Subscribe to Newsletter
Diagnostics Precision medicine, Oncology, Histology

Time, Money, and Tissue

At a Glance

  • Immunohistochemistry is a routine part of the anatomic pathology workflow
  • By multiplexing targets, users can maximize their return from a single piece of tissue – often a precious resource
  • Testing multiple targets in a single round also saves the laboratory time and money
  • To fully benefit from the opportunities multiplex IHC offers, the next step is automation

In the decades since Coons and colleagues published their revolutionary work on immunofluorescence detection of antigens in frozen tissue (1), immunohistochemistry (IHC) has become routine in the anatomic pathology laboratory. Each target antigen of interest has been individually identified within histological sections of formalin-fixed, paraffin-embedded (FFPE) tumors or other types of tissue (2). Single-marker IHC takes advantage of the labeling capabilities of horseradish peroxidase and alkaline phosphatase enzymes, in combination with their respective reactant chromogens, to produce a colorimetric stain for visualization under a light microscope (2,3). Alternatively, fluorescent reporters – fluorochromes – can visualize the antibody-antigen interaction, either by conjugation to the primary antibody (direct immunofluorescence) or via attachment to a secondary antibody that detects the species-specific primary (indirect immunofluorescence).

More recently, IHC users have shifted from the single-marker approach to multiplexed marker detection. Multiplex IHC methods can visualize multiple target antigens within a single tissue sample and can be further subcategorized as sequential or simultaneous (4). Generally, if the primary antibodies used are from the same host species, sequential multiplex IHC is required; otherwise, they can be cocktailed and incubated simultaneously.

But why use multiplex IHC at all? There are numerous advantages to visualizing multiple antigens simultaneously. Multiplex IHC maximizes the amount of data acquired from a single tissue sample – critical in conserving precious patient tissue. Unlike next-generation sequencing and mass spectrometry, in which the tissue sample is destroyed to test for individual target molecules, multiplex IHC also allows users to examine the spatial arrangements, interactions, and co-localizations of proteins of interest within the tissue architecture (5).

The complexity of the multiplex IHC protocol necessitates properly trained, highly skilled staff to achieve the most accurate and reproducible diagnoses. In clinical application, these technologically complex techniques require automation to achieve a simple, efficient, and easily understandable result for the clinical pathologist not well-versed in advanced multiplexing methods. Additionally, as the enhanced diagnostic utility of multiplex IHC is realized, the histology laboratory will experience increasing demand. Automation (and the associated standardization and reduction of variability) allows labs to achieve the quality, reproducibility, and speed necessary to meet this demand (6).

Multiplex IHC for diagnostics

One driving factor in the early adoption of multiplex IHC for clinical diagnostics was urologists’ collective move to use the smallest possible needle gauge to perform prostate biopsies. This caused pathologists great difficulty in reliably diagnosing (or ruling out) small foci of cancer; examining multiple minute tissue fragments was time-consuming and reproducibility of diagnosis was poor. The most glaring clinical need was the ability to differentiate between prostatic intraepithelial neoplasia (PIN) and carcinoma of the prostate. We also needed a way of clearly identifying, with high accuracy and specificity, any microinvasion or micrometastasis into adjacent prostate tissue. And, of course, this had to be done while conserving the limited amount of sample (prostate needle biopsies are thin filaments of tissue) and reducing the time to result.

It consists of one or two antibodies against high molecular weight cytokeratin (CK HMW), as well as antibodies to p63 and p504S (also known as AMACR enzyme).

With these constraints and needs in mind, a widely used antibody cocktail known as PIN-4 was developed (see Figure 1). It consists of one or two antibodies against high molecular weight cytokeratin (CK HMW), as well as antibodies to p63 and p504S (also known as AMACR enzyme). Studies have shown that combinations of CK HMW [34βE12], p63, and/or AMACR may be useful in differentiating normal prostate glands from PIN and prostatic adenocarcinoma (PCa) (7,8,9). In prostate tissue, CK HMW [34βE12] is a useful marker of basal cells of normal glands and PIN, a precursor lesion to prostatic adenocarcinoma; invasive prostatic adenocarcinoma, in contrast, typically lacks a basal cell layer (9,10,11).

p63, a homolog of the tumor suppressor p53, has been detected in nuclei of the basal epithelium in normal prostate glands; however, it is not expressed in malignant tumors of the prostate (12). α-methylacyl coenzyme A racemase (AMACR), also known as P504S, has been shown to be a specific marker of prostatic adenocarcinoma (13,14,15,16). Additionally, prostate glands involved in PIN have been found to express AMACR, whereas the enzyme was nearly undetectable in benign glands (16,17).

Figure 1: Prostate cancer stained with CK HMW + p63 + AMACR (RM). Multiplex IHC detection of CK HMW plus antibodies to p63 and AMACR in prostate adenocarcinoma biopsy. Strong AMACR (red) expression observed across the middle of the photomicrograph without corresponding basal cell layer (lack of CK HMW and p63 expression [DAB; brown]) indicates invasive PCa. Glands still containing basal cells (upper and lower portions of image) show signs of loss of a continuous basal cell layer surrounding the glands, indicative of prostatic intraepithelial neoplasia (PIN).

Pathologists encountered great difficulty in colocalizing the three individual antibody signals (CK HMW + p63 + AMACR) on three separate slides. In many cases, it proved impossible because the biopsies themselves were small, with the suspected PIN or PCa areas smaller or even absent as the tissue block was cut deeper for serial sectioning. However, applying these antibodies to an individual tissue section in a multiplex IHC cocktail format allows for the simultaneous pathological evaluation of each of these critical markers in the same focus or foci of interest, drastically improving reproducibility. It also improves the diagnostic accuracy of invasive prostatic adenocarcinoma to near certainty (18) – which is why the PIN-4 cocktail has become the standard of care in testing prostate needle biopsies.

The clinical application of multiplex IHC has since been expanded to other tissues to give enhanced differential diagnostic information. In breast, the ADH-5 (CK5/14 + p63 + CK7/18) multiplex cocktail (see Figure 2) can aid in the differential diagnoses of usual ductal hyperplasia (UDH), atypical ductal hyperplasia (ADH), and ductal carcinoma in situ (DCIS) (19). UDH carries minimal or no increased risk of breast cancer, and these patients do not undergo any additional procedures; however, ADH and DCIS progress to invasive carcinoma in 4–5 percent and 8–10 percent of cases, respectively. ADH and DCIS patients are advised to undergo excision surgery, with radiation treatment added for DCIS patients (20).

Studies have demonstrated that up to 40 percent of lesions diagnosed as ADH on core biopsies ended up reclassified as benign upon re-examination after surgical excision.

Histological differentiation between UDH, ADH, and DCIS has historically been difficult, with poor concordance among pathologists giving rise to potential misdiagnosis and improper treatment. The addition of the ADH-5 multiplex cocktail to routine histopathology testing significantly reduced overdiagnosis of ADH lesions, reclassifying those lesions as UDH. Studies have demonstrated that up to 40 percent of lesions diagnosed as ADH on core biopsies ended up reclassified as benign upon re-examination after surgical excision (21,22) – meaning that all of those patients could have avoided unnecessary surgery upon initial testing using the ADH-5 multiplex cocktail.

Figure 2: Breast lesions stained with CK5/14 + p63 + CK7/18. Multiplex IHC detection of cytokeratins (CK5, CK14, CK7, CK18) plus p63 in breast lesion biopsies. Breast basal cells express cytokeratins 5 and 14 (DAB; brown), myoepithelial cells express those same cytokeratins along with p63 (DAB; brown), and luminal cells express cytokeratins 7 and 18 (red). Left: In UDH, a polymorphic neoplastic proliferation results in myoepithelial/basal cells intermingled with luminal cells to reveal a heterogeneous, mosaic staining pattern. Right: ADH is a monomorphic neoplasia, typically derived from luminal cells, showing a homogeneous staining pattern across the affected ductal structure with little to no staining of myoepithelial/basal cells.

Multiplex IHC for therapeutic decisions

As the cancer treatment landscape has evolved, so have the diagnostic tools we use to make those critical therapeutic decisions. Now, immunotherapy is on the rise. A number of immune checkpoint inhibitors targeting either cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed cell death protein 1 (PD-1), or its ligand (PD-L1), have recently received FDA approval for the treatment of multiple cancer types. However, up to 60 percent of patients treated with these inhibitors see little to no benefit (23). A critical aspect in proper application of these newly designed immunotherapeutics is to establish valid predictive biomarkers to enhance patient selection.

One proposed approach to determining patient response to immune checkpoint inhibitors is to analyze the tumor microenvironment. An immune-active tumor microenvironment is critical to patient response to immunotherapy (24). We must not only understand the dynamic nature of the tumor, but also determine its interactions with its microenvironment to define the algorithm of biomarkers that will predict response to checkpoint inhibitors. Multiplex IHC is well-suited to resolve and define these elements and interactions (25,26).Profiling the tumor microenvironment within tissues requires evaluating multiple markers, including inflammatory cell subpopulations, tumor-infiltrating lymphocytes, and immune checkpoints (see Figure 3).

Figure 3: Melanoma stained with SOX10, PD-L1, and CD8. Multiplex IHC detection of SOX10, PD-L1, and CD8 helps define high proliferation zones in melanoma. SOX10 nuclear staining (blue) is observed in melanoma cells, with a subset of the melanoma (left) showing PD-L1(+) membranous co-expression (DAB; brown). High CD8 cytotoxic T cell staining (red) is associated with strong PD-L1 expression in the melanoma tumor cells. The SOX10/PD-L1/CD8 triple stain can help discriminate tumor cells from non-tumor cells and may facilitate quantifying or immunoscoring for accurate assessment.

The next step forward

The automation of multiplex IHC is the next evolutionary step to maximizing its potential. The considerable throughput and performance demands placed on diagnostic laboratories for accurate, consistent, high-quality staining results will only increase as novel assays are developed. In turn, laboratories will demand more from their automated IHC staining platforms. New innovations that increase efficiency – such as simultaneous multiplex IHC technology capability, online deparaffinization, and energy-efficient, parallel-processing antigen retrieval – will allow laboratories to meet the throughput and performance demands and beyond, all while continuing to provide high-quality results. As we increasingly move toward multiplexing and automation in the anatomic pathology laboratory, we’ll save time, money, and precious patient tissue.

Disclosure: Jason Ramos is Vice President of Research and Development at Biocare Medical, Pacheco, USA.

Receive content, products, events as well as relevant industry updates from The Pathologist and its sponsors.
Stay up to date with our other newsletters and sponsors information, tailored specifically to the fields you are interested in

When you click “Subscribe” we will email you a link, which you must click to verify the email address above and activate your subscription. If you do not receive this email, please contact us at [email protected].
If you wish to unsubscribe, you can update your preferences at any point.

  1. AH Coons et al., “Immunological properties of an antibody containing a fluorescent group”, Exp Biol Med, 47, 200–202 (1941).
  2. JA Ramos-Vara, “Technical aspects of immunohistochemistry”, Vet Pathol, 42, 405 (2005). PMID: 16006601.
  3. CR Taylor et al., “Techniques of Immunohistochemistry: Principles, Pitfalls, and Standardization”, Diagnostic Immunohistochemistry, 3rd edition, 1–41. W.B. Saunders: 2011.
  4. CM van der Loos, Immunoenzyme Multiple Staining Methods. Bios Scientific Publishers: 1999.
  5. AR Dixon et al., “Recent developments in multiplexing techniques for immunohistochemistry”, Expert Rev Mol Diagn, 15, 1171 (2015). PMID: 26289603.
  6. JW Prichard, “Overview of automated immunohistochemistry”, Arch Pathol Lab Med, 138, 1578 (2014). PMID: 25427039.
  7. RB Shah et al., “Usefulness of basal cell cocktail (34betaE12 + p63) in the diagnosis of atypical prostate glandular proliferations”, Am J Clin Pathol, 122, 517 (2004). PMID: 15487448.
  8. MT Sung et al., “Alpha-methylacyl-CoA racemase (P504S)/34betaE12/p63 triple cocktail stain in prostatic adenocarcinoma after hormonal therapy”, Hum Pathol, 38, 322 (2007). PMID: 17134736.
  9. DG Bostwick, J Qian, “High-grade prostatic intraepithelial neoplasia”, Mod Pathol, 17, 360 (2004). PMID: 14739906.
  10. PA Humphrey, “Diagnosis of adenocarcinoma in prostate needle biopsy tissue”, J Clin Pathol, 60, 35 (2007). PMID: 17213347.
  11. RB Shah et al., “Comparison of the basal cell-specific markers, 34betaE12 and p63, in the diagnosis of prostate cancer”, Am J Surg Pathol, 26, 1161 (2002). PMID: 12218572.
  12. S Signoretti et al., “p63 is a prostate basal cell marker and is required for prostate development”, Am J Pathol, 157, 1769 (2000). PMID: 11106548.
  13. J Xu et al., “Identification of differentially expressed genes in human prostate cancer using subtraction and microarray”, Cancer Res, 60, 1677 (2000). PMID: 10749139.
  14. MA Rubin et al., “alpha-Methylacyl coenzyme A racemase as a tissue biomarker for prostate cancer”, JAMA, 287, 1662 (2002). PMID: 11926890.
  15. J Luo et al., “Alpha-methylacyl-CoA racemase: a new molecular marker for prostate cancer”, Cancer Res, 62, 2220 (2002). PMID: 11956072.
  16. M Zhou et al., “Alpha-Methylacyl-CoA racemase: a novel tumor marker over-expressed in several human cancers and their precursor lesions”, Am J Surg Pathol, 26, 926 (2002). PMID: 12131161.
  17. CL Wu et al., “Analysis of alpha-methylacyl-CoA racemase (P504S) expression in high-grade prostatic intraepithelial neoplasia”, Hum Pathol, 35, 1008 (2004). PMID: 15297968.
  18. H Yaziji et al., “Immunohistochemistry cocktails are here to stay: Center for Medicare and Medicaid Services should revise its new reimbursement policy”, Am J Clin Pathol, 138, 10 (2012). PMID: 22706851.
  19. RK Jain et al., “Atypical ductal hyperplasia: interobserver and intraobserver variability”, Mod Pathol, 24, 917 (2011). PMID: 21532546.
  20. WD Dupont, DL Page, “Risk factors for breast cancer in women with proliferative breast disease”, N Engl J Med, 312, 146 (1985). PMID: 3965932.
  21. CR Smart et al., “Twenty-year follow-up of the breast cancers diagnosed during the Breast Cancer Detection Demonstration Project”, CA Cancer J Clin, 47, 134 (1997). PMID: 9152171.
  22. EC Gal-Gombos et al., “Large-needle core biopsy in atypical intraductal epithelial hyperplasia including immunohistochemical expression of high molecular weight cytokeratin: analysis of results of a single institution”, Breast J, 8, 269 (2002). PMID: 12199753.
  23. S Maleki Vareki et al., “Biomarkers of response to PD-1/PD-L1 inhibition”, Crit Rev Oncol Hematol, 116, 116 (2017). PMID: 28693793.
  24. RR Ji et al., “An immune-active tumor microenvironment favors clinical response to ipilimumab”, Cancer Immunol Immunother, 61, 1019 (2012). PMID: 22146893.
  25. J Kalra, J Baker, “Multiplex immunohistochemistry for mapping the tumor microenvironment”, Methods Mol Biol, 1554, 237 (2017). PMID: 28185197.
  26. H Halse et al., “Multiplex immunohistochemistry accurately defines the immune context of metastatic melanoma”, Sci Rep, 8, 11158 (2018). PMID: 30042403.
About the Author
Jason Ramos

Vice President of Research and Development at Biocare Medical.

Register to The Pathologist

Register to access our FREE online portfolio, request the magazine in print and manage your preferences.

You will benefit from:
  • Unlimited access to ALL articles
  • News, interviews & opinions from leading industry experts
  • Receive print (and PDF) copies of The Pathologist magazine

Register