Immunology: Current Research
Open Access

Our Group organises 3000+ Global Conferenceseries Events every year across USA, Europe & Asia with support from 1000 more scientific Societies and Publishes 700+ Open Access Journals which contains over 50000 eminent personalities, reputed scientists as editorial board members.

Open Access Journals gaining more Readers and Citations
700 Journals and 15,000,000 Readers Each Journal is getting 25,000+ Readers

This Readership is 10 times more when compared to other Subscription Journals (Source: Google Analytics)
  • Editorial   
  • Immunol Curr Res, Vol 1(1)

Immunohistochemistry as an Important Tool for Cancer Immunotherapy: From Diagnosis to Drug Development

Rances Blanco*
Laboratory of Recognition and Biological Activity Assays, Center of Molecular Immunology, Havana, Cuba
*Corresponding Author: Rances Blanco, Laboratory of Recognition and Biological Activity Assays, Center of Molecular Immunology, Havana, Cuba, Tel: 537-2717933-3464, Email: rances@cim.sld.cu

Received: 03-Oct-2017 / Accepted Date: 03-Oct-2017 / Published Date: 10-Oct-2017

Keywords: Antibody, antigen

Editorial

Immunohistochemistry (IHC) is a well-established method for identifying both cellular and tissue antigens by means of specific antigen-antibody reactions [1]. This method is a unique analytical tool combining molecular detection with morphological features and can be used on formalin-fixed and paraffin-embedded sections, frozen sections, conventional cytological smears, and other cytological preparations (e.g. isolated circulating tumor cells) [2]. The use of IHC in cancer research has expanded to involve diagnostic classification, anti-tumor drug development, disease prognosis, therapy selection, prediction to response and follow-up, contributing significantly to personalized medicine.

Between the most important applications of IHC in cancer diagnostic are: determination of the cell lineage of undifferentiated malignant tumors, characterization of metastatic malignancies of unknown primary tumors [3,4] and molecular classification of neoplasias [5]. The molecular classification of cancer also permits to identify which patients are most likely to benefit from targeted drugs, leading to a more appropriate therapeutic strategy [6,7]. At present, some IHC-based diagnostic kits are commercially available for the detection of anti-cancer therapeutic targets (e.g. HercepTest™, EGFR pharmDx™, PD-L1 IHC 22C3 pharmDx) and guidelines have been written by expert panels to standardize technique steps, data interpretation and reporting of results [8,9].

In many types of cancer, some IHC analysis such as proliferating antigens, angiogenesis-related molecules, growth factor and their receptors and tumor-infiltrating lymphocytes (immunoscore) are commonly used to obtain information about prognosis as well as a complement to histological grading of tumors [10,11]. Moreover, newer IHC biomarkers, scoring systems and cutoffs are being continuously explored for prediction of outcome [12] or response to moleculartargeted therapies [13,14]. Recently, the immunocytochemical detection of tumor antigens (e.g. PD-1/PD-L1 checkpoint pathway) expressed in circulating tumor cells (CTCs) highlighted the application of IHC into clinical practice permitting the follow-up of patients in a minimally invasive manner [15].

IHC also helps identifying the most relevant animal species with a similar target expression profile to human in order to perform both invivo toxicity and in-vivo pharmacodynamic modeling studies [16]. In combination, these assessments provide useful quantitative information about the effects of cross-reacting biologics predicting potential toxicity in humans. Other application of IHC using animal models is the study of the mechanisms of action for immunotherapeutics [17]. The use of cell lines of known antigens profile expression to induce experimental primary tumors and/or metastasis permit to evaluate, by means of IHC, the anti-tumoral properties of novel biological products as well as the molecular pathways involved in this effect [18].

Some techniques could be used to evaluate the target antigenbinding attributes of biologic drugs (e.g. tissue-based lysate western blots). However, screening studies using IHC provide a unique evaluation combining various immunostaining features such as pattern, distribution, intensity and frequency across panels of frozen tissues from normal humans (tissue cross-reactivity studies). This evaluation permit to identify both previously unknown sites of the target antigen and cross-reactive epitopes (unexpected targets) alerting researches to potential toxicity toward certain organs that could be observed in first-in-human clinical trials [19]. Consequently, tissue cross-reactivity studies constitute an essential part of the preclinical safety assessment package [20,21].

Tissue cross-reactivity studies also permit to compare first generation monoclonal antibodies (usually murine) with the second generation or recombinant versions of these Mabs (e.g. chimeric, humanized) [22]. These studies contribute to detect alterations in the affinity and/or in the specificity of therapeutic drugs when changes in molecule structure, production process or facilities are introduced. In the context of biosimilar antibodies, the IHC comparative studies also allow confirming the pattern of recognition of innovators vs. biosimilar candidates [23]. Frequently, IHC using fetal and malignant human tissues permit to obtain a complete characterization about the expression profile of target molecules and binding properties of therapeutic agents [22].

Despite of significant advances in a variety of newer technologies (e.g. next generation sequencing, super-resolution microscopy, proteomic mass spectrometry), up to day IHC remains one of the best choices for both cancer research and routine clinical use [12,13,18]. However, recurrently the confiability and reproducibility of IHC studies are strongly questioned. The lack of standardization of some preanalytical (e.g. handling of tissue sample, fixation, processing steps), analytical (e.g. antigen retrieval, primary antibody, detection system) and post-analytical (e.g. analysis, data interpretation, final report) factors is not completely resolved. Inconsistencies in IHC studies are considered responsible of both intra and inter-laboratory reported variabilities.

Some recommendations for a better performance of IHC and to recover it reliability include, but are not limited to, adequate use of negative and positive controls in each staining run, application of internal quality control systems for routine immunohistochemical procedures in each laboratory, validation of all immunostaining tests before the introduction into clinical practice, implementation of equivalence studies when deviations during validated technique performance occur, harmonization of scores and cutoff points to reduce the inter-laboratory variability in reports and introduction of automated tissue image analysis for an improved signal quantification of the immunohistochemical results.

References

  1. Kabiraj A, Gupta J, Khaitan T, Bhattacharya PT (2015) Principle and techniques of immunohistochemistry –a review. Int J Biol Med Res 6: 5204-5210.
  2. Weller P, Nel I, Hassenkamp P, Gauler T, Schlueter A, et al. (2014) Detection of circulating tumor cell subpopulations in patients with head and neck squamous cell carcinoma (HNSCC). PLoS One 9: e113706.
  3. Pavlidis N, Khaled H, Gaafar R (2015) A mini review on cancer of unknown primary site: A clinical puzzle for the oncologists. J Adv Res 6: 375-382.
  4. Kandalaft PL, Gown AM (2016) Practical applications in immunohistochemistry: Carcinomas of unknown primary site. Arch Pathol Lab Med 140: 508-523.
  5. Rakha EA, Green AR (2017) Molecular classification of breast cancer: What the pathologist needs to know. Pathology 49: 111-119.
  6. Ock CY, Lee KW, Kim JW, Kim JS, Kim TY, et al. (2015) Optimal patient selection for trastuzumab treatment in HER2-positive advanced gastric cancer.Clin Cancer Res 21: 2520-2529.
  7. Ma X, Wang L, Li H, Zhang Y, Gao Y, et al. (2016) Predictive immunohistochemical markers related to drug selection for patients treated with sunitinib or sorafenib for metastatic renal. Cell Cancer. Sci Rep 6: 30886.
  8. Varga Z, Noske A (2015) Impact of modified 2013 ASCO/CAP guidelines on HER2 testing in breast cancer. One Year Experience. PLoS One10: e0140652.
  9. Cree IA, Booton R, Cane P, Gosney J, Ibrahim M, et al. (2016) PD-L1 testing for lung cancer in the UK: recognizing the challenges for implementation. Histopathology 69: 177-186.
  10. Zavadova E, Spacek J, Vocka M, Konopasek B, Fucikova T, et al. (2015) Immunoscore and its predictive value for colorectal cancer. Klin Onkol 28: 82-85.
  11. Mlecnik B, Bindea G, Angell HK, Maby P, Angelova M, et al. (2016) Integrative analyses of colorectal cancer show immunoscore is a stronger predictor of patient survival than microsatellite instability. Immunity 44: 698-711.
  12. Blanco R, Domínguez E, Morales O, Blanco D, Martínez D, et al. (2015) Prognostic significance of N-Glycolyl GM3 ganglioside expression in non-small cell lung carcinoma patients: New evidences. Patholog Res Int 2015: 1-12.
  13. Pirker R, Pereira JR, von Pawel J, Krzakowski M, Ramlau R, et al. (2012) EGFR expression as a predictor of survival for first-line chemotherapy plus cetuximab in patients with advanced non-small-cell lung cancer: Analysis of data from the phase 3 FLEX study. Lancet Oncol 13: 33-42.
  14. Garon EB, Rizvi NA, Hui R, Leighl N, Balmanoukian AS, et al. (2015) Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med 372: 2018-2028.
  15. Nicolazzo C, Raimondi C, Mancini M, Caponnetto S, Gradilone A, et al. (2016) Monitoring PD-L1 positive circulating tumor cells in non-small cell lung cancer patients treated with the PD-1 inhibitor Nivolumab. Sci Rep 6: 31726.
  16. Maceira M, Rengifo E, Cedeno M, Merino NVMD, Parada AC (2004) Immunohistochemicalrecognition of the epidermal growth factor receptor by the H-R3 antibody in the skin of experimental animals. Appl Immunohistochem Mol Morphol 12: 360-363.
  17. Diaz A, Blanco B, Pineda I, Lemm M, Aciego T, et al. (2012) Preclinical efficacy of nimotuzumab, an anti-EGFR monoclonal antibody as a single agent therapy in human GBM U87MG xenografts. Cancer Ther 3: 245-255.
  18. Palomo AG, Santana RB, Pérez XE, Santana DB, Gabri MR, et al. (2016) Frequent co-expression of EGFR and NeuGcGM3 ganglioside in cancer: It’s potential therapeutic implications. ClinExp Metastasis 33:717-725.
  19. Leach MW, Halpern WG, Johnson CW, Rojko JL, MacLachlan TK, et al. (2010) Use of tissue cross-reactivity studies in the development of antibody-based biopharmaceuticals: history, experience, methodology, and future directions. Toxicol Pathol 38: 1138-1166.
  20. Food and Drug Administration (FDA) (1997) Points to consider in the manufacture and testing of monoclonal antibody products for human use. U. S. Department of Health and Human Services.
  21. Committee For Medicinal Products For Human Use (2008) Guideline on development, production, characterization and specifications for monoclonal antibodies and related products. European Medicines Agency.
  22. Cedeno-AriasM, Rengifo CE, Batista YR, Calzado ER, Rodríguez T (2007) Immunohistochemicalevaluation of H-R3 a novel humanized monoclonal antibody that neutralizes the EGF-receptor. Appl Immunohistochem Mol Morphol 15: 213-222.
  23. Reichert J, Beck A, Lyer H (2009) European medicines agency workshop on biosimilar monoclonal antibodies. mAbs 1: 3 94-416.

Citation: Blanco R (2017) Immunohistochemistry as an Important Tool for Cancer Immunotherapy: From Diagnosis to Drug Development. Immunol Curr Res 1: e103.

Copyright: © 2017 Blanco R. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.

Top