Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
1.
Mod Pathol ; 37(6): 100489, 2024 Apr 06.
Article in English | MEDLINE | ID: mdl-38588883

ABSTRACT

Somatic tumor testing in prostate cancer (PCa) can guide treatment options by identifying clinically actionable variants in DNA damage repair genes, including acquired variants not detected using germline testing alone. Guidelines currently recommend performing somatic tumor testing in metastatic PCa, whereas there is no consensus on the role of testing in regional disease, and the optimal testing strategy is only evolving. This study evaluates the frequency, distribution, and pathologic correlates of somatic DNA damage repair mutations in metastatic and localized PCa following the implementation of pathologist-driven reflex testing at diagnosis. A cohort of 516 PCa samples were sequenced using a custom next-generation sequencing panel including homologous recombination repair and mismatch repair genes. Variants were classified based on the Association for Molecular Pathology/American Society of Clinical Oncology/College of American Pathologists guidelines. In total, 183 (35.5%) patients had at least one variant, which is as follows: 72 of 516 (13.9%) patients had at least 1 tier I or tier II variant, whereas 111 of 516 (21.5%) patients had a tier III variant. Tier I/II variant(s) were identified in 27% (12/44) of metastatic biopsy samples and 13% (61/472) of primary samples. Overall, 12% (62/516) of patients had at least 1 tier I/II variant in a homologous recombination repair gene, whereas 2.9% (10/516) had at least 1 tier I/II variant in a mismatch repair gene. The presence of a tier I/II variant was not significantly associated with the grade group (GG) or presence of intraductal/cribriform carcinoma in the primary tumor. Among the 309 reflex-tested hormone-naive primary tumors, tier I/II variants were identified in 10% (31/309) of cases, which is as follows: 9.2% (9/98) GG2; 9% (9/100) GG3; 9.1% (4/44) GG4; and 13.4% (9/67) GG5 cases. Our findings confirm the use of somatic tumor testing in detecting variants of clinical significance in PCa and provide insights that can inform the design of testing strategies. Pathologist-initiated reflex testing streamlines the availability of the results for clinical decision-making; however, pathologic parameters such as GG and the presence of intraductal/cribriform carcinoma may not be reliable to guide patient selection.

3.
Diagn Cytopathol ; 52(3): 163-170, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38095142

ABSTRACT

BACKGROUND: PD-L1 immunoexpression in head and neck squamous-cell carcinoma (HNSCC) determines immunotherapy eligibility. Patients are often diagnosed using fine-needle aspiration (FNA) of metastatic lymph nodes, however, the cytohistologic correlation of the combined positive score (CPS) is largely unknown. METHODS: This study retrospectively identified 96 paired histologic (HS) and cytologic specimens (CyS), between 2016 and 2020, diagnosed with HNSCC. Cases with <100 tumor cells (n = 54) or missing block(s) (n = 8) were excluded. All 34 case pairs were scored with CPS using the PD-L1 22C3 pharmDx assay at clinically relevant cut-offs of <1%, 1%-19%, and ≥20% independently by three observers blinded to the case pairs (CyS with corresponding HS). RESULTS: The CPS (<1/1-19/≥20) for CyS and HS were as follows: 10(29.4%)/10(29.4%)/14(41.2%) and 2(5.9%)/13(38.2%)/19(55.9%), respectively. There was fair overall cytohistologic agreement (OA) of 76.5% (k = 0.261) at the CPS cut-off of 1. The OA did not differ significantly between site-matched (n = 13) and -unmatched (n = 21) case pairs (p = .4653). CyS has a specificity and positive predictive value (PPV) of 100% but a negative predictive value (NPV) of only 20% as compared to its paired HS. CONCLUSIONS: Our study demonstrates fair CPS cytohistologic correlation in HNSCC specimens using the PD-L1 IHC 22C3 pharmDx assay with high PPV but low NPV. This suggest that determining PD-L1 status in FNA specimens can play an important role in the clinical management of HNSCC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Head and Neck Neoplasms , Lung Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Biopsy, Fine-Needle , Retrospective Studies , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Immunohistochemistry , Biomarkers, Tumor/metabolism
4.
Mod Pathol ; 37(1): 100384, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37972928

ABSTRACT

Tumor-agnostic testing for NTRK1-3 gene rearrangements is required to identify patients who may benefit from TRK inhibitor therapies. The overarching objective of this study was to establish a high-quality pan-TRK immunohistochemistry (IHC) screening assay among 18 large regional pathology laboratories across Canada using pan-TRK monoclonal antibody clone EPR17341 in a ring study design. TRK-fusion positive and negative tumor samples were collected from participating sites, with fusion status confirmed by panel next-generation sequencing assays. Each laboratory received: (1) unstained sections from 30 cases of TRK-fusion-positive or -negative tumors, (2) 2 types of reference standards: TRK calibrator slides and IHC critical assay performance controls (iCAPCs), (3) EPR17341 antibody, and (4) suggestions for developing IHC protocols. Participants were asked to optimize the IHC protocol for their instruments and detection systems by using iCAPCs, to stain the 30 study cases, and to report the percentage scores for membranous, cytoplasmic, and nuclear staining. TRK calibrators were used to assess the analytical sensitivity of IHC protocols developed by using the 2 reference standards. Fifteen of 18 laboratories achieved diagnostic sensitivity of 100% against next-generation sequencing. The diagnostic specificity ranged from 40% to 90%. The results did not differ significantly between positive scores based on the presence of any type of staining vs the presence of overall staining in ≥1% of cells. The median limit of detection measured by TRK calibrators was 76,000 molecules/cell (range 38,000 to >200,000 molecules/cell). Three different patterns of staining were observed in 19 TRK-positive cases, cytoplasmic-only in 7 samples, nuclear and cytoplasmic in 9 samples, and cytoplasmic and membranous in 3 samples. The Canadian multicentric pan-TRK study illustrates a successful strategy to accelerate the multicenter harmonization and implementation of pan-TRK immunohistochemical screening that achieves high diagnostic sensitivity by using laboratory-developed tests where laboratories used centrally developed reference materials. The measurement of analytical sensitivity by using TRK calibrators provided additional insights into IHC protocol performance.


Subject(s)
Neoplasms , Humans , Immunohistochemistry , Canada , Antibodies, Monoclonal , Receptor, trkA/genetics , Oncogene Proteins, Fusion/genetics , Biomarkers, Tumor/genetics
5.
Lung Cancer ; 160: 127-135, 2021 10.
Article in English | MEDLINE | ID: mdl-34509095

ABSTRACT

Patients with non-small cell lung cancer (NSCLC) harboring ROS proto-oncogene 1 (ROS1) gene rearrangements show dramatic response to the tyrosine kinase inhibitor (TKI) crizotinib. Current best practice guidelines recommend that all advanced stage non-squamous NSCLC patients be also tested for ROS1 gene rearrangements. Several studies have suggested that ROS1 immunohistochemistry (IHC) using the D4D6 antibody may be used to screen for ROS1 fusion positive lung cancers, with assays showing high sensitivity but moderate to high specificity. A break apart fluorescence in situ hybridization (FISH) test is then used to confirm the presence of ROS1 gene rearrangement. The goal of Canadian ROS1 (CROS) study was to harmonize ROS1 laboratory developed testing (LDT) by using IHC and FISH assays to detect ROS1 rearranged lung cancers across Canadian pathology laboratories. Cell lines expressing different levels of ROS1 (high, low, none) were used to calibrate IHC protocols after which participating laboratories ran the calibrated protocols on a reference set of 24 NSCLC cases (9 ROS1 rearranged tumors and 15 ROS1 non-rearranged tumors as determined by FISH). Results were compared using a centralized readout. The stained slides were evaluated for the cellular localization of staining, intensity of staining, the presence of staining in non-tumor cells, the presence of non-specific staining (e.g. necrosis, extracellular mater, other) and the percent positive cells. H-score was also determined for each tumor. Analytical sensitivity and specificity harmonization was achieved by using low limit of detection (LOD) as either any positivity in the U118 cell line or H-score of 200 with the HCC78 cell line. An overall diagnostic sensitivity and specificity of up to 100% and 99% respectively was achieved for ROS1 IHC testing (relative to FISH) using an adjusted H-score readout on the reference cases. This study confirms that LDT ROS1 IHC assays can be highly sensitive and specific for detection of ROS1 rearrangements in NSCLC. As NSCLC can demonstrate ROS1 IHC positivity in FISH-negative cases, the degree of the specificity of the IHC assay, especially in highly sensitive protocols, is mostly dependent on the readout cut-off threshold. As ROS1 IHC is a screening assay for a rare rearrangements in NSCLC, we recommend adjustment of the readout threshold in order to balance specificity, rather than decreasing the overall analytical and diagnostic sensitivity of the protocols.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Canada , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Protein-Tyrosine Kinases/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/genetics , Proto-Oncogenes , Reactive Oxygen Species
6.
Mod Pathol ; 33(1): 4-17, 2020 01.
Article in English | MEDLINE | ID: mdl-31383961

ABSTRACT

Different clones, protocol conditions, instruments, and scoring/readout methods may pose challenges in introducing different PD-L1 assays for immunotherapy. The diagnostic accuracy of using different PD-L1 assays interchangeably for various purposes is unknown. The primary objective of this meta-analysis was to address PD-L1 assay interchangeability based on assay diagnostic accuracy for established clinical uses/purposes. A systematic search of the MEDLINE database using PubMed platform was conducted using "PD-L1" as a search term for 01/01/2015 to 31/08/2018, with limitations "English" and "human". 2,515 abstracts were reviewed to select for original contributions only. 57 studies on comparison of two or more PD-L1 assays were fully reviewed. 22 publications were selected for meta-analysis. Additional data were requested from authors of 20/22 studies in order to enable the meta-analysis. Modified GRADE and QUADAS-2 criteria were used for grading published evidence and designing data abstraction templates for extraction by reviewers. PRISMA was used to guide reporting of systematic review and meta-analysis and STARD 2015 for reporting diagnostic accuracy study. CLSI EP12-A2 was used to guide test comparisons. Data were pooled using random-effects model. The main outcome measure was diagnostic accuracy of various PD-L1 assays. The 22 included studies provided 376 2×2 contingency tables for analyses. Results of our study suggest that, when the testing laboratory is not able to use an Food and Drug Administration-approved companion diagnostic(s) for PD-L1 assessment for its specific clinical purpose(s), it is better to develop a properly validated laboratory developed test for the same purpose(s) as the original PD-L1 Food and Drug Administration-approved immunohistochemistry companion diagnostic, than to replace the original PD-L1 Food and Drug Administration-approved immunohistochemistry companion diagnostic with a another PD-L1 Food and Drug Administration-approved companion diagnostic that was developed for a different purpose.


Subject(s)
B7-H1 Antigen/analysis , Immunohistochemistry/methods , Humans , Immunohistochemistry/standards
7.
Appl Immunohistochem Mol Morphol ; 27(10): 699-714, 2019.
Article in English | MEDLINE | ID: mdl-31584451

ABSTRACT

Since 2014, programmed cell death protein 1 (PD-1)/programmed cell death ligand 1 (PD-L1) checkpoint inhibitors have been approved by various regulatory agencies for the treatment of multiple cancers including melanoma, lung cancer, urothelial carcinoma, renal cell carcinoma, head and neck cancer, classical Hodgkin lymphoma, colorectal cancer, gastroesophageal cancer, hepatocellular cancer, and other solid tumors. Of these approved drug/disease combinations, a subset also has regulatory agency-approved, commercially available companion/complementary diagnostic assays that were clinically validated using data from their corresponding clinical trials. The objective of this document is to provide evidence-based guidance to assist clinical laboratories in establishing fit-for-purpose PD-L1 biomarker assays that can accurately identify patients with specific tumor types who may respond to specific approved immuno-oncology therapies targeting the PD-1/PD-L1 checkpoint. These recommendations are issued as 38 Guideline Statements that address (i) assay development for surgical pathology and cytopathology specimens, (ii) reporting elements, and (iii) quality assurance (including validation/verification, internal quality assurance, and external quality assurance). The intent of this work is to provide recommendations that are relevant to any tumor type, are universally applicable and can be implemented by any clinical immunohistochemistry laboratory performing predictive PD-L1 immunohistochemistry testing.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , B7-H1 Antigen/metabolism , Biomarkers/metabolism , Immunotherapy/methods , Neoplasms/therapy , B7-H1 Antigen/antagonists & inhibitors , Canada , Clinical Laboratory Techniques , Evidence-Based Medicine , Humans , Immunohistochemistry , Neoplasms/diagnosis , Neoplasms/immunology , Patient Selection , Practice Guidelines as Topic , Predictive Value of Tests , Prognosis , Quality Assurance, Health Care
8.
Appl Immunohistochem Mol Morphol ; 27(4): 251-257, 2019 04.
Article in English | MEDLINE | ID: mdl-30789848

ABSTRACT

PD-L1 testing by immunohistochemistry (IHC) has presented significant challenges not only for clinical laboratories, but also for external quality assurance (EQA) entities that provide proficiency testing (PT) for clinical laboratories. Canadian Immunohistochemistry Quality Control (CIQC) has used educational runs to explore approaches to sample design and analysis of results that would enhance patient safety. As PT for predictive biomarkers requires modeling at every level (design of the run, assessment of the run, and reporting of "pass" or "fail") based on "fit-for-purpose" principles, CIQC has applied those principles to PD-L1 PT runs. Each laboratory received unstained slides with TMA tissue cores from 104 randomly selected primary NSCLC and tonsil tissues to test with their current PD-L1 assay. Diagnostic sensitivity and specificity were calculated against designated gold standards based on the "3D" approach (drug-disease-diagnostic assay). Depending on the selection of fit-for-purpose gold standards and also on the selection of what was considered fit-for-purpose cut-off points, great variation in the performance (accuracy) of both companion/complementary diagnostic assays and laboratory developed tests was seen. "Fit-for-purpose" in PT for PD-L1 testing entails that the purpose(s) of each PT run is declared a priori, that the PT program has selected/designated purpose-specific gold standard results for the PT challenge, and that the PT materials for the PT run are designed and constructed to enable calculations of diagnostic accuracy.


Subject(s)
B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung , Laboratory Proficiency Testing , Lung Neoplasms , Canada , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Female , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Sensitivity and Specificity
9.
J Clin Pathol ; 72(3): 237-243, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30782622

ABSTRACT

The ability to effectively monitor key indicators is important for continuous quality improvement in laboratory immunohistochemistry. This article deals specifically with laboratory turnaround time (TAT) as a key delivery indicator and the impact of laboratory workflow on laboratory TATs. While our laboratory has traditionally relied on the manual calculation of slide-TAT (S-TAT) to monitor delivery, we have determined that automated calculation of case-TAT (C-TAT) would be superior as a delivery indicator. AABACUS (Automatable Activity-Based Approach to Complexity Unit Scoring) is an activity-based workload model designed to function primarily as a decision support tool to monitor pathologist staffing levels. We devised a high-level proof-of-principle approach to determine whether it is possible to apply AABACUS as a decision support tool for quality improvement through analysis of alternative laboratory workflows that have potential to impact C-TAT. Our use of AABACUS in this proof-of-principle quality improvement endeavour was two-fold: (1) we leveraged the ability of AABACUS to link data at the slide level to data at the case level, which enabled the automated calculation of C-TAT; and (2) we adapted AABACUS to evaluate the impact of laboratory workflow activities (specifically workflow bifurcation activities) on the calculated C-TATs. We have coined the term 'L-AABACUS' to describe the adaptation of AABACUS to the analysis of laboratory workflow.


Subject(s)
Decision Support Techniques , Laboratories, Hospital/organization & administration , Quality Improvement , Workflow , Humans , Time Factors , Workload
10.
J Clin Pathol ; 72(5): 363-367, 2019 May.
Article in English | MEDLINE | ID: mdl-30755496

ABSTRACT

AIMS: Renal tumour biopsy (RTB) is increasingly recognised as a useful diagnostic tool in the management of small renal masses, particularly those that are incidentally found. Intratumoural heterogeneity with respect to morphology, grade and molecular features represents a frequently identified limitation to the use of RTB. While previous studies have evaluated pathological correlation between RTB and nephrectomy, no studies to date have focused specifically on the role of RTB for the diagnosis of papillary renal cell carcinoma (PRCC) and its further subclassification into clinically relevant subtypes. METHODS: This single-institution study evaluated 60 cases of PRCC for concordance between RTB and nephrectomy with respect to diagnosis, grading and subtyping (type 1/type 2). RESULTS: We observed 93% concordance (55 of 59 evaluable cases) between RTB and nephrectomy for the diagnosis of PRCC, although seven tumours (12%) were undergraded on RTB. Subtyping of PRCC on RTB was concordant with nephrectomy in 89% of cases reported as type 1 PRCC on RTB (31/35), but only 40% of cases reported as type 2 PRCC on RTB (4/10). Morphological misclassification of PRCC on RTB was most likely to occur in tumours showing a solid growth pattern. Discordant PRCC subtyping most often occurred in tumours with eosinophilia/oncocytic change. CONCLUSION: There was good concordance between RTB and nephrectomy for the primary diagnosis of PRCC. Although further subtyping of PRCC can aid therapeutic stratification, this can be challenging on RTB and tumours with overlapping or ambiguous features are best reported as PRCC not otherwise specified pending development of more robust methods to facilitate definitive subclassification.


Subject(s)
Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Nephrectomy , Adult , Aged , Biopsy , Carcinoma, Renal Cell/classification , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/surgery , Databases, Factual , Female , Humans , Kidney Neoplasms/classification , Kidney Neoplasms/diagnosis , Kidney Neoplasms/surgery , Male , Middle Aged , Retrospective Studies
11.
Appl Immunohistochem Mol Morphol ; 26(5): 299-304, 2018.
Article in English | MEDLINE | ID: mdl-29734239

ABSTRACT

OBJECTIVES: The occurrence of uneven staining (UES) in automated immunohistochemistry (IHC) has been experienced by clinical laboratories and has the potential to confound readout, interpretation, and reporting of IHC assays despite the presence optimally stained on-slide controls. However, there are no studies of this phenomenon in regard to the type, frequency, and association with different automated IHC platforms. We studied the occurrence of UES in automated IHC assays with real world examples from clinical practice and by using a laboratory developed methodology to monitor baseline and periodic performance of automated IHC instruments. MATERIALS AND METHODS: Sections of formalin-fixed, paraffin-embedded normal liver tissue were mounted on 180 glass slides and stained for HepPar1 on 6 automated IHC instruments (4 different models from 3 different manufacturers). Macroscopic and microscopic defects of staining were recorded. RESULTS: Only 8% of slides showed completely uniform staining. UES, including areas of both increased and decreased staining, occurred with all instruments. Decreased staining was often zonal, involving large regions of the slide. Decreased staining mostly localized in an instrument-dependent manner. Increased staining tended to occur in small foci with a random distribution. CONCLUSIONS: The common occurrence of UES (particularly decreased staining) has important implications for the reliable read-out of IHC assays on biopsy samples. Baseline and periodic quality assurance testing for UES is recommended for all automated IHC instruments.


Subject(s)
Automation, Laboratory/methods , Immunohistochemistry/methods , Liver/pathology , Staining and Labeling/methods , Biopsy , Cold Temperature , Hot Temperature , Humans , Immunohistochemistry/instrumentation , Quality Control
12.
Appl Immunohistochem Mol Morphol ; 25(2): 79-85, 2017 02.
Article in English | MEDLINE | ID: mdl-28182587

ABSTRACT

All laboratory tests have test performance characteristics (TPCs), whether or not they are explicitly known to the laboratorian or the pathologist. TPCs are thus also an integral characteristic of immunohistochemistry (IHC) tests and other in situ, cell-based molecular assays such as DNA or RNA in situ hybridization or aptamer-based testing. Because of their descriptive, in situ, cell-based nature, IHC tests have a limited repertoire of appropriate TPCs. Although only a few TPCs are relevant to IHC, proper selection of informative TPCs is nonetheless essential for the development of and adherence to appropriate quality assurance measures in the IHC laboratory. This paper describes the TPCs that are relevant to IHC testing and emphasizes the role of TPCs in the validation of IHC tests. This is part 2 of the 4-part series "Evolution of Quality Assurance for Clinical Immunohistochemistry in the Era of Precision Medicine."


Subject(s)
Immunohistochemistry/standards , Precision Medicine , Humans , Reproducibility of Results , Sensitivity and Specificity
13.
Appl Immunohistochem Mol Morphol ; 25(3): 151-159, 2017 03.
Article in English | MEDLINE | ID: mdl-28187030

ABSTRACT

Validation of immunohistochemistry (IHC) assays is a subject that is of great importance to clinical practice as well as basic research and clinical trials. When applied to clinical practice and focused on patient safety, validation of IHC assays creates objective evidence that IHC assays used for patient care are "fit-for-purpose." Validation of IHC assays needs to be properly informed by and modeled to assess the purpose of the IHC assay, which will further determine what sphere of validation is required, as well as the scope, type, and tier of technical validation. These concepts will be defined in this review, part 3 of the 4-part series "Evolution of Quality Assurance for Clinical Immunohistochemistry in the Era of Precision Medicine."


Subject(s)
Laboratories/standards , Precision Medicine , Quality Control , Immunohistochemistry
14.
Appl Immunohistochem Mol Morphol ; 25(5): 308-312, 2017.
Article in English | MEDLINE | ID: mdl-26657875

ABSTRACT

Appropriate controls are critical for the correct interpretation of immunohistochemistry (IHC) assays and help to detect unsuccessful/suboptimal slides. We performed an audit of slides that were designated as being "failed" by the IHC laboratory (ie, laboratory-failed slides) of a large North American oncology and transplant center. All slides were run with on-slide controls. The study included analysis of only those failed slides where staining of both internal and external controls were unsuccessful/suboptimal in a period of 65 days. Failed slides were categorized based on the reason why the laboratory failed the slides. The study compared frequencies of failed slides across 9 automated stainers from 2 manufacturers and between class 1 and class 2 biomarkers. Distinction between "failed slides" and "false-negative/false-positive tests" is emphasized. The study included 22,234 IHC slides in the study period. Of those, 452 (2%) were designated as "failed" by the laboratory. Class 1 and class 2 tests showed failure rates of 0.8% and 9%, respectively. The most frequent reason for failed slides on one platform related to "no or weak staining," whereas the other had more failed slides due to "high signal-to-noise ratio" (P<0.0001, χ test). Although the slides were run in groups of the same as well as different IHC protocols, unsuccessful/suboptimal testing typically manifested as individual slides (92%) and not as groups of slides; this indicates that so-called "batch controls" are not suitable as controls for automated platforms. We conclude that in the era of automated IHC staining platforms, on-slide controls allow for the proper identification of IHC slides that should be failed by the IHC laboratory and represent a powerful tool for preventing the reporting of false-negative/false-positive tests.


Subject(s)
Clinical Audit , Immunohistochemistry/standards , Laboratories/standards , Quality Control , Diagnostic Errors , Humans , Reference Standards
15.
Appl Immunohistochem Mol Morphol ; 25(4): 227-230, 2017 04.
Article in English | MEDLINE | ID: mdl-27941560

ABSTRACT

The numbers of diagnostic, prognostic, and predictive immunohistochemistry (IHC) tests are increasing; the implementation and validation of new IHC tests, revalidation of existing tests, as well as the on-going need for daily quality assurance monitoring present significant challenges to clinical laboratories. There is a need for proper quality tools, specifically tissue tools that will enable laboratories to successfully carry out these processes. This paper clarifies, through the lens of laboratory tissue tools, how validation, verification, and revalidation of IHC tests can be performed in order to develop and maintain high quality "fit-for-purpose" IHC testing in the era of precision medicine. This is the final part of the 4-part series "Evolution of Quality Assurance for Clinical Immunohistochemistry in the Era of Precision Medicine."


Subject(s)
Immunohistochemistry/methods , Laboratories , Quality Assurance, Health Care , Clinical Laboratory Techniques , Humans , Immunohistochemistry/instrumentation , Precision Medicine
16.
Appl Immunohistochem Mol Morphol ; 25(1): 4-11, 2017 01.
Article in English | MEDLINE | ID: mdl-27922482

ABSTRACT

Technical progress in immunohistochemistry (IHC) as well as the increased utility of IHC for biomarker testing in precision medicine avails us of the opportunity to reassess clinical IHC as a laboratory test and its proper characterization as a special type of immunoassay. IHC, as used in current clinical applications, is a descriptive, qualitative, cell-based, usually nonlinear, in situ protein immunoassay, for which the readout of the results is principally performed by pathologists rather than by the instruments on which the immunoassay is performed. This modus operandi is in contrast to other assays where the instrument also performs the readout of the test result (eg, nephelometry readers, mass spectrometry readers, etc.). The readouts (results) of IHC tests are used either by pathologists for diagnostic purposes or by treating physicians (eg, oncologists) for patient management decisions, the need for further testing, or follow-up. This paper highlights the distinction between the original purpose for which an IHC test is developed and its subsequent clinical uses, as well as the role of pathologists in the analytical and postanalytical phases of IHC testing. This paper is the first of a 4-part series, under the general title of "Evolution of Quality Assurance for Clinical Immunohistochemistry in the Era of Precision Medicine."


Subject(s)
Biomarkers/metabolism , Precision Medicine , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence
17.
J Clin Pathol ; 69(1): 47-52, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26193899

ABSTRACT

AIMS: First, to determine the frequency of intravascular granulomas (IVGs) in seminomas and assess for the presence of entrapped seminoma cells. Second, to identify the relationship of this unusual form of vascular space invasion with tumour relapse and/or dissemination. METHODS: 86 cases of seminoma were reviewed to identify IVGs. Immunostaining for OCT3/4 and CD68 was performed. Pathological stage, presence of conventional vascular and rete testis invasion, parenchymal granulomas and follow-up were recorded. Multivariable analysis incorporating tumour size, vascular invasion (conventional granulomas and IVGs) and rete testis invasion was performed. RESULTS: IVGs were identified in 13 cases (13/86). CD68 confirmed histiocytes in all cases. OCT3/4 identified tumour cells in 9/13 seminomas. 27 patients had disease progression with either dissemination at presentation (n=11) or relapse (n=16). Of these 27 patients, 8 had IVG (29.6%). By comparison, 6 of 57 clinical stage 1 seminomas that did not relapse had IVG (10.53%). Multivariable analysis revealed that no single parameter was statistically significant at predicting tumour relapse and/or dissemination (size: HR 1.65; CI 0.71 to 3.82, p=0.24, rete testis invasion: HR 1.04; CI 0.48 to 2.26, p=0.92, lymphovascular space invasion/IVG: HR 1.62; CI 0.65 to 4.01, p=0.30). CONCLUSIONS: IVGs may represent a previously unrecognised form of vascular space invasion in seminomas. Studies on larger cohorts are needed to demonstrate its clinical value.


Subject(s)
Blood Vessels/pathology , Granuloma/pathology , Neoplastic Cells, Circulating/pathology , Seminoma/secondary , Testicular Neoplasms/pathology , Adult , Aged , Antigens, CD/analysis , Antigens, Differentiation, Myelomonocytic/analysis , Biomarkers, Tumor/analysis , Blood Vessels/chemistry , Disease Progression , Granuloma/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Multivariate Analysis , Neoplasm Invasiveness , Neoplasm Recurrence, Local , Neoplasm Staging , Neoplastic Cells, Circulating/chemistry , Octamer Transcription Factor-3/analysis , Predictive Value of Tests , Proportional Hazards Models , Retrospective Studies , Risk Factors , Seminoma/chemistry , Seminoma/therapy , Testicular Neoplasms/chemistry , Testicular Neoplasms/therapy , Time Factors , Treatment Outcome , Tumor Burden , Young Adult
18.
Appl Immunohistochem Mol Morphol ; 23(10): 677-81, 2015.
Article in English | MEDLINE | ID: mdl-26551338

ABSTRACT

Intrachromosomal rearrangements involving the ALK gene are found in 3% to 5% of non-small cell lung cancers. Crizotinib is a tyrosine kinase inhibitor that has been shown to prolong progression-free survival in patients with advanced non-small cell lung cancer harboring ALK gene rearrangements. In Canada, ALK immunohistochemistry (IHC) is used as a screening test before confirmation by fluorescence in situ hybridization (FISH). Canadian Immunohistochemistry Quality Control (CIQC) provides ALK (Lung Cancer) proficiency testing (PT) for Canadian IHC laboratories. Samples included 32 previously characterized cases (IHC and FISH) either from the Canadian ALK (CALK) project or from CIQC reference laboratories. The same design was used for both runs. A total of 20 laboratories participated in Run 1 and 22 in Run 2. Some laboratories participated in the anticipation of future need and used the PT exercise as a part of test development and validation. Results of the IHC testing were first self-reported using the CIQC TMA Scorer and then evaluated by expert assessment. FISH results were self-reported only. Participants also reported details about IHC and FISH protocols. The κ-values were calculated, for which values >0.80 were used as acceptable results, respectively. The pass rate between the 2 runs and between different primary antibodies were compared. Six of the 22 protocols (27%) in Run 1 and 15 of the 22 (68%) protocols in Run 2 passed the CIQC PT criteria for IHC testing. The increase in the pass rate for Run 2 was significant (P=0.03, Wilcoxon signed-rank test). All reported FISH results were correct. CALK laboratories had significantly higher κ-values than non-CALK laboratories (P=0.002, t test). PT for IHC for rare diseases such as ALK-positive lung cancer is feasible, but challenging. The academic nature of the CIQC program and collaboration on a national level facilitated the development of appropriate PT samples. Participating laboratories made use of the PT exercise either to confirm that their testing was properly calibrated or to improve their protocols, which was confirmed by the achievement of significantly better results in Run 2. They also used CIQC's PT program for new test development and optimization.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/enzymology , Lung Neoplasms/pathology , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Female , Humans , Immunohistochemistry/standards , In Situ Hybridization/standards , Male , Quality Control
19.
Mod Pathol ; 28(3): 324-39, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25216230

ABSTRACT

Pathologists provide diagnoses relevant to the disease state of the patient and identify specific tissue characteristics relevant to response to therapy and prognosis. As personalized medicine evolves, there is a trend for increased demand of tissue-derived parameters. Pathologists perform increasingly complex analyses on the same 'cases'. Traditional methods of workload assessment and reimbursement, based on number of cases sometimes with a modifier (eg, the relative value unit (RVU) system used in the United States), often grossly underestimate the amount of work needed for complex cases and may overvalue simple, small biopsy cases. We describe a new approach to pathologist workload measurement that aligns with this new practice paradigm. Our multisite institution with geographically diverse partner institutions has developed the Automatable Activity-Based Approach to Complexity Unit Scoring (AABACUS) model that captures pathologists' clinical activities from parameters documented in departmental laboratory information systems (LISs). The model's algorithm includes: 'capture', 'export', 'identify', 'count', 'score', 'attribute', 'filter', and 'assess filtered results'. Captured data include specimen acquisition, handling, analysis, and reporting activities. Activities were counted and complexity units (CUs) generated using a complexity factor for each activity. CUs were compared between institutions, practice groups, and practice types and evaluated over a 5-year period (2008-2012). The annual load of a clinical service pathologist, irrespective of subspecialty, was ∼40,000 CUs using relative benchmarking. The model detected changing practice patterns and was appropriate for monitoring clinical workload for anatomical pathology, neuropathology, and hematopathology in academic and community settings, and encompassing subspecialty and generalist practices. AABACUS is objective, can be integrated with an LIS and automated, is reproducible, backwards compatible, and future adaptable. It can be applied as a robust decision support tool for the assessment of overall and targeted staffing needs as well as utilization analyses for resource allocation.


Subject(s)
Algorithms , Pathology , Physicians , Workload , Humans
20.
Am J Clin Pathol ; 142(5): 629-33, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25319977

ABSTRACT

OBJECTIVES: Formalin-fixed, paraffin-embedded unstained archived diagnostic tissue sections are frequently exchanged between clinical laboratories for immunohistochemical staining. The manner in which such sections are prepared represents a type of preanalytical variable that must be taken into account given the growing importance of immunohistochemical assays, especially predictive and prognostic tests, in personalized medicine. METHODS: Recommendations were derived from review of the literature and expert consensus of the Canadian Association of Pathologists-Association canadienne des pathologists National Standards Committee for High Complexity Testing/Immunohistochemistry. RESULTS: Relevant considerations include the type of glass slide on which to mount the unstained sections; the thickness of the tissue sections; the time from slide preparation to testing; the environment, particularly the temperature at which the unstained sections will be maintained prior to testing; the inclusion of on-slide positive control tissue where possible; and whether patient identifier(s) should be included on slide labels. CONCLUSIONS: Clear communication between requesting and releasing laboratories will facilitate the proper preparation of unstained sections and also ensure that applicable privacy considerations are addressed.


Subject(s)
Clinical Laboratory Techniques , Immunohistochemistry/standards , Paraffin Embedding/standards , Practice Guidelines as Topic , Archives , Canada , Clinical Laboratory Techniques/standards , Formaldehyde/standards , Humans , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL
...