | Monosomy 22 | hSNF5/INII deletions and mutations | Choroid plexus carcinomas are also associated with non- function of this gene (70%) | |
| Rhabdomyosarcoma | ||||
| Alveolar | t(2;13)(q35;q14) | PAX3-FKHR fusion | >75% | Poor 4-year survival if metastatic (8%) |
| t(1;13)(p36;q14), double minutes | PAX7-FKHR fusion | 10-20% | Better 4-year survival if metastatic (75%) | |
| Embryonal | Trisomies 2q, 8, and 20 | LOH 11p15 | >75% >75% | |
| Schwannoma and perineurioma | Deletion of 22q | NF2 inactivation | >80% | 5% of cases of vestibular schwannomas are associated with neurofibromatosis type 2 (germline NF2 mutations) |
| Synovial sarcoma | ||||
| Monophasic | t(X;18)(p11;q11) | SYT-SSXI/SYT-SSX2 fusion | >90% | |
| Biphasic | t(X;18)(p11;q11) | SYT-SSI fusion | >90% | |
| Thyroid carcinoma | ||||
| Papillary | 10q11 rearrangement | RET fusion oncogenes | >30% | |
| 1q21 rearrangement | NTRKI fusion oncogenes | >10% | ||
| BRAF oncogenes | 30% | |||
| Follicular | t(2;3)(q13;p25) | PAX8-PPARG fusion | >40% | |
| Medullary | ||||
| Sporadic (75%) | RET activating mutations | >90% | ||
| Hereditary (25%) | Germline RET, MEN2A, or MEN2B mutations | >90% | Indication for screening for pheochromocytoma and screening family members | |
| Wilms' tumor, pediatric | Deletion 11p13 | WTI inactivation | 25% | Germline mutations occur in several syndromes. WT1 mutations also occur in sporadic tumors |
| Trisomy 12 | 40% | |||
For additional information on specific genes, see Online Mendelian Inheritance in Man (OMIM; www.ncbi.nlm.nih.gov).
aTrastuzumab (Herceptin) is a monoclonal antibody directed against the HER-2/neu receptor. Patients are selected for treatment by testing carcinomas with IHC or FISH.
Cetuximab (C225, Erbitux ™) is a monoclonal antibody directed against the EGFR receptor. A test has been approved by the FDA for the determination of EGFR (DakoCyomation, EGFR PharmDX). This test is not used for lung carcinomas (see note “d” below).
- Bcr-Abl tyrosine kinase: CML, ALL (Ph+)
- KIT tyrosine kinase: GIST, systemic mastocytosis, some types of AML
- PDGFR kinase: CMML, chronic eosinophilic leukemia, rare cases of GIST.
- The KIT protein (CD117) is encoded by the c-K/T proto-oncogene and is a transmembrane receptor protein with tyrosine kinase activity. Mutations may render KIT independent of its ligand, SCF (stem cell factor). Mutated proteins may or may not respond to therapy with imatinib. Wild-type KIT and KIT with mutations in the juxtamembrane domain (the intracellular segment between the transmembrane and tyrosine kinase domains) are found in GISTs and are sensitive to imatinib. Other tumor types are associated with mutations in the enzymatic domain and the altered protein is generally not sensitive to imatinib. Overexpression of the protein is detected by IHC.
Gefitinib (Iressa) is a tyrosine kinase inhibitor effective against a small subset of lung adenocarcinomas with specific activating mutations in EGFR. IHC for EGFR is not helpful for identifying carcinomas likely to respond to treatment.
Table 7-34.
Common cytogenetic changes in lymphomas and leukemias
| TUMOR TYPE | CYTOGENETIC CHANGES | MOLECULAR EVENTS | FREQUENCY | COMMENTS |
|---|---|---|---|---|
| Chronic leukemias and mastocytosis | ||||
| CML (Ph1) |
t(9;22)(q34;q11.2) Other variants or cryptic translocations |
BCR-ABL fusion (usually p210, but also p190 and p230 fusion proteins) | 90-95% |
|
| BCR-ABL fusion (usually p210, but also p190 and p230 fusion proteins) | 5-10% | |||
| CML, accelerated phase or blast phase | Additional changes: extra Ph, +8, or i(17)(q10) | 80% | May be myeloid (70%) or lymphoid (30%) | |
| Chronic myelomonocytic leukemia with eosinophilia | t(5;12)(q33;p13) | ETV6(also called TEL) -PDGRFbeta fusion | Rare | Excellent response to imatiniba |
| Chronic eosinophilic leukemia/hyper- eosinophilic syndrome |
Cryptic del(4)(q12) - interstitial 800 kb deletion | FIPILI-PDGFRalpha fusion | ∼50% | The fusion protein is an activated tyrosine kinase. Excellent response with the tyrosine kinase inhibitor imatiniba May be more common in infants and women. Excellent response to imatiniba |
| t(1;5)(q23;q33) | myorn egalin-PDGFRbeta fusion protein | ? Rare | ||
| Mastocytosis | c-KIT point mutations (Asp816Val) | 100% | CD117 (c-kit) is detected by IHC in normal and abnormal mast cells. The most common mutations do not result in proteins sensitive to imatinib Found in mastocytosis with associated eosinophilia. These patients do not have the typical c-KIT mutation. Excellent response to treatment with imatiniba |
|
| Cryptic del(4)(q12) - interstitial 800 kb deletion | FIPILI-PDGFRalpha fusion | ∼60% of patients with eosinophilia | ||
| Acute myeloid leukemia | ||||
| AML | Normal karyotype | 20% | ||
| FLT3 (13q12) internal tandem duplications (ITD, 20%) or point mutations (7%) | 20-30% of AML with normal karyotype | More common in monocytic AML (M5), less common in myeloblastic leukemia with maturation (M2) or erythroleukemia (M6). Less common in AML with cytogenetic changes (10%). Poor prognostic factor Results in an activated tyrosine kinase. Current trials are evaluating response to a kinase inhibitor, PKC412. |
||
| AML (M1, M2, or M4) | t(6;9)(p23;q34) | DEK-CAN fusion | 1% of all AML | Poor prognosis |
| FLT3 ITDs | 90% of this AML type | |||
| Acute myeloid leukemia | ||||
| AML with t(8;2l) (M2) |
t(8;2l)(q22;q22) | AMLI-ETO fusion | 5-12% of AML | 30% of cases of AML with karyotypic abnormalities and maturation. Maturation in neutrophilic lineage. Usually younger patients, good prognosis |
| c-KIT mutations | ∼50% of this AML type | Response to imatinib3 untested | ||
| Acute promyelocytic leukemia (M3, M3v.) |
t(IS;l7)(q22;q11-12) | PML-RARa fusion | 5-8% of AML (95-100% of APML) | Abnormal promyelocytes predominate. Usually occurs in adults in mid-life. Treatment with all trans-retinoic acid acts to differentiate the cells. Favorable prognosis |
| t(l I;l7)(q23;p2l) | PLZF-RARa fusion | |||
| t(S;l7)(q34;ql2) | NPM i-RARa fusion | |||
| t(l I;l7)(pl3;q2l) | NUMA-RARa fusion | |||
| FLT3 ITDs | 32% of APML | |||
| AML with inv or t(l6;l6) |
inv (I6)(pl 3)(q22) t(l6;l6)(pl3;q22) del(l6q) Other rare variants or cryptic translocations |
CBFbeta-MYHI1 fusion | 10-12% of AML (100% of M4EO) | Monocytic and granulocytic differentiation and abnormal eosinophils in the marrow. Usually younger patients. Favorable prognosis |
| c-KIT mutations | ∼50% of this AML type | Response to imatinib3 untested | ||
| AML with 1 Iq23 abnormalities | 1 Iq23 abnormalities | MLL fusion with numerous different partners | 5-6% of AML | Usually associated with monocytic features. Occurs in infants and in patients after therapy with topoisomerase II inhibitors. Intermediate prognosis |
| AML and MDS, therapy related | Sq-/7q-/12p-/20q- t(9;l l), t(l I;l9), t(6;l 1) Other less common changes |
Occurs after alkylating agents and/or radiation, usually 5 to 6 years after treatment. Poor prognosis | ||
| MLL balanced translocations | Occurs after DNA- topoisomerase II inhibitors, usually 3 years after treatment. Long-term prognosis unknown | |||
| B Cell | ||||
| Precursor B-lympho- blastic leukemia/lymphoblastic lymphoma (ALL) | t(9;22)(q34;ql 1.2) | BCR-ABL fusion (usually pi90 (esp. in children), but also p210 protein) | 5% of childhood ALL 20-25% of adult ALL | Philadelphia chromosome Poor prognosis |
| t with 1 Iq23 | MLL rearrangements | Poor prognosis. Usually infants | ||
| t(l2;2l)(pl3;q22) | TEL-AMLI fusion | >50% of childhood ALL or hyperdiploid | Good prognosis. This translocation is not detected by standard cytogenetics | |
| t(l;l9)(q23;pl 3.3) | PBXI-E2A fusion | 5-6% | Pre-B-ALL; most common translocation in childhood. Unfavorable but modified by therapy |
|
| Hypodiploid | Poor prognosis | |||
| Hyperdiploid >50 | Good prognosis (= DNA Index 1.16 to 1.6) | |||
| t(S;l4)(q3 I;q32) | IL3-IGH fusion | Poor prognosis | ||
| t(8;l4)(q24;q32) | MYC-IGH fusion | Good prognosis | ||
| t(2;8)(pl2;q24) | IGK-MYC fusion | Good prognosis | ||
| t(8;22)(q24;q11) | MYC-IGL fusion | Good prognosis | ||
| t(l7;l9)(q2l;pl 3) | HLF-E2A fusion | Poor prognosis | ||
| t(4;l I)(q2l;q23) | MLL-AF4 fusion | Poor prognosis | ||
| ALL, therapy related | Similar to therapy related AML | |||
| Small lymphocytic lymphoma/CLL | Trisomy 12 | 16% | Usually do not have lgVH mutations. Aggressive clinical course | |
| del(l lq22-23)—ATR | 18% | Poor prognosis Detected by FISH | ||
| del(13q14) —DBS319 | 55% | Usually do have lgVH mutations Long-term survival Detected by FISH | ||
| 17p — pS3 | 7% | Worse prognosis Detected by FISH | ||
| lgVH not mutated | 40-50% | Worse prognosis (<8 year median survival) | ||
| lgVH (mutated, >2% difference in nucleotide sequence) | 50-60% | Better prognosis (median survival >24 years) | ||
| Lymphoplasmacytic lymphoma/Waldenström's macroglobulinemia | t(9;l4)(pl 3;q32) | PAX-5-IGH fusion | 50% | This rearrangement may be less common in cases associated with Waldenström's macroglobulinemia or if node-based |
| Mantle cell lymphoma | t(l I;l4)(q13;q32) | CCND l-IGH fusion ATM point mutations | >95% | Overexpression of cyclin D1 detected by IHC |
| Marginal zone lymphoma (MALT) | +3 | 60% | ||
| t(l;l4)(p2l;q32) | BCL-IO-IGH fusion | |||
| t(l I;l8)(q2l;q2l) | API2-MALT1 fusion | 25-50% | ||
| t(l I;l4)(q2l;q32) | MALTI-IGH fusion | |||
| Follicular lymphoma | t(l4;l8)(q32;q2l) | IGH-BCL-2 fusion | 70-95% | |
| t(2;l8)(pl2;q2l) | IGK-BCL-2 fusion | Rare | ||
| Burkitt's lymphoma and | t(8;l4)(q24;q32) | MYC-IGH fusion | 85% | |
| Burkitt-like lymphoma | t(2;8)(pl2;q24) | MYC-IGK fusion | Rare | |
| t(8;22)(q24;q11) | MYC-IGL fusion | Rare | ||
| Mediastinal (thymic) large B-cell lymphoma | 9p+ | REL amplification | ||
| Diffuse large B-cell lymphoma | t(3q27;v) | BCL6 translocations with many partners | 30% | BCL6 is detected by IHC in most cases, BCL2 in some cases |
| t(l4;l8)(q32;q2l) | BCL2-IGH fusion | 20-30% | ||
| Hairy cell leukemia | No consistent changes | |||
| Primary effusion lymphoma | No consistent changes | |||
| Plasmacytoma/myeloma | t(l I;l4)(ql 3;q32) | CCND l-IGH fusion | Best prognosis | |
| t(6;l4)(p2l;q32) | CCND3-IGH fusion | |||
| t(4;14)(p16;q32) | FGF23-IGH fusion | Adverse prognosis | ||
| t(l4;l6)(q32;q23) | IGH-MAF fusion | Adverse prognosis | ||
| Monosomy I3/I3q- | 15-40% | |||
| T Cell | ||||
| Precursor lymphoblastic leukemia/lymphoblastic lymphoma | Translocations involving TCR alpha, beta, delta, and gamma and partner genes MYC, TALI, RBTNI, RBTN2, HOXI/, and LCK | 30% | ||
| del(l) | Tail (small deletion) | 30% | Adolescents | |
| t(1;14) | Tall-TCRdelta fusion | 25% | Adolescents | |
| t(5;14) | HOXI IL2-TCRdelta fusion | >30% | Young children | |
| del(9p) | CDKN2A deletion | |||
| T-cell prolymphocytic leukemia | inv (q1 I)(q32) | TCRa/β-TCLI & TCLIb fusion | 80% | |
| t(l4;l4)(ql I;q32) | TCRa/β-TCLI & TCLIb fusion | 10% | ||
| t(7;l4)(q3S;q32.1) chrom 8 abnormalities | TCRβ-TCL IA fusion | 70-80% | ||
| Adult T-cell lymphoma/leukemia | No consistent changes | |||
| Mycosis fungoides and Sézary syndrome | No consistent changes | |||
| Peripheral T-cell lymphoma, NOS | No consistent changes | |||
| Hepatosplenic T-cell lymphoma | i(7q)(ql0) | 100% | ||
| Panniculitis-like T-cell lymphoma | No consistent changes | |||
| Angioimmunoblastic lymphoma | Trisomy 3, trisomy 5, + X | |||
| Enteropathy-type T-cell lymphoma | No consistent changes. | |||
| Anaplastic large cell lymphoma (CD30+) | t(2;S)(p23;q3S) | NPMI-ALK fusion protein (p80) | 70-80% | ALK detected by IHC in nucleus, nucleolus, and cytoplasm |
| 2p23 rearrangements | ALK fusion with other partners | ALK detected by IHC in cytoplasm | ||
| Extranodal NK/T-cell lymphoma, nasal type | No consistent changes. | |||
| Blastic NK-cell lymphoma | No consistent changes. | |||
For additional information on specific genes, see Online Mendelian Inheritance in Man (OMIM; www.ncbi.nlm.nih.gov).
- Bcr-Abl tyrosine kinase: CML, ALL (Ph+)
- KIT tyrosine kinase: GIST, systemic mastocytosis, some types of AML
- PDGFR kinase: CMML, chronic eosinophilic leukemia, rare cases of GIST
- The KIT protein is encoded by the c-KIT proto-oncogene and is a transmembrane receptor protein with tyrosine kinase activity. Mutations may render KIT independent of its ligand, SCF (stem cell factor). Mutated proteins may or may not respond to therapy with imatinib. Wild-type KIT and KIT with mutations in the juxtamembrane domain (the intracellular segment between the transmembrane and tyrosine kinase domains) are found in GISTs and are sensitive to imatinib. Other tumor types are associated with mutations in the enzymatic domain and the altered protein is generally not sensitive to imatinib.
Indications
-
•
B-cell proliferations: clonal rearrangements of the immunoglobulin heavy and light chain genes; specific translocations (see Table 7-34).
-
•
T-cell proliferations: rearrangements of the γ and β T-cell receptor genes.
-
•
Leukemias (see Table 7-34).
-
•
Post-transplant lymphoproliferative disorders: clonal populations of EBV-infected cells.
-
•
Oligodendrogliomas: PCR-based LOH analysis for 1p/19q deletions.
Method of submitting tissue
Fresh or frozen tissue (e.g., snap-frozen tissue) as well as fluids may be used. Cytologic preparations can be used for FISH.
Results
The results are usually either reported separately or incorporated into the surgical pathology report.
CYTOGENETICS
Cytogenetic studies have been demonstrated to be useful in several areas important to pathology:
Tumor classification, particularly sarcomas, lymphomas, brain tumors, and other unusual tumors (Ewing's sarcoma, synovial sarcoma).
Benign versus malignant lesions, for example:
-
•
Reactive mesothelial cells versus mesothelioma
-
•
Lipoma versus liposarcoma.
Prognosis, for example in neuroblastoma and multiple myeloma.
Research: Translocations are common to many tumors and usually identify genes important to the pathogenesis of the tumor.
Cells may be cultured to perform complete karyotype analysis or tissues can be analyzed for specific chromosomal alterations by fluorescence in situ hybridization (FISH). FISH studies can be performed on cultured cells, cytology preparations, and fixed and embedded tissues.
Indications
Cytogenetic studies are indicated for soft tissue tumors, mesotheliomas (tissue or pleural fluid), unusual tumors, poorly differentiated tumors, all subcutaneous lipomas larger than 5 cm, all subfascial lipomas (for karyotype), and oligodendrogliomas (for FISH).
Method for submitting tissue
Tissue for karyotyping must be fresh, viable, and relatively sterile. However, tissue may be submitted even if it has not been handled under strictly sterile conditions (contamination is not usually a problem). If specimens are to be held overnight, the tissue should be minced (into 1-mm cubes) in a sterile specimen container, covered with culture medium, and held overnight in the refrigerator. Fluids may also be submitted for analysis (especially pleural effusions with a suspicion of mesothelioma).
Results
The results of the cytogenetic analysis should be incorporated into the final diagnosis.
Tumors and diseases associated with germline mutations
The following features are suggestive of a hereditary susceptibility to cancer:
-
•
Two or more close relatives on the same side of the family with cancer
-
•
Evidence of autosomal dominant transmission
-
•
Early development of cancer in the patient and relatives (in general, under 50 years of age)
-
•
Multiple primary cancers
-
•
Multiple types of cancers
-
•
Unusual pathologic features of tumors (Table 7-35 )
-
•
A constellation of tumors suggestive of a specific syndrome (Table 7-36 ).
Pathologists can aid in the detection of hereditary carcinomas by being aware of the types and pathologic characteristics of carcinomas associated with these syndromes. Patients with germline mutations are important to identify in order to:
-
•
Screen patients for other common tumors or other components of the disease
-
•
Consider prophylactic surgery or preventive interventions
-
•
Offer screening to family members at risk and genetic counseling.
Although the sporadic forms of cancers in general far outnumber cases associated with germline mutations, in some cases the appearance or site of a carcinoma is highly suggestive of a known syndrome and further investigation may be warranted.
Table 7-35.
Pathologic features of tumors and diseases suggestive of a germline mutation
| TYPE OF TUMOR | PERCENTAGE OF CASES RELATED TO KNOWN GERMLINE MUTATIONS | SYNDROMES/GENES INVOLVED | CLUES FOR THE PATHOLOGIST |
|---|---|---|---|
| Adrenocortical carcinoma in children | 50-100% | Li-Fraumeni, Beckwith-Wiedemann, MEN1 | Unusual occurrence in a child |
| Angiomyolipoma of kidney | 20% | Tuberous sclerosis | Patients may be screened for other features of tuberous sclerosis |
| Basal cell carcinoma | Rare if solitary | Nevoid basal cell carcinoma syndrome | Risk of a mutation is increased if multiple or if tumor occurs at <30 years of age |
| Breast cancer, poorly differentiated, ER negativea | >25% if <35 years old, <10% if >35 years old | BRCAI | BRCA1 cancers are more likely to have “medullary” features, and be ER- PR- HER-2/neu-.BRCAI mutation more likely if patient has a family history or has bilateral cancer |
| Breast cancer, male | 4-14% | BRCA2 | Cancers are of no specific type |
| Colorectal carcinoma, poorly differentiated, mucinous, or with prominent lympho- cytic infiltrate |
∼10–15% overall, ∼80% if patient is <40 |
HNPCC |
HNPCC carcinomas are more likely right-sided (two thirds), poorly differentiated (“medullary”), mucinous, signet ring, lymphocytic infiltrate. |
| IHC for MSH2 and MLH1 can be used to detect many, but not all, cases, but MLH1 may also be absent in sporadic cases | |||
| GI neuroendocrine tumors: |
MEN! mutations |
MEN! mutations are also found in 15-70% of sporadic neuroendocrine tumors |
|
| Somatostatinoma | 45% | ||
| PPoma | 18-44% | ||
| Non-functioning | 18-44% | ||
| Gastrinoma | 20-25% | ||
| Glucagonoma | 1-20% | ||
| VIPoma | 6% | ||
| Insulinoma | 4-5% | ||
| Carcinoid | Rare | ||
| Hirschsprung's disease | 20-40% | MEN2A (RET mutations in codons 609, 618, 620) | |
| Juvenile (hamar- tomatous) polyps | Rare if solitary | Juvenile polyposis syndrome (JPS) | Suspect JPS if there are >5 polyps, if present throughout the GI tract, or if there is a family history of juvenile polyps |
| Medullary carcinoma of the thyroid |
25% |
MEN2A, MEN2B, Familial medullary carcinoma (RET mutations) |
May be multiple and associated with C-cell hyperplasia |
| Cancers in occur in children in MEN2B and in young adults in MEN2A | |||
| Medulloblastoma | Rare (?) | Nevoid basal cell carcinoma syndrome | If <3 years of age or of desmoplastic type, risk of mutation is increased |
| Myxoma, cardiac | <5% | Carney complex | Increased likelihood if multiple, right sided, and/or recurrent and in young patients (<30) |
| Neurofibromas | ∼10% if solitary but > 90% if plexiform | Neurofibromatosis type 1 | Increased risk if there are >2 neurofibromas or one plexiform neurofibroma |
| Ovarian carcinoma |
Rare |
BRCAI, BRCA2 |
Increased risk if there is a history of breast cancer |
| BRCAI-associated carcinomas are more likely to be serous in type | |||
| Pheochromocytoma | 30% of all cases, 59% if patient is <18, 84% if bilateral | MEN2A, MEN2B, VHL, isolated familial pheochromocytoma | Multiple tumors, hyperplasia of the medulla |
| Primary pigmented nodular adreno- cortical disease |
>90% |
Carney complex |
May present with Cushing's syndrome |
| Most are associated with germline mutations, but patients may not have other manifestations of the Carney complex | |||
| Retinoblastoma | 40% of all cases, 100% if bilateral or with a positive family history | RB mutations (13q14.1-q14.2) | |
| Rhabdomyoma of heart in infants | 50% | Tuberous sclerosis | |
| Sarcoma, children | 7-33% | Li-Fraumeni, basal cell nevus syndrome, neurofibromatosis type 1, pleuropulmonary blastoma syndrome | |
| Sebaceous carcinoma | ∼10% if ocular, 40% if above the chin, 80% if elsewhere | HNPCC | Increased likelihood if the tumor has cystic degeneration or features of keratoacanthoma |
| Usually due to germline MSH2 mutations | |||
| Schwannoma, psammomatous melanotic | >50% | Carney complex | Higher likelihood if patient is young (<30 years) and/or multiple tumors present |
| Schwannoma, vestibular | 5% | Neurofibromatosis type 2 | Risk is increased if the patient is <30 or if there is bilateral involvement |
| Sporadic cases almost all have somatic NF2 mutations | |||
| Sertoli cell tumor, large-cell calcifying | 25-35% | Carney complex, Peutz-Jeghers | Most are bilateral and multifocal in young patients. Rarely malignant |
| Trichilemmoma, facial, multiple | ∼80% | PTEN | Sporadic tumors also have loss of PTEN, which can be shown by IHC. |
| Wilms' tumor | 10-15% | Germline mutations in WTI (11p13) | Nephrogenic rests are present and may be extensive |
| |||
| Associated with WAGR syndrome (Wilms' tumor, aniridia, GU anomalies, mental retardation) and Denys-Drash syndrome | |||
See reference 17 for additional information relating pathologic characteristics to risk of a BRCAI mutation.
Table 7-36.
Hereditary syndromes associated with multiple tumors
| SYNDROME | GERMLINE MUTATIONS | TUMORS (% OF PATIENTS DEVELOPING TUMOR) | COMMENTS |
|---|---|---|---|
| Beckwith- | 11p15 abnormalities (loss of methylation, uniparental disomy, mutations in CDKN1C) |
Wilms' tumor, neuroblastoma, hepatoblastoma, adrenocortical carcinoma, rhabdomyosarcoma |
Macrosomia, macroglossia, visceromegaly, ear creases and pits, omphalocele, hypoglycemia |
| Wiedemann syndrome | |||
| BRCA1 and 2 | BRCAI (17q21),BRCA2 (13q12.3) | Breast (85%), ovary (BRCA1 63%, BRCA2 27%), prostate carcinoma, others | BRCA1 breast cancers are more often poorly differentiated, have medullary features, are ER- PR- HER-2/neu-, and have p53 mutations. Ovarian carcinomas are generally serous (90%), high grade, and bilateral. BRCA2 cancers do not have specific pathologic features |
| Carney complex | Type 1 (CNC1): PRKAR1A (17q23-24) | Myxomas (cardiac, cutaneous, breast), primary pigmented nodular adrenocortical disease | |
| Type 2 (CNC2): locus at 2p16 | (25%), large-cell calcifying Sertoli cell tumors (>90% males), multiple thyroid nodules or carcinoma (75%), growth hormone producing pituitary adenoma (10%), psammomatous melanotic schwannoma (10%), breast duct adenomas, osteochondromyxoma of bone | ||
|
|||
| Carney triad |
Unknown |
Gastric gastrointestinal stromal tumor, pulmonary chondroma, extra-adrenal paraganglioma | Most patients are young and female. Only 22% have all three tumors. Most family members are not affected |
| Also esophageal leiomyomas and adrenocortical tumors | |||
| Familial adenomatous polyposis (FAP; including Gardner syndrome and Turcot syndrome) | APC (5q21-22) | Colorectal carcinoma, upper GI carcinoma, desmoid, osteoma, thyroid, brain (one third to two thirds are medulloblastomas—Turcot syndrome) | |
| Familial medullary thyroid carcinoma | RET mutations in codons 10, 11, 13, 14 (10q11.2) | Medullary thyroid carcinoma | Cancers usually occur in adults |
| Hereditary diffuse gastric cancer syndrome | CDHI (e-cadherin) (16q22.1) | Signet ring cell carcinoma of the stomach (67% men, 83% women), lobular carcinoma of the breast (39% women) | 50% of sporadic signet ring cell carcinomas have CDHI somatic mutations and all show loss of e-cadherin by IHC |
| Hereditary non- polyposis syndrome |
Mismatch repair genes: MSH2 (2p22-p21) (40%), MLHI (3p21.3) (40%), MSH6 (2p16) (5-7%), PMS2 (7p22) (rare) |
Colon carcinoma (80%), endometrial carcinoma (20-60%), ovarian carcinoma (9-12%), stomach carcinoma (11–19%), hepatobiliary tumors (2-7%), transitional cell carcinoma (4-5%, esp. ureter and renal pelvis), small bowel tumors (1-4%), lymphoma (rare) | Colon carcinomas are more likely (overall, 66%) to be on the right side, poorly differentiated (“medullary”), mucinous, signet ring, or undifferentiated, with a prominent lymphocytic infiltrate |
| Sebaceous skin tumors, adenomas, epitheliomas, carcinoma, keratoacanthomas (Muir-Torre, usually MSH2) | IHC can be used to detect the absence of MSH2 (usually due to germline mutations) and MLHI (can be due to germline mutations, epigenetic changes, or less commonly, somatic mutations) in many patients MSI testing is also used | ||
| Juvenile polyposis syndrome | MADH4 (or SMAD4) (18q21.10) (15%) or BMPRIA (10q22.3) (25%) | Hamartomatous (juvenile) polyps, GI carcinomas | |
| Li-Fraumeni | p53 (17p13.1), rarely CHEK2 (22q12.1) | Sarcomas, breast cancer, leukemia, osteosarcomas, brain tumors, adrenocortical carcinoma, others | |
| MEN1 | MENI (11q13) | Pituitary adenoma, pancreatic islet cell tumors, parathyroid adenomas, adrenocortical tumors, carcinoids, lipomas | MEN! mutations also occur in 15-70% of sporadic neuroendocrine tumors |
| MEN2A |
RET exon 10 and 11 missense mutations (10q11.2) |
Medullary thyroid carcinoma (95%), hyperplasia of the parathyroids (15-30%), pheochromo- cytoma (50%), ganglioneuromatosis of GI tract | Specific mutations correlate with age at development of medullary thyroid carcinoma |
| Subsets of patients have Hirschsprung's disease or cutaneous lichen amyloidosus | |||
| MEN2B |
RET missense mutation in exon 16 (10q11.2) |
Medullary thyroid carcinoma (100%), pheochromocytoma (50%) | Marfanoid habitus, distinctive facies |
| Mucosal neuromas of lips and tongue | |||
| Nevoid basal cell carcinoma syndrome (Gorlin syndrome) | PTCH (9q22.3) | Basal cell carcinomas (90%), odontogenic keratocysts (90%), cardiac or ovarian fibromas (20%), medulloblastoma in childhood (5%) | Macrocephaly, skeletal anomalies, palmar or plantar pits, calcification of falx (90%) |
| Neurofibromatosis type 1 | NFI (17q11.2) | Neurofibromas (esp. plexiform) (100%), optic gliomas, adrenal ganglioneuromas, pheochromo- cytoma (0.1-6%), MPNST (10%), leukemia, ganglioneuromatosis of the GI tract | Café-au-lait macules (95%), iris hamartomas (Lisch nodules), axillary freckling |
| Neurofibromatosis type 2 | NF2 (22q12.2) | Bilateral vestibular schwannomas (100%, 40% have lobular pattern), schwannomas of other nerves, meningiomas (50%, often fibroblastic) | |
| Peutz-Jeghers (hamartomatous polyp) syndrome |
LKBI/STKI1 (19p13.3) |
Colon, breast, stomach, pancreas, small bowel, thyroid, lung, uterus, sex cord stromal tumors, calcifying Sertoli cell tumors | Perioral pigmentation |
| Hamartomatous polyps of GI tract | |||
| Pheochromo- cytoma or paraganglioma, familial | SDHB (1p36.1-p35), SDHD (11q23) SDHC (1q21) (paraganglioma) | Pheochromocytoma, paraganglioma | Patients are more commonly young (<40), with multifocal adrenal tumors, or extra- adrenal disease |
| SDHD is imprinted and only confers susceptibility after paternal transmission | |||
| PTEN hamartoma syndrome (including 80% of Cowden's syndrome, 50-60% of Bannayan-Riley -Ruvalcaba syndrome) |
PTEN (10q23.31) |
Breast cancer (25 to 50%), thyroid carcinoma (10%, esp. follicular), endometrial carcinoma (5-10%), hamartomatous polyps of GI tract | Macrocephaly (megalencephaly, 97th percentile), Lhermitte-Duclos disease |
| Multiple facial trichilemmomas, acral keratosis, oral papillomatous lesions, mucosal lesions | |||
| Tuberous sclerosis |
TSCI (9q34), TSC2 (16p13.3) |
Subependymal glial nodules (90%), cortical or subcortical tubers (70%), angiomyolipoma of kidney (70%), lymphangiomyomatosis of lung (1-6%), rhabdomyoma of heart (47-67%) | Seizures (80%), developmental delay or retardation (50%) |
| |||
| Von Hippel- Lindau (VHL) | VHL (3p26-p25) | Hemangioblastomas (retinal, cerebellar, spinal cord) (80%), renal cell carcinoma (40%), renal cysts, pancreatic cysts, Pheochromocytoma, endolymphatic sac tumors (10%), epididymal cystadenomas | |
For additional information on most syndromes, see http://www.genetests.org/and Online Mendelian Inheritance in Man (OMIM; www.ncbi.nlm.nih.gov).
ANALYTICAL CYTOLOGY (FLOW CYTOMETRY)
Flow cytometers analyze populations of thousands of disaggregated cells as they pass by stationary detectors. Cell size and cytoplasmic granularity can be measured as well as DNA content and the presence or absence of immunohistochemical markers added to the cell suspension. Newer techniques can analyze three or more features simultaneously to divide cells into unique populations. DNA content can be used to determine the number of cells in S-phase (a measure of proliferation—S-phase fraction). Because cells are not visualized by this technique, it is important to be sure that only lesional tissue is submitted.
Indications for ploidy and S-phase analysis
-
•
Hydatidiform moles: complete (diploid), partial (triploid).
-
•
Some carcinomas: DNA ploidy and S-phase fraction have been reported to be of prognostic significance for some carcinomas (e.g., colon, breast, and prostate) but the analysis is not routinely performed at all institutions or used by all oncologists.
Indications for cell surface marker analysis
-
•
Lymphomas.
-
•
Leukemias.
Method for submitting tissue
Single cell suspensions are necessary for analysis. For fresh tissues, cells must be viable. Fresh tissue (approximately 0.3 to 0.5 cm3) is placed in a specimen container and kept moist with HBSS. Tissues can be held overnight in the refrigerator.
Formalin-fixed paraffin-embedded sections may also be used for DNA ploidy analysis by the Hedley method, although the results are not as satisfactory due to nuclear fragmentation.
Results
The results are usually incorporated into the final surgical pathology report.
CYTOLOGIC PREPARATIONS FROM SURGICAL SPECIMENS
Cytologic preparations of surgical specimens often provide additional information.
Intraoperative diagnosis.
Touch preps or smears are especially valuable for:
-
•
Infectious cases (to avoid contamination of the cryostat and aerosolization of infectious agents)
-
•
Neuropathology cases, for diagnosis and for the performance of cytogenetic (FISH) analysis
-
•
Tumors (for excellent cytologic detail, especially lymphomas and papillary carcinomas of the thyroid).
Special stains.
Stains for microorganisms can be performed the same day on cytologic smears of specimens from critically ill patients. Do not submit air dried smears of infectious cases for staining as the unfixed material may constitute a hazard to laboratory personnel.
Fat is dissolved during routine processing, but can be demonstrated with fat stains on air dried slides.
Genetic studies (FISH).
Nuclei are intact in touch preparations, unlike tissue sections in which the only partial nuclei are present. This feature makes touch preparations superior for techniques such as FISH and image analysis.
It is always a useful exercise to look at cytology preparations and the corresponding surgical specimen to learn the comparative morphology of these techniques.
SPECIMEN RADIOGRAPHY
Specimen radiographs are often preferred to patient radiographs:
-
•
A permanent record of the radiograph can be kept with the case.
-
•
A radiograph of the specimen may reveal more details of the underlying process (e.g., fewer structures may be present to complicate the appearance).
-
•
A significant time interval may have elapsed between the patient radiograph and the surgical excision.
-
•
The radiograph often indicates sites that are important to examine histologically (tumor invasion into a rib or microcalcifications in a breast biopsy).
-
•
The specimen radiograph can confirm that the clinical lesion was removed.
Indications
-
•
Tumors of bone and cartilage.
-
•
Tumors invading into bone.
-
•
Avascular necrosis.
-
•
All bioprosthetic heart valves (to document the degree of calcification).
-
•
Breast biopsies or mastectomies performed for mammographic lesions that cannot be located grossly. Paraffin blocks of breast tissue can be radiographed if microcalcifications were seen by specimen radiography but not in histologic sections and were not identified prior to processing.
Calcifications can dissolve in formalin over several days. If the demonstration of calcifications is important (e.g., mammographically detected calcifications) it is preferable to process the tissue within 1 to 2 days. If processing is to be delayed, the tissue can be stored in ethanol.
Method
Radiographic equipment is available in radiology departments and in some pathology departments. The specimen may be placed on a piece of wax paper (to keep the surfaces clean) lying on the film. Specimens can be radiographed after decalcification (not all calcium is removed) but best results are obtained on fresh undecalcified specimens. Lungs should not be inflated prior to radiography.
If the specimen is small, two exposures at different settings or at different angles may be useful. Lead sheets can be used to allow two exposures on one piece of film.
If the film is too dark (overexposed), the exposure is too high and a lower setting should be tried. If the film is too light (i.e., unexposed) the exposure is too low and a higher setting is indicated.
Special injection techniques with radiocontrast media are available for unusual specimens such as a recipient lung with pulmonary hypertension or vascular ectasia of the bowel.
Octreotide and sentinel nodes.
Labeled compounds are sometimes used to localize certain types of tumors (generally neuroendocrine) or sentinel lymph nodes. The patient is injected with the isotope prior to surgery and the surgeon uses a handheld probe to identify the labeled tissue. The amount of radioactivity in the tissue is small; generally it does not pose a hazard to pathologists handling the tissue and does not need special disposal methods. However, each pathology department should consult with the radiation safety department to ensure appropriate handling of such tissues. In some cases, if a gross lesion is not present corresponding to the area of octreotide uptake, specimens can be imaged using a gamma camera.
Results
The radiographs are documented in the gross description and any information gained from the radiograph is incorporated into the surgical pathology report.
TISSUE FOR RESEARCH: TUMOR BANK
The pathology department is a unique resource for researchers who need human tissues. The pathologist plays a key role as patient advocate and diagnostician in order to provide appropriate human tissues for biologic research. Most hospitals have a policy that allows the release of tissue for research if it would otherwise be discarded. Therefore, tissue is never given away for research until all necessary tissue has been taken for diagnosis. Tissue from primary diagnostic breast biopsies and open lung biopsies without gross lesions should not be given away. It is in the best interest of the patient that a pathologist evaluates the specimen rather than have tissue given away by a nonpathologist who is not aware of what is needed for diagnosis.
Indications
By request of researchers who have obtained permission from the hospital Human Studies Committee.
Method
Adequate information must be provided by the clinician to allow the pathologist to determine how much of the tissue is needed for diagnostic purposes. Research laboratories should provide containers for the transport of specimens. The name of the laboratory, the type of tissue, and the amount of tissue allocated for research must be carefully documented. Tissue should never be given away if there is any question as to the need for the tissue for diagnostic purposes. In some cases it may be preferable (or possibly required) to withhold the name or other identifiers of the patient for medical confidentiality.
MICROBIOLOGIC CULTURE AND SMEARS
The investigation of infectious disease by culture is complementary to its investigation by histologic sections (Table 7-37 ).
Table 7-37.
Identification of infectious diseases
| CULTURE | HISTOLOGIC SECTIONS |
|---|---|
| Can be performed on aspirates, swabs, fluids, or tissues | Requires surgical excision of tissues |
| Cultures amplify the number of organisms present, allowing them to be recognized | Organisms may be rare, or not seen in tissue sections |
| The specific organism can be identified and tested for drug susceptibility | Categories of organisms can be recognized but specific identification may not be possible |
| Some organisms cannot be cultured | Many organisms can be identified that will not grow in culture or that require long culture times (e.g., Mycobacterium tuberculosis) |
| It may be difficult to exclude contamination for a positive culture | Morphologic evidence of an inflammatory response provides evidence for a clinical infection. The location of the infection may be of diagnostic importance (e.g., cellulitis versus necrotizing fasciitis or superficial colonization of devitalized tissue versus deep infections involving viable tissues) |
Indications
-
•
Suspected infectious processes.
-
•
Suspected sarcoid to exclude an infectious process.
Method
Tissue is kept as sterile as possible. Suture removal kits are a convenient source of sterile scissors and forceps. Serially section the specimen to determine whether there are focal lesions. Place representative sections in a sterile specimen container. Label with the patient's name and unit number, patient's physician, type of specimen, collection date, and time of collection (required for JCAHO accreditation).
Three different types of culture are often requested (requiring three different requisition forms):
1. Routine culture.
The usual request for routine specimens would be:
-
•
Bacteria (only includes aerobic culture)
-
•
Mycobacteria
-
•
Fungal.
Other organisms require special culture techniques and must be specifically requested:
-
•
Anaerobic bacteria
-
•
Salmonella, Shigella, and Campylobacter
-
•
Nocardia
-
•
Neisseria gonorrhoeae
-
•
Brucella
-
•
Legionella
-
•
Francisella tularensis
-
•
Helicobacter.
2. Viral culture.
CMV, varicella zoster, adenovirus, and herpes simplex are most commonly requested. Cultures for influenza A and B, Respiratory synctial virus, and parainfluenza require special techniques.
3. Mycoplasma.
Usually requires special cultures. Occasionally, mycoplasma can be detected on anaerobic cultures, but this is not the optimal means for identifying this organism.
Results
The results are generally reported by the microbiology laboratory. It is helpful to correlate the results with the pathologic findings, when possible.
REFERENCES
- 1.Werner M, Chott A, Fabiano A, Battifora H. Effect of formalin fixation and processing on immunohistochemistry. Am J Surg Pathol. 2000;24:1016–1019. doi: 10.1097/00000478-200007000-00014. [DOI] [PubMed] [Google Scholar]
- 2.Arber JM, Arber DA, Jenkins KA, Battifora H. Effect of decalcification and fixation in paraffin-section immunohistochemistry. Appl Immunohistochem. 1996;4:241–248. [Google Scholar]
- 3.DiVito KA, Charette LA, Rimm DL, Camp RL. Long-term preservation of antigenicity on tissue microarrays. Lab Invest. 2004;84:1071–1078. doi: 10.1038/labinvest.3700131. [DOI] [PubMed] [Google Scholar]
- 4.Jacobs TW, Prioleau JE, Stillman IE, Schnitt SJ. Loss of tumor marker immunostaining intensity on stored paraffin slides of breast cancer. J Natl Cancer Inst. 1996;88:1054–1059. doi: 10.1093/jnci/88.15.1054. [DOI] [PubMed] [Google Scholar]
- 5.Bertheau P, Cazals-Hatem D, Meignin V. Variability of immunohistochemical reactivity on paraffin slides. J Clin Pathol. 1998;51:370–374. doi: 10.1136/jcp.51.5.370. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.van den Broek LJ, van de Vijver MJ. Assessment of problems in diagnostic and research immunohistochemistry associated with epitope instability in stored paraffin sections. Appl Immunohistochem Mol Morphol. 2000;8:316–321. [PubMed] [Google Scholar]
- 7.Battifora H. Assessment of antigen damage in immunohistochemistry, the vimentin internal control. Am J Clin Pathol. 1991;6:669–671. doi: 10.1093/ajcp/96.5.669. [DOI] [PubMed] [Google Scholar]
- 8.Chu PG, Weiss LM. Keratin expression in human tissues and neoplasms. Histopathology. 2002;40:403–439. doi: 10.1046/j.1365-2559.2002.01387.x. [DOI] [PubMed] [Google Scholar]
- 9.Gyure KA, Morrison AC. Cytokeratin 7 and 20 expression in choroid plexus tumors: utility in differentiating these neoplasms from metastatic carcinomas. Mod Pathol. 2000;13:638–643. doi: 10.1038/modpathol.3880111. [DOI] [PubMed] [Google Scholar]
- 10.Wang NP, Zee S, Zarbo RJ, Bacchi CE, Gown AM. Coordinate expression of cytokeratins 7 and 20 defines unique subsets of carcinomas. Appl Immunohistochem. 1995;3:99–107. [Google Scholar]
- 11.Mierau GW, Berry PJ, Malott RL. Appraisal of the comparative utility of immunohistochemistry and electron microscopy in the diagnosis of childhood round cell tumors. Ultrastruct Pathol. 1996;20:507–517. doi: 10.3109/01913129609016355. [DOI] [PubMed] [Google Scholar]
- 12.Varadhachary GR, Abbruzzese JL, Lenzi R. Diagnostic strategies for unknown primary cancer. Cancer. 2004;100:1776–1785. doi: 10.1002/cncr.20202. [DOI] [PubMed] [Google Scholar]
- 13.Corson J. Pathology of mesothelioma. Thorac Surg Clin. 2004;14:447–460. doi: 10.1016/j.thorsurg.2004.06.007. [DOI] [PubMed] [Google Scholar]
- 14.Banks PM. Incorporation of immunostaining data in anatomic pathology reports. Am J Surg Pathol. 1992;16:808. doi: 10.1097/00000478-199208000-00011. [DOI] [PubMed] [Google Scholar]
- 15.Electron microscopy of tumors. Semin Diagn Pathol 20: 2003. The entire February 2003 issue is devoted to EM.
- 16.Wittel VA, Goel A, Varshney GC, Batra SK. Mucin antibodies – new tools in diagnosis and therapy of cancer. Front Biosci. 2001;6(D):1296–1310. doi: 10.2741/wittel. [DOI] [PubMed] [Google Scholar]
- 17.Lakhani SR. The pathology of familial breast cancer: predictive value of immunohistochemical markers estrogen receptor, progesterone receptor, HER-2, and p53 in patients with mutations in BRCA1 and BRCA2. J Clin Oncol. 2002;20:2310–2318. doi: 10.1200/JCO.2002.09.023. [DOI] [PubMed] [Google Scholar]
