Immunohistochemistry

Last updated
Main staining patterns on chromogenic immunohistochemistry. Main staining patterns on immunohistochemistry.jpg
Main staining patterns on chromogenic immunohistochemistry.
Immunofluorescence of human skin using an anti-IgA antibody. The skin is from a patient with Henoch-Schonlein purpura: IgA deposits are found in the walls of small superficial capillaries (yellow arrows). The pale wavy green area on top is the epidermis, the bottom fibrous area is the dermis. HSP IF IgA.jpg
Immunofluorescence of human skin using an anti-IgA antibody. The skin is from a patient with Henoch–Schönlein purpura: IgA deposits are found in the walls of small superficial capillaries (yellow arrows). The pale wavy green area on top is the epidermis, the bottom fibrous area is the dermis.

Immunohistochemistry (IHC) is a form of immunostaining. It involves the process of selectively identifying antigens (proteins) in cells and tissue, by exploiting the principle of antibodies binding specifically to antigens in biological tissues. Albert Hewett Coons, Ernest Berliner, Norman Jones and Hugh J Creech was the first to develop immunofluorescence in 1941. This led to the later development of immunohistochemistry. [1] [2]

Contents

Immunohistochemical staining is widely used in the diagnosis of abnormal cells such as those found in cancerous tumors. In some cancer cells certain tumor antigens are expressed which make it possible to detect. Immunohistochemistry is also widely used in basic research, to understand the distribution and localization of biomarkers and differentially expressed proteins in different parts of a biological tissue. [3]

Sample preparation

IHC can be performed on tissue that has been fixed and embedded in paraffin, but also cryopreservated (frozen) tissue. Based on the way the tissue is preserved, there are different steps to prepare the tissue for IHC, but the general method includes proper fixation, antigen retrieval incubation with primary antibody, then incubation with secondary antibody. [4] [5]

Tissue preparation and fixation

Fixation of the tissue is important to preserve the tissue and maintaining cellular morphology. The fixation formula, ratio of fixative to tissue and time in the fixative, will affect the result. The fixation solution (fixative) is often 10% neutral buffer formalin (NBF). Normal fixation time is 24 hours in room temperature. The ratio of fixative to tissue ranges from 1:1 to 1:20. After the tissue is fixed it can be embedded in paraffin wax. [4] [5]

For frozen sections, fixation is usually performed after sectioning if not new antibodies are going to be tested. Then acetone or NBF can be used. [5]

Sectioning

Sectioning of the tissue sample is done using a microtome. For paraffin embedded tissue 4 μm is normal thickness, and for frozen sections 4 – 6 μm. [5] The thickness of the sliced sections matters, and is an important factor in IHC. If you compare a section of brain tissue measuring 4 μm with a section measuring 7 μm, some of what you see in the 7 μm thick section might be lacking in the 4 μm section. This shows the importance of detailed methods related to this methodology. [6] The paraffin embedded tissues should be deparaffinized to remove all the paraffin on and around the tissue sample in xylene or a good substitute, followed by alcohol. [7]

Antigen retrieval

Antigen retrieval is sometimes necessary to make the epitopes in the tissue accessible. The epitopes are the binding sites for antibodies used to visualize the target and may be masked due to the fixation. Fixation of the tissue may cause formation of methylene bridges or crosslinking of amino groups, so that the epitopes no longer are available. The most common way to perform antigen retrieval is with heat induced epitop retrieval (HIER). This can be done in different ways, for example by using microwave oven, autoclaves, heating plates or water baths. For frozen sections, antigen retrieval is generally not necessary, but for frozen section that has been fixed in aceton or NBF, can antigen retrieval improve the IHC signal. [5]

Blocking

Non-specific binding of antibodies can cause background staining. Although antibodies bind to specific epitopes, they may also partially or weakly bind to sites on nonspecific proteins that are similar to the binding site on the target protein. By incubating the tissue with normal serum isolated from the species which the secondary antibody was produced, the background staining can be reduced. It is also possible to use commercially available universal blocking buffers. Other common blocking buffers include normal serum, non-fat dry milk, BSA, or gelatin. [4] [5] Endogenous enzyme activity may also cause background staining but can be reduced if the tissue is treated with hydrogen peroxide. [4]

Sample labeling

After preparing the sample, the target can be visualized by using antibodies labeled with fluorescent compounds, metals or enzymes. There are direct and indirect methods for labeling the sample. [5] [8]

Antibody types

The antibodies used for detection can be polyclonal or monoclonal. Polyclonal antibodies are made by using animals like guinea pig, rabbit, mouse, rat, or goat. The animal is injected with the antigen of interest and trigger an immune response. The antibodies can be isolated from the animal’s whole serum. Polyclonal antibody production will result in a mixture of different antibodies and will recognize multiple epitopes. Monoclonal antibodies are made by injecting the animal with the antigen of interest and then isolating an antibody-producing B cell, typically from the spleen. The antibody producing cell is then fused with a cancer cell line. This causes the antibodies to show specificity for a single epitope. [9]

For immunohistochemical detection strategies, antibodies are classified as primary or secondary reagents. Primary antibodies are raised against an antigen of interest and are typically unconjugated (unlabeled). Secondary antibodies are raised against immunoglobulins of the primary antibody species. The secondary antibody is usually conjugated to a linker molecule, such as biotin, that then recruits reporter molecules, or the secondary antibody itself is directly bound to the reporter molecule. [8]

Detection methods

The direct method is a one-step staining method and involves a labeled antibody reacting directly with the antigen in tissue sections. While this technique utilizes only one antibody and therefore is simple and rapid, the sensitivity is lower due to little signal amplification, in contrast to indirect approaches. [8]

The indirect method involves an unlabeled primary antibody that binds to the target antigen in the tissue. Then a secondary antibody, which binds with the primary antibody is added as a second layer. As mentioned, the secondary antibody must be raised against the antibody IgG of the animal species in which the primary antibody has been raised. This method is more sensitive than direct detection strategies because of signal amplification due to the binding of several secondary antibodies to each primary antibody. [8]

The indirect method, aside from its greater sensitivity, also has the advantage that only a relatively small number of standard conjugated (labeled) secondary antibodies needs to be generated. For example, a labeled secondary antibody raised against rabbit IgG, is useful with any primary antibody raised in rabbit. This is particularly useful when a researcher is labeling more than one primary antibody, whether due to polyclonal selection producing an array of primary antibodies for a singular antigen or when there is interest in multiple antigens. With the direct method, it would be necessary to label each primary antibody for every antigen of interest. [8]

Chromogenic immunohistochemistry: The cell is exposed to a primary antibody (red) that binds to a specific antigen (purple square). The primary antibody binds a secondary (green) antibody that is chemically coupled to an enzyme (blue). The enzyme changes the color of the substrate to a more pigmented one (brown star). Chromogenic immunohistochemistry.jpg
Chromogenic immunohistochemistry: The cell is exposed to a primary antibody (red) that binds to a specific antigen (purple square). The primary antibody binds a secondary (green) antibody that is chemically coupled to an enzyme (blue). The enzyme changes the color of the substrate to a more pigmented one (brown star).

Reporter molecules

Reporter molecules vary based on the nature of the detection method, the most common being chromogenic and fluorescence detection. In chromogenic immunohistochemistry an antibody is conjugated to an enzyme, such as alkaline phosphate (AP) and horseradish peroxidase (HRP), that can catalyze a color-producing reaction in the presence of a chromogenic substrate like diaminobenzidine (DAB). [4] The colored product can be analyzed with an ordinary light microscope. [10] In immunofluorescence the antibody is tagged to a fluorophore, such as fluorescein isothiocyanate (FITC), tetramethylrhodamine isothiocyanate (TRITC), aminomethyl Coumarin acetate (AMCA) or Cyanine5 (Cy5). Synthetic fluorochromes from Alexa Fluors is also commonly used. [10] [11] The fluorochromes can be visualized by a fluorescence or confocal microscope. [10]

For chromogenic and fluorescent detection methods, densitometric analysis of the signal can provide semi- and fully quantitative data, respectively, to correlate the level of reporter signal to the level of protein expression or localization. [5]

Counterstains

Immunohistochemistry stain versus hematoxylin counterstain. Immunohistochemistry stain versus counterstain.png
Immunohistochemistry stain versus hematoxylin counterstain.

After immunohistochemical staining of the target antigen, another stain is often applied. The counterstain provide contrast that helps the primary stain stand out and makes it easier to examine the tissue morphology. It also helps with orientation and visualization of the tissue section. Hematoxylin is commonly used. [5] [12]

Troubleshooting

In immunohistochemical techniques, there are several steps prior to the final staining of the tissue that can cause a variety of problems. It can be strong background staining, weak target antigen staining and presence of artifacts. It is important that antibody quality and the IHC techniques are optimized. [13] Endogenous biotin, reporter enzymes or primary/secondary antibody cross-reactivity are common causes of strong background staining. [8] [10] Weak or absent staining may be caused by inaccurate fixation of the tissue or to low antigen levels. These aspects of IHC tissue prep and antibody staining must be systematically addressed to identify and overcome staining issues. [4] [5]

Methods to eliminate background staining include dilution of the primary or secondary antibodies, changing the time or temperature of incubation, and using a different detection system or different primary antibody. Quality control should as a minimum include a tissue known to express the antigen as a positive control and negative controls of tissue known not to express the antigen, as well as the test tissue probed in the same way with omission of the primary antibody (or better, absorption of the primary antibody). [4]

Diagnostic IHC markers

Chromogenic immunohistochemistry of a normal kidney targeting the protein CD10. Kidney cd10 ihc.jpg
Chromogenic immunohistochemistry of a normal kidney targeting the protein CD10.

IHC is an excellent detection technique and has the tremendous advantage of being able to show exactly where a given protein is located within the tissue examined. It is also an effective way to examine the tissues. This has made it a widely used technique in neuroscience, enabling researchers to examine protein expression within specific brain structures. Its major disadvantage is that, unlike immunoblotting techniques where staining is checked against a molecular weight ladder, it is impossible to show in IHC that the staining corresponds with the protein of interest. For this reason, primary antibodies must be well-validated in a Western Blot or similar procedure. The technique is even more widely used in diagnostic surgical pathology for immunophenotyping tumors (e.g. immunostaining for e-cadherin to differentiate between DCIS (ductal carcinoma in situ: stains positive) and LCIS (lobular carcinoma in situ: does not stain positive) [14] ). More recently, immunohistochemical techniques have been useful in differential diagnoses of multiple forms of salivary gland, head, and neck carcinomas. [15]

The diversity of IHC markers used in diagnostic surgical pathology is substantial. Many clinical laboratories in tertiary hospitals will have menus of over 200 antibodies used as diagnostic, prognostic and predictive biomarkers. Examples of some commonly used markers include:

PIN-4 staining of a benign gland (left) and prostate adenocarcinoma (right) using the PIN-4 cocktail. The adenocarcinoma lacks the basal epithelial cells (stained dark brown by p63, CK-5 and CK-14). Also, in PIN-4 stained samples, adenocarcinoma cells generally display red cytoplasms (stained by AMACR). PIN-4 staining of benign prostate gland and adenocarcinoma.jpg
PIN-4 staining of a benign gland (left) and prostate adenocarcinoma (right) using the PIN-4 cocktail. The adenocarcinoma lacks the basal epithelial cells (stained dark brown by p63, CK-5 and CK-14). Also, in PIN-4 stained samples, adenocarcinoma cells generally display red cytoplasms (stained by AMACR).

Directing therapy

A variety of molecular pathways are altered in cancer and some of the alterations can be targeted in cancer therapy. Immunohistochemistry can be used to assess which tumors are likely to respond to therapy, by detecting the presence or elevated levels of the molecular target.

Chemical inhibitors

Tumor biology allows for a number of potential intracellular targets. Many tumors are hormone dependent. The presence of hormone receptors can be used to determine if a tumor is potentially responsive to antihormonal therapy. One of the first therapies was the antiestrogen, tamoxifen, used to treat breast cancer. Such hormone receptors can be detected by immunohistochemistry. [18] Imatinib, an intracellular tyrosine kinase inhibitor, was developed to treat chronic myelogenous leukemia, a disease characterized by the formation of a specific abnormal tyrosine kinase. Imitanib has proven effective in tumors that express other tyrosine kinases, most notably KIT. Most gastrointestinal stromal tumors express KIT, which can be detected by immunohistochemistry. [19]

Monoclonal antibodies

Many proteins shown to be highly upregulated in pathological states by immunohistochemistry are potential targets for therapies utilising monoclonal antibodies. Monoclonal antibodies, due to their size, are utilized against cell surface targets. Among the overexpressed targets are members of the EGFR family, transmembrane proteins with an extracellular receptor domain regulating an intracellular tyrosine kinase. [20] Of these, HER2/neu (also known as Erb-B2) was the first to be developed. The molecule is highly expressed in a variety of cancer cell types, most notably breast cancer. As such, antibodies against HER2/neu have been FDA approved for clinical treatment of cancer under the drug name Herceptin. There are commercially available immunohistochemical tests, Dako HercepTest, Leica Biosystems Oracle [21] and Ventana Pathway. [22]

Similarly, EGFR (HER-1) is overexpressed in a variety of cancers including head and neck and colon. Immunohistochemistry is used to determine patients who may benefit from therapeutic antibodies such as Erbitux (cetuximab). [23] Commercial systems to detect EGFR by immunohistochemistry include the Dako pharmDx.

Mapping protein expression

Immunohistochemistry can also be used for a more general protein profiling, provided the availability of antibodies validated for immunohistochemistry. The Human Protein Atlas displays a map of protein expression in normal human organs and tissues. The combination of immunohistochemistry and tissue microarrays provides protein expression patterns in a large number of different tissue types. Immunohistochemistry is also used for protein profiling in the most common forms of human cancer. [24] [25]

See also

Related Research Articles

<span class="mw-page-title-main">Histology</span> Study of the microscopic anatomy of cells and tissues of plants and animals

Histology, also known as microscopic anatomy or microanatomy, is the branch of biology that studies the microscopic anatomy of biological tissues. Histology is the microscopic counterpart to gross anatomy, which looks at larger structures visible without a microscope. Although one may divide microscopic anatomy into organology, the study of organs, histology, the study of tissues, and cytology, the study of cells, modern usage places all of these topics under the field of histology. In medicine, histopathology is the branch of histology that includes the microscopic identification and study of diseased tissue. In the field of paleontology, the term paleohistology refers to the histology of fossil organisms.

Immunoperoxidase is a type of immunostain used in molecular biology, medical research, and clinical diagnostics. In particular, immunoperoxidase reactions refer to a sub-class of immunohistochemical or immunocytochemical procedures in which the antibodies are visualized via a peroxidase-catalyzed reaction.

<span class="mw-page-title-main">Immunostaining</span> Biochemical technique

In biochemistry, immunostaining is any use of an antibody-based method to detect a specific protein in a sample. The term "immunostaining" was originally used to refer to the immunohistochemical staining of tissue sections, as first described by Albert Coons in 1941. However, immunostaining now encompasses a broad range of techniques used in histology, cell biology, and molecular biology that use antibody-based staining methods.

<span class="mw-page-title-main">Immunofluorescence</span> Technique used for light microscopy

Immunofluorescence(IF) is a light microscopy-based technique that allows detection and localization of a wide variety of target biomolecules within a cell or tissue at a quantitative level. The technique utilizes the binding specificity of antibodies and antigens. The specific region an antibody recognizes on an antigen is called an epitope. Several antibodies can recognize the same epitope but differ in their binding affinity. The antibody with the higher affinity for a specific epitope will surpass antibodies with a lower affinity for the same epitope.

<span class="mw-page-title-main">Hybridoma technology</span> Method for producing lots of identical antibodies

Hybridoma technology is a method for producing large numbers of identical antibodies. This process starts by injecting a mouse with an antigen that provokes an immune response. A type of white blood cell, the B cell, produces antibodies that bind to the injected antigen. These antibody producing B-cells are then harvested from the mouse and, in turn, fused with immortal myeloma cancer cells, to produce a hybrid cell line called a hybridoma, which has both the antibody-producing ability of the B-cell and the longevity and reproductivity of the myeloma. The hybridomas can be grown in culture, each culture starting with one viable hybridoma cell, producing cultures each of which consists of genetically identical hybridomas which produce one antibody per culture (monoclonal) rather than mixtures of different antibodies (polyclonal). The myeloma cell line that is used in this process is selected for its ability to grow in tissue culture and for an absence of antibody synthesis. In contrast to polyclonal antibodies, which are mixtures of many different antibody molecules, the monoclonal antibodies produced by each hybridoma line are all chemically identical.

<span class="mw-page-title-main">Carcinoembryonic antigen</span> Glycoprotein secreted into the luminal surface of the epithelia in the gastrointestinal tract

Carcinoembryonic antigen (CEA) describes a set of highly-related glycoproteins involved in cell adhesion. CEA is normally produced in gastrointestinal tissue during fetal development, but the production stops before birth. Consequently, CEA is usually present at very low levels in the blood of healthy adults. However, the serum levels are raised in some types of cancer, which means that it can be used as a tumor marker in clinical tests. Serum levels can also be elevated in heavy smokers.

<span class="mw-page-title-main">Tissue microarray</span>

Tissue microarrays consist of paraffin blocks in which up to 1000 separate tissue cores are assembled in array fashion to allow multiplex histological analysis.

<span class="mw-page-title-main">Immunocytochemistry</span>

Immunocytochemistry (ICC) is a common laboratory technique that is used to anatomically visualize the localization of a specific protein or antigen in cells by use of a specific primary antibody that binds to it. The primary antibody allows visualization of the protein under a fluorescence microscope when it is bound by a secondary antibody that has a conjugated fluorophore. ICC allows researchers to evaluate whether or not cells in a particular sample express the antigen in question. In cases where an immunopositive signal is found, ICC also allows researchers to determine which sub-cellular compartments are expressing the antigen.

<span class="mw-page-title-main">Fixation (histology)</span> Preservation of biological tissue

In the fields of histology, pathology, and cell biology, fixation is the preservation of biological tissues from decay due to autolysis or putrefaction. It terminates any ongoing biochemical reactions and may also increase the treated tissues' mechanical strength or stability. Tissue fixation is a critical step in the preparation of histological sections, its broad objective being to preserve cells and tissue components and to do this in such a way as to allow for the preparation of thin, stained sections. This allows the investigation of the tissues' structure, which is determined by the shapes and sizes of such macromolecules as proteins and nucleic acids.

<span class="mw-page-title-main">Ki-67 (protein)</span> Mammalian protein found in humans

Antigen Kiel 67, also known as Ki-67 or MKI67, is a protein that in humans is encoded by the MKI67 gene.

<span class="mw-page-title-main">CD68</span> Mammalian protein found in Homo sapiens

CD68 is a protein highly expressed by cells in the monocyte lineage, by circulating macrophages, and by tissue macrophages.

<span class="mw-page-title-main">Immunolabeling</span> Procedure for detection and localization of an antigen

Immunolabeling is a biochemical process that enables the detection and localization of an antigen to a particular site within a cell, tissue, or organ. Antigens are organic molecules, usually proteins, capable of binding to an antibody. These antigens can be visualized using a combination of antigen-specific antibody as well as a means of detection, called a tag, that is covalently linked to the antibody. If the immunolabeling process is meant to reveal information about a cell or its substructures, the process is called immunocytochemistry. Immunolabeling of larger structures is called immunohistochemistry.

The estrogen receptor test (ERT) is a laboratory test to determine whether cancer cells have estrogen receptors. This information can guide treatment of the cancer.

<span class="mw-page-title-main">Epithelial cell adhesion molecule</span> Transmembrane glycoprotein

Epithelial cell adhesion molecule (EpCAM), also known as CD326 among other names, is a transmembrane glycoprotein mediating Ca2+-independent homotypic cell–cell adhesion in epithelia. EpCAM is also involved in cell signaling, migration, proliferation, and differentiation. Additionally, EpCAM has oncogenic potential via its capacity to upregulate c-myc, e-fabp, and cyclins A & E. Since EpCAM is expressed exclusively in epithelia and epithelial-derived neoplasms, EpCAM can be used as diagnostic marker for various cancers. It appears to play a role in tumorigenesis and metastasis of carcinomas, so it can also act as a potential prognostic marker and as a potential target for immunotherapeutic strategies.

<span class="mw-page-title-main">B3GAT1</span> Protein-coding gene in the species Homo sapiens

3-beta-glucuronosyltransferase 1 (B3GAT1) is an enzyme that in humans is encoded by the B3GAT1 gene, whose enzymatic activity creates the CD57 epitope on other cell surface proteins. In immunology, the CD57 antigen is also known as HNK1 or LEU7. It is expressed as a carbohydrate epitope that contains a sulfoglucuronyl residue in several adhesion molecules of the nervous system.

<span class="mw-page-title-main">Frozen tissue array</span>

Frozen tissue array consists of fresh frozen tissues in which up to 50 separate tissue cores are assembled in array fashion to allow simultaneous histological analysis.

Porocarcinoma (PCA) is a rare form of skin cancer that develops in eccrine sweat glands, i.e. the body's widely distributed major type of sweat glands, as opposed to the apocrine sweat glands which are located primarily in the armpits and perineal area. This cancer typically develops in individuals as a single cutaneous tumor in the intraepidermal spiral part of these sweat glands' ducts at or near to where they open on the skin's surface. PCA tumors are classified as one form of the cutaneous adnexal tumors; in a study of 2,205 cases, PCA was the most common (11.8%) form of these tumors.

Chromogenic in situ hybridization (CISH) is a cytogenetic technique that combines the chromogenic signal detection method of immunohistochemistry (IHC) techniques with in situ hybridization. It was developed around the year 2000 as an alternative to fluorescence in situ hybridization (FISH) for detection of HER-2/neu oncogene amplification. CISH is similar to FISH in that they are both in situ hybridization techniques used to detect the presence or absence of specific regions of DNA. However, CISH is much more practical in diagnostic laboratories because it uses bright-field microscopes rather than the more expensive and complicated fluorescence microscopes used in FISH.

Tissues that have been preserved with formaldehyde, a highly reactive compound, contain a variety of chemical modifications that can reduce the detectability of proteins in biomedical procedures such as immunohistochemistry. Antigen retrieval is an approach to reducing or eliminating these chemical modifications. The two primary methods of antigen retrieval are heat-mediated epitope retrieval (HIER) and proteolytic induced epitope retrieval (PIER).

Mammary analogue secretory carcinoma (MASC), also termed MASCSG, is a salivary gland neoplasm. It is a secretory carcinoma which shares the microscopic pathologic features with other types of secretory carcinomas including mammary secretory carcinoma, secretory carcinoma of the skin, and salivary gland–type carcinoma of the thyroid. MASCSG was first described by Skálová et al. in 2010. The authors of this report found a chromosome translocation in certain salivary gland tumors, i.e. a (12;15)(p13;q25) fusion gene mutation. The other secretory carcinoma types carry this fusion gene.

References

  1. Ortiz Hidalgo C (2022), Del Valle L (ed.), "Immunohistochemistry in Historical Perspective: Knowing the Past to Understand the Present", Immunohistochemistry and Immunocytochemistry, Methods in Molecular Biology, vol. 2422, New York, NY: Springer US, pp. 17–31, doi:10.1007/978-1-0716-1948-3_2, ISBN   978-1-0716-1947-6, PMID   34859396, S2CID   244861186 , retrieved 2024-02-22
  2. "Immunohistochemistry: Origins, Tips, and a Look to the Future". The Scientist Magazine®. Retrieved 2024-02-22.
  3. Duraiyan J, Govindarajan R, Kaliyappan K, Palanisamy M (2012). "Applications of immunohistochemistry". Journal of Pharmacy and Bioallied Sciences. 4 (6): S307-9. doi: 10.4103/0975-7406.100281 . ISSN   0975-7406. PMC   3467869 . PMID   23066277.
  4. 1 2 3 4 5 6 7 Magaki S, Hojat SA, Wei B, So A, Yong WH (2019), Yong WH (ed.), "An Introduction to the Performance of Immunohistochemistry", Biobanking, vol. 1897, New York, NY: Springer New York, pp. 289–298, doi:10.1007/978-1-4939-8935-5_25, ISBN   978-1-4939-8933-1, PMC   6749998 , PMID   30539453
  5. 1 2 3 4 5 6 7 8 9 10 Kim SW, Roh J, Park CS (2016-11-15). "Immunohistochemistry for Pathologists: Protocols, Pitfalls, and Tips". Journal of Pathology and Translational Medicine. 50 (6): 411–418. doi:10.4132/jptm.2016.08.08. ISSN   2383-7837. PMC   5122731 . PMID   27809448.
  6. Libard S, Cerjan D, Alafuzoff I (January 2019). "Characteristics of the tissue section that influence the staining outcome in immunohistochemistry". Histochemistry and Cell Biology. 151 (1): 91–96. doi:10.1007/s00418-018-1742-1. ISSN   0948-6143. PMC   6328518 . PMID   30357509.
  7. Binch A, Snuggs J, Le Maitre CL (2020-05-15). "Immunohistochemical analysis of protein expression in formalin fixed paraffin embedded human intervertebral disc tissues". Jor Spine. 3 (3): e1098. doi:10.1002/jsp2.1098. ISSN   2572-1143. PMC   7524243 . PMID   33015573.
  8. 1 2 3 4 5 6 Ramos-Vara JA (2005-07-04). "Technical Aspects of Immunohistochemistry". Veterinary Pathology. 42 (4): 405–426. doi:10.1354/vp.42-4-405. ISSN   0300-9858. PMID   16006601. S2CID   6229029.
  9. Peltomaa R, Barderas R, Benito-Peña E, Moreno-Bondi MC (2021-08-21). "Recombinant antibodies and their use for food immunoanalysis". Analytical and Bioanalytical Chemistry. 414 (1): 193–217. doi:10.1007/s00216-021-03619-7. ISSN   1618-2642. PMC   8380008 . PMID   34417836.
  10. 1 2 3 4 Krenacs T, Krenacs L, Raffeld M (2010), Oliver C, Jamur MC (eds.), "Multiple Antigen Immunostaining Procedures", Immunocytochemical Methods and Protocols, vol. 588, Totowa, NJ: Humana Press, pp. 281–300, doi:10.1007/978-1-59745-324-0_28, ISBN   978-1-58829-463-0, PMC   7670878 , PMID   20012839
  11. Im K, Mareninov S, Diaz MF, Yong WH (2019), Yong WH (ed.), "An Introduction to Performing Immunofluorescence Staining", Biobanking, vol. 1897, New York, NY: Springer New York, pp. 299–311, doi:10.1007/978-1-4939-8935-5_26, ISBN   978-1-4939-8933-1, PMC   6918834 , PMID   30539454
  12. Zehntner SP, Chakravarty MM, Bolovan RJ, Chan C, Bedell BJ (2008-06-03). "Synergistic Tissue Counterstaining and Image Segmentation Techniques for Accurate, Quantitative Immunohistochemistry". Journal of Histochemistry & Cytochemistry. 56 (10): 873–880. doi:10.1369/jhc.2008.950345. ISSN   0022-1554. PMC   2544616 . PMID   18574255.
  13. Ward JM, Rehg JE (2013-09-27). "Rodent Immunohistochemistry: Pitfalls and Troubleshooting". Veterinary Pathology. 51 (1): 88–101. doi:10.1177/0300985813503571. ISSN   0300-9858. PMID   24078006. S2CID   20084645.
  14. O'Malley F, Pinder S (2006). Breast Pathology (First ed.). Edinburgh: Churchill Livingstone/Elsevier. ISBN   978-0-443-06680-1.
  15. Zhu S, Schuerch C, Hunt J (January 2015). "Review and updates of immunohistochemistry in selected salivary gland and head and neck tumors". Archives of Pathology & Laboratory Medicine. 139 (1): 55–66. doi: 10.5858/arpa.2014-0167-RA . PMID   25549144.
  16. Taupin P (January 2007). "BrdU immunohistochemistry for studying adult neurogenesis: paradigms, pitfalls, limitations, and validation". Brain Research Reviews. 53 (1): 198–214. doi:10.1016/j.brainresrev.2006.08.002. PMID   17020783. S2CID   23557588.
  17. Leader M, Patel J, Makin C, Henry K (December 1986). "An analysis of the sensitivity and specificity of the cytokeratin marker CAM 5.2 for epithelial tumours. Results of a study of 203 sarcomas, 50 carcinomas and 28 malignant melanomas". Histopathology. 10 (12): 1315–1324. doi:10.1111/j.1365-2559.1986.tb02574.x. PMID   2434403. S2CID   28142859.
  18. Jørgensen JT, Nielsen KV, Ejlertsen B (April 2007). "Pharmacodiagnostics and targeted therapies - a rational approach for individualizing medical anticancer therapy in breast cancer". The Oncologist. 12 (4): 397–405. doi: 10.1634/theoncologist.12-4-397 . PMID   17470682. S2CID   19065186.
  19. Gold JS, Dematteo RP (August 2006). "Combined surgical and molecular therapy: the gastrointestinal stromal tumor model". Annals of Surgery. 244 (2): 176–184. doi:10.1097/01.sla.0000218080.94145.cf. PMC   1602162 . PMID   16858179.
  20. Harari PM (December 2004). "Epidermal growth factor receptor inhibition strategies in oncology". Endocrine-Related Cancer. 11 (4): 689–708. doi: 10.1677/erc.1.00600 . PMID   15613446.
  21. "leicabiosystems.com". leicabiosystems.com. Retrieved 2013-06-16.
  22. Press MF, Sauter G, Bernstein L, Villalobos IE, Mirlacher M, Zhou JY, et al. (September 2005). "Diagnostic evaluation of HER-2 as a molecular target: an assessment of accuracy and reproducibility of laboratory testing in large, prospective, randomized clinical trials". Clinical Cancer Research. 11 (18): 6598–6607. doi: 10.1158/1078-0432.CCR-05-0636 . PMID   16166438.
  23. Bibeau F, Boissière-Michot F, Sabourin JC, Gourgou-Bourgade S, Radal M, Penault-Llorca F, et al. (September 2006). "Assessment of epidermal growth factor receptor (EGFR) expression in primary colorectal carcinomas and their related metastases on tissue sections and tissue microarray". Virchows Archiv. 449 (3): 281–287. doi:10.1007/s00428-006-0247-9. PMC   1888717 . PMID   16865406.
  24. "The Human Protein Atlas". www.proteinatlas.org. Retrieved 2017-10-02.
  25. Uhlén M, Fagerberg L, Hallström BM, Lindskog C, Oksvold P, Mardinoglu A, et al. (January 2015). "Proteomics. Tissue-based map of the human proteome". Science. 347 (6220): 1260419. doi:10.1126/science.1260419. PMID   25613900. S2CID   802377.

Further reading