1
|
Fassan M, Kuwata T, Matkowskyj KA, Röcken C, Rüschoff J. Claudin-18.2 Immunohistochemical Evaluation in Gastric and Gastroesophageal Junction Adenocarcinomas to Direct Targeted Therapy: A Practical Approach. Mod Pathol 2024; 37:100589. [PMID: 39098518 DOI: 10.1016/j.modpat.2024.100589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/14/2024] [Accepted: 07/29/2024] [Indexed: 08/06/2024]
Abstract
Claudin-18.2 (CLDN18.2) expression evaluated by immunohistochemistry is a new biomarker for gastric and gastroesophageal junction adenocarcinomas that will soon have market authorization for implementation into routine clinical practice. Despite successful testing in the setting of clinical trials, no specific practical testing guidelines have been proposed. Several preanalytical and analytical variables may interfere with adequate CLDN18.2 staining interpretation; thus, this article provides practical guidance on CLDN18.2 testing and scoring in gastric and gastroesophageal junction adenocarcinomas to identify patients who may respond to targeted therapy with monoclonal antibodies directed against CLDN18.2. Based on available data, moderate to strong (2+/3+) membrane staining in ≥75% of adenocarcinoma cells is the proposed cutoff for clinical use of monoclonal antibody anti-CLDN18.2 (zolbetuximab).
Collapse
Affiliation(s)
- Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua, Italy; Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy.
| | - Takeshi Kuwata
- Department of Genetic Medicine and Services, National Cancer Center Hospital East, Chiba, Japan
| | | | - Christoph Röcken
- Department of Pathology, University-Hospital Schleswig-Holstein (UKSH), Kiel, Germany
| | - Josef Rüschoff
- Discovery Life Sciences Biomarker Services, Kassel, Germany
| |
Collapse
|
2
|
Heymann JJ, Pineda CM, Booth CN, Jenkins E, Menke JR, Monaco SE, Nayar R, Nishino M, Roy-Chowdhuri S, Ruiz-Cordero R, Russell DK, Saqi A, Sundling KE, Thrall MJ, Torous VF, VandenBussche CJ, VanderLaan PA, Zhang ML, Siddiqui MT. Tools, techniques, and challenges in preparing cytology specimens for ancillary studies: results of the ASC Optimizing Cytology and Small Biopsy Specimen Processing for Ancillary Studies task force survey. J Am Soc Cytopathol 2024:S2213-2945(24)00215-1. [PMID: 39547835 DOI: 10.1016/j.jasc.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/25/2024] [Accepted: 10/09/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION Ancillary testing on cytopathology and other small biopsy specimens is crucial for diagnosis and provides critical information to clinicians. Testing is dependent on preanalytic factors and would benefit from standardization of specimen collection protocols across laboratories. To assess institutional practices and areas of need for evidence-based standards, we surveyed current practices across cytopathology laboratories. MATERIALS AND METHODS A twelve-question electronic survey was distributed to American Society of Cytopathology (ASC) members through email, social media, and the ASC from January 8, 2024 to March 1, 2024. Survey responses were tabulated. RESULTS Of 294 respondents, 257 (87%) completed at least 10/12 questions. Formalin-fixed, paraffin-embedded cell blocks (CBs) are utilized for immunohistochemistry, molecular testing, and in situ hybridization by 89%, 84%, and 71% of respondents, respectively. For fine needle aspirations, no collection medium is utilized by a majority of respondents. In contrast, 61% utilize no collection medium for fluids; 64% predominantly utilize liquid-based preservatives for other exfoliative specimens. For CB preparation, 58% of respondents use coagulating agent; 67% use no fixative before formalin. The two most significant factors limiting clinical utility of ancillary testing in cytology specimens are low cellularity and lack of validation (49% and 23% of respondents, respectively). CONCLUSIONS There is wide variation in current practices among laboratories, reflecting lack of consensus. Although laboratories utilize different collection media for different specimen types, for CB utilization, current survey results are similar to those reported previously. ASC has convened a task force to facilitate specimen standardization and minimize variability among preanalytic factors.
Collapse
Affiliation(s)
- Jonas J Heymann
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital-Weill Cornell Medicine, New York, New York.
| | - Cristiana M Pineda
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | | | | | - Joshua R Menke
- Department of Pathology, Stanford University, Stanford, California
| | - Sara E Monaco
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, Pennsylvania
| | - Ritu Nayar
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Michiya Nishino
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Sinchita Roy-Chowdhuri
- Division of Pathology and Laboratory Medicine, Department of Anatomic Pathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Roberto Ruiz-Cordero
- Department of Pathology and Laboratory Medicine, Miller School of Medicine, University of Miami, Miami, Florida
| | | | - Anjali Saqi
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, New York
| | - Kaitlin E Sundling
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Michael J Thrall
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | - Vanda F Torous
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | - Paul A VanderLaan
- Department of Pathology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - M Lisa Zhang
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Momin T Siddiqui
- Department of Pathology and Laboratory Medicine, New York-Presbyterian Hospital-Weill Cornell Medicine, New York, New York
| |
Collapse
|
3
|
Carvalho FM, Carvalho JP. Unraveling the Heterogeneity of Deficiency of Mismatch Repair Proteins in Endometrial Cancer: Predictive Biomarkers and Assessment Challenges. Cancers (Basel) 2024; 16:3452. [PMID: 39456546 PMCID: PMC11505891 DOI: 10.3390/cancers16203452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Endometrial cancer (EC) poses a significant global health challenge, with increasing prevalence in 26 of 43 countries and over 13,000 deaths projected in the United States by 2024. This rise correlates with aging populations, the obesity epidemic, and changing reproductive patterns, including delayed childbearing. Despite the early diagnosis in 67% of cases, approximately 30% of cases present with regional or distant spread, leading to nearly 20% mortality rates. Unlike many cancers, EC mortality rates are escalating, outpacing therapeutic advancements until recently. One of the reasons for this was the lack of effective therapeutic options for advanced disease until recently. The introduction of immunotherapy has marked a turning point in EC treatment, particularly benefiting patients with defects in mismatch repair proteins (dMMRs). However, dMMR status alone does not ensure a favorable response, underscoring the need for precise patient selection. This review explores the pivotal role of mismatch repair proteins in EC, emphasizing their heterogeneity, the challenges in their assessment, and their potential as predictive biomarkers.
Collapse
Affiliation(s)
- Filomena M. Carvalho
- Department of Pathology, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo 01246-903, Brazil
| | - Jesus P. Carvalho
- Department of Obstetrics and Gynecology, Instituto do Cancer do Estado de Sao Paulo, Faculdade de Medicina da Universidade de Sao Paulo, São Paulo 01246-903, Brazil;
| |
Collapse
|
4
|
Hicks DG, Turner BM. Optimized biomarker evaluation and molecular testing in the era of breast cancer precision medicine. Biotech Histochem 2024:1-13. [PMID: 39258585 DOI: 10.1080/10520295.2024.2390179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024] Open
Abstract
Ground breaking advances in medicine, driven in part by major technologic developments in molecular biology have led us to a new model for cancer care that has been termed personalized, or precision medicine. Precision medicine is a model for making medical decisions that employs an innovative clinical approach and advanced tumor testing methods that are tailored to understanding an individual patient's tumor biology and the molecular drivers of their disease. This medical model includes a combination of diagnostic testing and specific treatment options that can be offered to patients at presentation and in theory throughout the course of their disease as new mutations arise with the development of disease recurrence. Although the precision medicine model offers incredible potential to transform cancer care, these advances are only meaningful when they reach the correct patients. The evolving paradigm of precision medicine is changing the practice of pathology, and the pathology community needs to be mindful of these changes because every tissue specimen represents a patient's life, and those patients are depending on the pathology community to handle their tissue correctly. The diagnostic tests performed in the pathology laboratory for precision medicine are increasingly complex, and pathologists along with the entire laboratory and clinical communities need to take steps to ensure that the right diagnosis is given to the right patient to inform the right treatment options, at the right time, along every step of the continuum of care for cancer patients. While hormone receptors and human epidermal growth factor receptor 2 (HER2) overexpression and/or amplification have been the mainstay for risk-stratification, and treatment decision making in breast cancer since the early 2000's, the seminal work on gene expression by Perou and colleagues in the early 2000's opened the door for molecular testing in the prognostic and predictive assessment of breast cancer. Molecular testing is now part of the standard of care in the precision medicine model for breast cancer care. In this article, the reader will gain a better understanding of how the lack of standardization of pre-analytic factors has the potential to negatively impact the quality of the tissue specimen for downstream biomarker and molecular testing, which ultimately can negatively affect patient care. The reader will also gain insight into the current climate surrounding molecular testing in breast cancer.
Collapse
Affiliation(s)
- David G Hicks
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| | - Bradley M Turner
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
5
|
Charania AA, Pokal AG, Zuaiter DR, Crawford CL, Esnakula AK, Islam M, Kim AC, Budhwani KI. A comprehensive preanalytical protocol for fresh solid tumor biospecimens. Methods 2024; 229:108-114. [PMID: 38909975 PMCID: PMC11296924 DOI: 10.1016/j.ymeth.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 06/25/2024] Open
Abstract
Nearly seventy percent of diagnostic lab test errors occur due to variability in preanalytical factors. These are the parameters involved with all aspects of tissue processing, starting from the time tissue is collected from the patient in the operating room, until it is received and tested in the laboratory. While there are several protocols for transporting fixed tissue, organs, and liquid biopsies, such protocols are lacking for transport and handling of live solid tumor tissue specimens. There is a critical need to establish preanalytical protocols to reduce variability in biospecimen integrity and improve diagnostics for personalized medicine. Here, we provide a comprehensive protocol for the standard collection, handling, packaging, cold-chain logistics, and receipt of solid tumor tissue biospecimens to preserve tissue viability.
Collapse
Affiliation(s)
| | | | | | | | - Ashwini K Esnakula
- The Ohio State University Wexner Medical Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Mozaffarul Islam
- The Ohio State University Wexner Medical Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | - Alex C Kim
- The Ohio State University Wexner Medical Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH, USA
| | | |
Collapse
|
6
|
Ribeiro R, Carvalho FM, Baiocchi G, Guindalini RSC, da Cunha JR, Anjos CHD, de Nadai Costa C, Gifoni ACLVC, Neto RC, Cagnacci AQC, Carneiro VCG, Calabrich A, Moretti-Marques R, Pinheiro RN, de Castro Ribeiro HS. Guidelines of the Brazilian Society of Surgical Oncology for anatomopathological, immunohistochemical, and molecular testing in female tumors. J Surg Oncol 2024; 130:882-895. [PMID: 39038206 DOI: 10.1002/jso.27717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/04/2024] [Indexed: 07/24/2024]
Abstract
INTRODUCTION Precision medicine has revolutionized oncology, providing more personalized diagnosis, treatment, and monitoring for patients with cancer. In the context of female-specific tumors, such as breast, ovarian, endometrial, and cervical cancer, proper tissue collection and handling are essential for obtaining tissue, immunohistochemical (IHC), and molecular data to guide therapeutic decisions. OBJECTIVES To establish guidelines for the collection and handling of tumor tissue, to enhance the quality of samples for histopathological, IHC, genomic, and molecular analyses. These guidelines are fundamental in informing therapeutic decisions in cancer treatment. METHOD The guidelines were developed by a multidisciplinary panel of renowned specialists between June 12, 2013 and February 12, 2024. Initially, the panel deliberated on critical and controversial topics related to conducting precision medicine studies focusing on female tumors. Subsequently, 22 pivotal topics were identified within the framework and assigned to groups. These groups reviewed relevant literature and drafted preliminary recommendations. Following this, the recommendations were reviewed by the coordinators and received unanimous approval. Finally, the groups made the final adjustments, classified the level of evidence, and ranked the recommendations. CONCLUSION The collection of surgical samples requires minimum quality standards to enable histopathological, IHC, genomic, and molecular analyses. These analyses provide crucial data for informing therapeutic decisions, significantly impacting potential survival gains for patients with female tumors.
Collapse
Affiliation(s)
- Reitan Ribeiro
- Department of Gynecology Oncology, Erasto Gaertner Hospital, Curitiba, Paraná, Brazil
| | - Filomena Marino Carvalho
- Department of Pathology, Faculdade de Medicina (FMUSP), Universidade de São Paulo, São Paulo, São Paulo, Brazil
| | - Glauco Baiocchi
- Department of Gynecologic Oncology, AC Camargo Cancer Center , São Paulo, São Paulo, Brazil
| | | | | | | | | | | | - Renato Cagnacci Neto
- Department of Mastology, Breast Cancer Reference Center, AC Camargo Cancer, CenterSão Paulo, São Paulo, Brazil
| | - Allyne Queiroz Carneiro Cagnacci
- Department of Oncology, Oncology Center, Hospital Alemão Oswaldo Cruz, São Paulo, São Paulo, Brazil
- Hereditary Cancer Department, Instituto do Câncer do Estado de São Paulo (ICESPSP), São Paulo, São Paulo, Brazil
| | - Vandré Cabral Gomes Carneiro
- Department of Gynecology Oncology, Instituto de Medicina Integral Professor Fernando Figueira (IMIP), Recife, Pernambuco, Brazil
- Research Department, Hospital de Câncer de Pernambuco, Recife, Brazil
- Department of Oncogenetic, Oncologia D'OR, Recife, Pernambuco, Brazil
| | - Aknar Calabrich
- Department of Oncology, Clínica AMO/DASA, Salvador, Bahia, Brazil
| | - Renato Moretti-Marques
- Department of Oncology, Albert Einstein Israelite Hospital, São Paulo, São Paulo, Brazil
| | | | | |
Collapse
|
7
|
Gerasimova Y, Ali H, Nadeem U. Challenges for pathologists in implementing clinical microbiome diagnostic testing. J Pathol Clin Res 2024; 10:e70002. [PMID: 39289163 PMCID: PMC11407905 DOI: 10.1002/2056-4538.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 08/11/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024]
Abstract
Recent research has established that the microbiome plays potential roles in the pathogenesis of numerous chronic diseases, including carcinomas. This discovery has led to significant interest in clinical microbiome testing among physicians, translational investigators, and the lay public. As novel, inexpensive methodologies to interrogate the microbiota become available, research labs and commercial vendors have offered microbial assays. However, these tests still have not infiltrated the clinical laboratory space. Here, we provide an overview of the challenges of implementing microbiome testing in clinical pathology. We discuss challenges associated with preanalytical and analytic sample handling and collection that can influence results, choosing the appropriate testing methodology for the clinical context, establishing reference ranges, interpreting the data generated by testing and its value in making patient care decisions, regulation, and cost considerations of testing. Additionally, we suggest potential solutions for these problems to expedite the establishment of microbiome testing in the clinical laboratory.
Collapse
Affiliation(s)
- Yulia Gerasimova
- Department of Infectious Diseases, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Haroon Ali
- Department of Medicine, Woodland Heights Medical Center, Lufkin, TX, USA
| | - Urooba Nadeem
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
8
|
Savitha BA, Shrivastava P, Bhagat R, Krishnamoorthy N, Shivashimpi DK, Bakre MM. Comparison of Risk Stratification by CanAssist Breast Test Performed on Core Needle Biopsies Versus Surgical Specimens in Hormone Receptor-Positive, Her2-Negative Early Breast Cancer. Cureus 2024; 16:e70054. [PMID: 39449944 PMCID: PMC11499627 DOI: 10.7759/cureus.70054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/26/2024] Open
Abstract
Introduction Core needle biopsies (CNB) are being increasingly utilized for biomarker, prognostic, and predictive testing in breast cancer (BC). CanAssist Breast (CAB) is a prognostic test performed to assess the 'risk of breast cancer recurrence' in early-stage hormone receptor-positive, Her2-negative BC patients. CAB segregates tumors as 'low risk' or 'high risk' for distant recurrence. Risk assessment done by CAB aids in planning and making adjuvant chemotherapy or hormone therapy decisions. CAB is typically performed on surgical specimens (SS). However, performing it on CNB does offer additional insights into tumor biology leading to different strategies for treatment planning; hence, we aimed to compare the risk stratification performance of CAB using CNB versus SS. Method We analyzed 103 paired formalin-fixed paraffin-embedded CNB and SS samples from hormone receptor-positive, Her2-negative early BC tissue samples submitted for performing CAB at OncoStem Diagnostics between November 2021 and September 2023. Concordance on 'risk categories' of CAB performed on CNB versus SS was reported using overall percentage agreement and Pearson correlation coefficient. Results We found excellent overall concordance of 92.2% for CAB risk stratification between paired CNB and SS tumor samples with a strong Pearson correlation coefficient of r= 0.8351 (p< 0.0001) when either SS or CNB was used as the gold standard. In prognostic testing patients with a 'low risk' of recurrence may avoid chemotherapy and hence it is crucial to assess the accuracy of CAB in the low-risk category. Additionally, in a real-world scenario, it is more likely that CAB will be performed on CNB first. Conclusion CAB when performed on CNB samples showed high concordance with SS thus demonstrating that CNB was a suitable sample for the CanAssist Breast test. The accuracy in the low-risk category is 97.5%, which ensures that physicians can reliably use prognostic information by testing CNB to guide adjuvant therapy decisions.
Collapse
Affiliation(s)
| | - Payal Shrivastava
- Technical and Analytical Division, OncoStem Diagnostics Pvt. Ltd, Bengaluru, IND
| | - Rahul Bhagat
- Technical and Analytical Division, OncoStem Diagnostics Pvt. Ltd, Bengaluru, IND
| | | | - Deepti K Shivashimpi
- Technical and Analytical Division, OncoStem Diagnostics Pvt. Ltd, Bengaluru, IND
| | - Manjiri M Bakre
- Design and Development, OncoStem Diagnostics Pvt. Ltd, Bengaluru, IND
| |
Collapse
|
9
|
Nibid L, Sabarese G, Andreotti L, Canalis B, Righi D, Longo F, Grazi M, Crucitti P, Perrone G. RNA-Seq Analysis in Non-Small Cell Lung Cancer: What Is the Best Sample from Clinical Practice? J Pers Med 2024; 14:851. [PMID: 39202042 PMCID: PMC11355753 DOI: 10.3390/jpm14080851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 07/23/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
RNA-based next-generation sequencing (RNA-seq) represents the gold standard for detecting gene fusion in non-small cell lung cancer (NSCLC). Despite this, RNA instability makes the management of tissue samples extremely complex, resulting in a significant number of test failures with missing data or the need to switch to other techniques. In the present study, we analyzed pre-analytical variables in 140 tumor tissue samples from patients affected by NSCLC to detect features that increase the chances of successful RNA-seq. We found that the success rate of the analysis positively correlates with the RNA concentration and fragmentation index. Interestingly, small biopsies were more suitable samples than surgical specimens and cell blocks. Among surgical specimens, wedge resections demonstrated better results than lobectomy. Moreover, samples stored for less than 30 days (1 month) had a better chance of success than older samples. Defining the role of pre-analytical variables in RNA-seq allows the detection of more suitable samples for analysis and more effective planning of molecular-based diagnostic approaches in NSCLC.
Collapse
Affiliation(s)
- Lorenzo Nibid
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (L.A.); (B.C.); (G.P.)
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (G.S.); (D.R.)
| | - Giovanna Sabarese
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (G.S.); (D.R.)
| | - Luca Andreotti
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (L.A.); (B.C.); (G.P.)
| | - Benedetta Canalis
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (L.A.); (B.C.); (G.P.)
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (G.S.); (D.R.)
| | - Daniela Righi
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (G.S.); (D.R.)
| | - Filippo Longo
- Research Unit of General Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (F.L.); (M.G.); (P.C.)
- Thoracic Surgery Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Margherita Grazi
- Research Unit of General Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (F.L.); (M.G.); (P.C.)
| | - Pierfilippo Crucitti
- Research Unit of General Surgery, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (F.L.); (M.G.); (P.C.)
- Thoracic Surgery Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy
| | - Giuseppe Perrone
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Via Alvaro del Portillo, 21, 00128 Roma, Italy; (L.A.); (B.C.); (G.P.)
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, 200, 00128 Roma, Italy; (G.S.); (D.R.)
| |
Collapse
|
10
|
Tsimberidou AM, Sireci A, Dumanois R, Pritchard D. Strategies to Address the Clinical Practice Gaps Affecting the Implementation of Personalized Medicine in Cancer Care. JCO Oncol Pract 2024; 20:761-766. [PMID: 38442324 PMCID: PMC11638942 DOI: 10.1200/op.23.00601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 12/15/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024] Open
Affiliation(s)
- Apostolia M Tsimberidou
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | |
Collapse
|
11
|
Plekhanov AA, Kozlov DS, Shepeleva AA, Kiseleva EB, Shimolina LE, Druzhkova IN, Plekhanova MA, Karabut MM, Gubarkova EV, Gavrina AI, Krylov DP, Sovetsky AA, Gamayunov SV, Kuznetsova DS, Zaitsev VY, Sirotkina MA, Gladkova ND. Tissue Elasticity as a Diagnostic Marker of Molecular Mutations in Morphologically Heterogeneous Colorectal Cancer. Int J Mol Sci 2024; 25:5337. [PMID: 38791375 PMCID: PMC11120711 DOI: 10.3390/ijms25105337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/25/2024] [Accepted: 05/04/2024] [Indexed: 05/26/2024] Open
Abstract
The presence of molecular mutations in colorectal cancer (CRC) is a decisive factor in selecting the most effective first-line therapy. However, molecular analysis is routinely performed only in a limited number of patients with remote metastases. We propose to use tissue stiffness as a marker of the presence of molecular mutations in CRC samples. For this purpose, we applied compression optical coherence elastography (C-OCE) to calculate stiffness values in regions corresponding to specific CRC morphological patterns (n = 54). In parallel to estimating stiffness, molecular analysis from the same zones was performed to establish their relationships. As a result, a high correlation between the presence of KRAS/NRAS/BRAF driver mutations and high stiffness values was revealed regardless of CRC morphological pattern type. Further, we proposed threshold stiffness values for label-free targeted detection of molecular alterations in CRC tissues: for KRAS, NRAS, or BRAF driver mutation-above 803 kPa (sensitivity-91%; specificity-80%; diagnostic accuracy-85%), and only for KRAS driver mutation-above 850 kPa (sensitivity-90%; specificity-88%; diagnostic accuracy-89%). To conclude, C-OCE estimation of tissue stiffness can be used as a clinical diagnostic tool for preliminary screening of genetic burden in CRC tissues.
Collapse
Affiliation(s)
- Anton A. Plekhanov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Dmitry S. Kozlov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Anastasia A. Shepeleva
- Nizhny Novgorod Regional Oncologic Hospital, 11/1 Delovaya St., 603126 Nizhny Novgorod, Russia
| | - Elena B. Kiseleva
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Liubov E. Shimolina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Irina N. Druzhkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Maria A. Plekhanova
- Nizhny Novgorod Regional Oncologic Hospital, 11/1 Delovaya St., 603126 Nizhny Novgorod, Russia
- Nizhny Novgorod City Polyclinic #1, 5 Marshala Zhukova Sq., 603107 Nizhny Novgorod, Russia
| | - Maria M. Karabut
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Ekaterina V. Gubarkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Alena I. Gavrina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Dmitry P. Krylov
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Alexander A. Sovetsky
- Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanova St., 603950 Nizhny Novgorod, Russia
| | - Sergey V. Gamayunov
- Nizhny Novgorod Regional Oncologic Hospital, 11/1 Delovaya St., 603126 Nizhny Novgorod, Russia
| | - Daria S. Kuznetsova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Vladimir Y. Zaitsev
- Institute of Applied Physics of the Russian Academy of Sciences, 46 Ulyanova St., 603950 Nizhny Novgorod, Russia
| | - Marina A. Sirotkina
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| | - Natalia D. Gladkova
- Institute of Experimental Oncology and Biomedical Technologies, Privolzhsky Research Medical University, 10/1 Minin and Pozharsky Sq., 603950 Nizhny Novgorod, Russia
| |
Collapse
|
12
|
Van Der Schueren C, Decruyenaere P, Avila Cobos F, Bult J, Deleu J, Dipalo LL, Helsmoortel HH, Hulstaert E, Morlion A, Ramos Varas E, Schoofs K, Trypsteen W, Vanden Eynde E, Van Droogenbroeck H, Verniers K, Vandesompele J, Decock A. Subpar reporting of pre-analytical variables in RNA-focused blood plasma studies. Mol Oncol 2024. [PMID: 38564603 DOI: 10.1002/1878-0261.13647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/13/2024] [Accepted: 03/22/2024] [Indexed: 04/04/2024] Open
Abstract
Extracellular RNA (cell-free RNA; exRNA) from blood-derived liquid biopsies is an appealing, minimally invasive source of disease biomarkers. As pre-analytical variables strongly influence exRNA measurements, their reporting is essential for meaningful interpretation and replication of results. The aim of this review was to chart to what extent pre-analytical variables are documented, to pinpoint shortcomings and to improve future reporting. In total, 200 blood plasma exRNA studies published in 2018 or 2023 were reviewed for annotation of 22 variables associated with blood collection, plasma preparation, and RNA purification. Our results show that pre-analytical variables are poorly documented, with only three out of 22 variables described in over half of the publications. The percentage of variables reported ranged from 4.6% to 54.6% (mean 24.84%) in 2023 and from 4.6% to 57.1% (mean 28.60%) in 2018. Recommendations and guidelines (i.e., BRISQ, ASCO-CAP, BloodPAC, PPMPT, and CEN standards) have currently not resulted in improved reporting. In conclusion, our results highlight the lack of reporting pre-analytical variables in exRNA studies and advocate for a consistent use of available standards, endorsed by funders and journals.
Collapse
Affiliation(s)
| | - Philippe Decruyenaere
- Department of Biomolecular Medicine, Ghent University, Belgium
- Department of Hematology, Ghent University Hospital, Belgium
| | - Francisco Avila Cobos
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Johanna Bult
- Department of Biomolecular Medicine, Ghent University, Belgium
- Department of Hematology, University Medical Center Groningen, The Netherlands
| | - Jill Deleu
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Laudonia Lidia Dipalo
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Hetty Hilde Helsmoortel
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Eva Hulstaert
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
- Department of Dermatology, AZ Sint-Blasius, Belgium
| | - Annelien Morlion
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Elena Ramos Varas
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Kathleen Schoofs
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
- Translational Oncogenomics and Bioinformatics Lab, Cancer Research Institute Ghent (CRIG), Belgium
- Center for Medical Biotechnology, VIB-UGent, Belgium
| | - Wim Trypsteen
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Eveline Vanden Eynde
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Hanne Van Droogenbroeck
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Kimberly Verniers
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| | - Jo Vandesompele
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
- CellCarta, Belgium
| | - Anneleen Decock
- Department of Biomolecular Medicine, Ghent University, Belgium
- OncoRNALab, Cancer Research Institute Ghent (CRIG), Belgium
| |
Collapse
|
13
|
L'Imperio V, Cazzaniga G, Mannino M, Seminati D, Mascadri F, Ceku J, Casati G, Bono F, Eloy C, Rocco EG, Frascarelli C, Fassan M, Malapelle U, Pagni F. Digital counting of tissue cells for molecular analysis: the QuANTUM pipeline. Virchows Arch 2024:10.1007/s00428-024-03794-9. [PMID: 38532196 DOI: 10.1007/s00428-024-03794-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/19/2024] [Accepted: 03/20/2024] [Indexed: 03/28/2024]
Abstract
The estimation of tumor cellular fraction (TCF) is a crucial step in predictive molecular pathology, representing an entry adequacy criterion also in the next-generation sequencing (NGS) era. However, heterogeneity of quantification practices and inter-pathologist variability hamper the robustness of its evaluation, stressing the need for more reliable results. Here, 121 routine histological samples from non-small cell lung cancer (NSCLC) cases with complete NGS profiling were used to evaluate TCF interobserver variability among three different pathologists (pTCF), developing a computational tool (cTCF) and assessing its reliability vs ground truth (GT) tumor cellularity and potential impact on the final molecular results. Inter-pathologist reproducibility was fair to good, with overall Wk ranging between 0.46 and 0.83 (avg. 0.59). The obtained cTCF was comparable to the GT (p = 0.129, 0.502, and 0.130 for surgical, biopsies, and cell block, respectively) and demonstrated good reliability if elaborated by different pathologists (Wk = 0.9). Overall cTCF was lower as compared to pTCF (30 ± 10 vs 52 ± 19, p < 0.001), with more cases < 20% (17, 14%, p = 0.690), but none containing < 100 cells for the algorithm. Similarities were noted between tumor area estimation and pTCF (36 ± 29, p < 0.001), partly explaining variability in the human assessment of tumor cellularity. Finally, the cTCF allowed a reduction of the copy number variations (CNVs) called (27 vs 29, - 6.9%) with an increase of effective CNVs detection (13 vs 7, + 85.7%), some with potential clinical impact previously undetected with pTCF. An automated computational pipeline (Qupath Analysis of Nuclei from Tumor to Uniform Molecular tests, QuANTUM) has been created and is freely available as a QuPath extension. The computational method used in this study has the potential to improve efficacy and reliability of TCF estimation in NSCLC, with demonstrated impact on the final molecular results.
Collapse
Affiliation(s)
- Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy.
| | - Giorgio Cazzaniga
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy
| | - Mauro Mannino
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy
| | - Davide Seminati
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy
| | - Francesco Mascadri
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy
| | - Joranda Ceku
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy
| | - Gabriele Casati
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy
| | - Francesca Bono
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy
| | - Catarina Eloy
- Pathology Laboratory, Institute of Molecular Pathology and Immunology of University of Porto (IPATIMUP), Porto, Portugal
- Pathology Department, Medical Faculty of University of Porto, Porto, Portugal
| | - Elena Guerini Rocco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Matteo Fassan
- Surgical Pathology and Cytopathology Unit, Department of Medicine, DIMED, University of Padua, Padua, Italy
- Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
14
|
Groiss S, Viertler C, Kap M, Bernhardt G, Mischinger HJ, Sieuwerts A, Verhoef C, Riegman P, Kruhøffer M, Svec D, Sjöback SR, Becker KF, Zatloukal K. Inter-patient heterogeneity in the hepatic ischemia-reperfusion injury transcriptome: Implications for research and diagnostics. N Biotechnol 2024; 79:20-29. [PMID: 38072306 DOI: 10.1016/j.nbt.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 12/07/2023] [Indexed: 01/16/2024]
Abstract
Cellular responses induced by surgical procedure or ischemia-reperfusion injury (IRI) may severely alter transcriptome profiles and complicate molecular diagnostics. To investigate this effect, we characterized such pre-analytical effects in 143 non-malignant liver samples obtained from 30 patients at different time points of ischemia during surgery from two individual cohorts treated either with the Pringle manoeuvre or total vascular exclusion. Transcriptomics profiles were analyzed by Affymetrix microarrays and expression of selected mRNAs was validated by RT-PCR. We found 179 mutually deregulated genes which point to elevated cytokine signaling with NFκB as a dominant pathway in ischemia responses. In contrast to ischemia, reperfusion induced pro-apoptotic and pro-inflammatory cascades involving TNF, NFκB and MAPK pathways. FOS and JUN were down-regulated in steatosis compared to their up-regulation in normal livers. Surprisingly, molecular signatures of underlying primary and secondary cancers were present in non-tumor tissue. The reported inter-patient variability might reflect differences in individual stress responses and impact of underlying disease conditions. Furthermore, we provide a set of 230 pre-analytically highly robust genes identified from histologically normal livers (<2% covariation across both cohorts) that might serve as reference genes and could be particularly suited for future diagnostic applications.
Collapse
Affiliation(s)
- Silvia Groiss
- Diagnostic & Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Christian Viertler
- Diagnostic & Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria
| | - Marcel Kap
- Pathology Department, Erasmus University Medical Center, 3015CN Rotterdam, the Netherlands
| | - Gerwin Bernhardt
- Division of General Surgery, Department of Surgery, Medical University of Graz, 8010 Graz, Austria; Department of Orthopedics and Trauma Surgery, Medical University of Graz, 8010 Graz, Austria
| | - Hans-Jörg Mischinger
- Division of General Surgery, Department of Surgery, Medical University of Graz, 8010 Graz, Austria
| | - Anieta Sieuwerts
- Department of Medical Oncology, Erasmus MC Cancer Institute and Cancer Genomics Netherlands, Erasmus University Medical Center, 3015CN Rotterdam, the Netherlands
| | - Cees Verhoef
- Department of Surgical Oncology, Erasmus MC Cancer Institute, Erasmus University Medical Center, 3015CN Rotterdam, the Netherlands
| | - Peter Riegman
- Pathology Department, Erasmus University Medical Center, 3015CN Rotterdam, the Netherlands
| | | | - David Svec
- Laboratory of Gene Expression, Institute of Biotechnology CAS, 252 50 Vestec, Czech Republic
| | | | | | - Kurt Zatloukal
- Diagnostic & Research Institute of Pathology, Medical University of Graz, 8010 Graz, Austria.
| |
Collapse
|
15
|
McConkey DJ, Baumann BC, Cooper Greenberg S, DeGraff DJ, Delacroix SE, Efstathiou JA, Foster J, Groshen S, Kadel EE, Khani F, Kim WY, Lerner SP, Levin T, Liao JC, Milowsky MI, Meeks JJ, Miyamoto DT, Mouw KW, Pietzak EJ, Solit DB, Sundi D, Tawab-Amiri A, West PJ, Wobker SE, Wyatt AW, Apolo AB, Black PC. Ensuring Successful Biomarker Studies in Bladder Preservation Clinical Trials for Non-muscle Invasive Bladder Cancer. Bladder Cancer 2024; 10:1-8. [PMID: 38993535 PMCID: PMC11181871 DOI: 10.3233/blc-230082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 01/18/2024] [Indexed: 07/13/2024]
Affiliation(s)
- David J. McConkey
- Johns Hopkins Greenberg Bladder Cancer Institute, Baltimore, MD, USA
| | - Brian C. Baumann
- Department of Radiation Oncology, Springfield Clinic, Springfield, IL, USA
- Department of Radiation Oncology, University of Pennsylvania, Philadelphia, PA, USA
| | | | - David J. DeGraff
- Department of Pathology and Laboratory Medicine, Pennsylvania State University, Hershey, PA, USA
- Department of Urology, Pennsylvania State University, Hershey, PA, USA
| | - Scott E. Delacroix
- Department of Urology, Louisiana State University Health Science Center, New Orleans, New Orleans, LA, USA
| | - Jason A. Efstathiou
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jared Foster
- Division of Cancer Treatment and Diagnosis, National Cancer Institute, NIH, Bethesda, MD, USA
| | - Susan Groshen
- Department of Population and Public Health Sciences, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Edward E. Kadel
- US Medical Affairs and Oncology Biomarker Development, Genentech Inc., South San Francisco, CA, USA
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
| | - William Y. Kim
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Seth P. Lerner
- Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Joseph C. Liao
- Department of Urology, Stanford University, Stanford, CA, USA
| | - Matthew I. Milowsky
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Joshua J. Meeks
- Department of Urology, Feinberg School of Medicine, Chicago, IL, USA
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Chicago, IL, USA
| | - David T. Miyamoto
- Department of Radiation Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, MA, USA
| | - Kent W. Mouw
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Eugene J. Pietzak
- Urology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David B. Solit
- Human Oncology and Pathogenesis Program, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Debasish Sundi
- Department of Urology, Ohio State University Comprehensive Cancer Center, Pelotonia Institute for Immuno-Oncology, Columbus, OH, USA
| | - Abdul Tawab-Amiri
- Coordinating Center for Clinical Trials, National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Sara E. Wobker
- University of North Carolina at Chapel Hill, Departments of Pathology and Laboratory Medicine and Urology, Chapel Hill, NC, USA
| | - Alexander W. Wyatt
- Vancouver Prostate Centre, Department of Urologic Sciences,University of British Columbia, Vancouver, BC, Canada
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - Andrea B. Apolo
- Genitourinary Malignancies Branch, National Cancer Institute, Center for Cancer Research, Bethesda, MD, USA
| | - Peter C. Black
- Vancouver Prostate Centre, Department of Urologic Sciences,University of British Columbia, Vancouver, BC, Canada
- University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
16
|
Watanabe K, Kohsaka S, Tatsuno K, Shinozaki-Ushiku A, Isago H, Kage H, Ushiku T, Aburatani H, Mano H, Oda K. Analysis of quality metrics in comprehensive cancer genomic profiling using a dual DNA-RNA panel. Pract Lab Med 2024; 39:e00368. [PMID: 38404525 PMCID: PMC10883814 DOI: 10.1016/j.plabm.2024.e00368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 02/13/2024] [Indexed: 02/27/2024] Open
Abstract
Background The nucleic acid quality from formalin-fixed paraffin-embedded (FFPE) tumor vary among samples, resulting in substantial variability in the quality of comprehensive cancer genomic profiling tests. The objective of the study is to investigate how nucleic acid quality affects sequencing quality. We also examined the variations in nucleic acid quality among different hospitals or cancer types. Methods Three nucleic acid quality metrics (ddCq, Q-value, and DV200) and five sequencing quality metrics (on-target rate, mean depth, coverage uniformity, target exon coverage, and coverage of the housekeeping gene) were examined using 585 samples from the Todai OncoPanel, a dual DNA-RNA panel. Results In the DNA panel, ddCq served as an indicator of sequencing depth and Q-value reflected the uniformity of sequencing across different regions. It was essential to have favorable values not only for ddCq but also for Q-value to obtain ideal sequencing results. For the RNA panel, DV200 proved to be a valuable metric for assessing the coverage of the housekeeping genes. Significant inter-hospital differences were observed for DNA quality (ddCq and Q-value), but not for RNA quality (DV200). Differences were also observed among cancer types, with Q-value being the lowest in lung and the highest in cervix, while DV200 was the highest in lung and the lowest in bowel. Conclusions We demonstrated distinct characteristics and high predictive performances of ddCq, Q-value, and DV200. Variations were observed in the nucleic acid quality across hospitals and cancer types. Further study is warranted on preanalytical factors in comprehensive cancer genomic profiling tests.
Collapse
Affiliation(s)
- Kousuke Watanabe
- Department of Clinical Laboratory, The University Tokyo, Tokyo, Japan
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Kenji Tatsuno
- Genome Science and Medicine Laboratory, RCAST, The University of Tokyo, Tokyo, Japan
| | - Aya Shinozaki-Ushiku
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hideaki Isago
- Department of Clinical Laboratory, The University Tokyo, Tokyo, Japan
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
| | - Hidenori Kage
- Department of Respiratory Medicine, The University of Tokyo, Tokyo, Japan
- Next-Generation Precision Medicine Development Laboratory, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Tetsuo Ushiku
- Department of Pathology, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Aburatani
- Genome Science and Medicine Laboratory, RCAST, The University of Tokyo, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Katsutoshi Oda
- Division of Integrative Genomics, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
17
|
Vyas M, Karamchandani DM. Essentials of macroscopic evaluation of specimens from gastrointestinal tract. J Clin Pathol 2024; 77:169-176. [PMID: 38373784 DOI: 10.1136/jcp-2023-208981] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Accepted: 07/20/2023] [Indexed: 02/21/2024]
Abstract
An astute macroscopic examination, coupled with correlating the gross findings with clinical indication and operative notes along with judicious, yet all pertinent sectioning for pathological examination is crucial for an accurate histopathological diagnosis, eventually leading to optimal patient care. This succinct review highlights the general concepts that lay the foundation of evaluating and grossing specimens from the luminal gastrointestinal (GI) tract. We also discuss the gross evaluation and sectioning of small therapeutic resections, along with a systematic approach and rationale when grossing and submitting histological sections from larger oncological resections from the luminal GI tract. Selected site-specific considerations, for example, grossing treated rectal and oesophageal cancers or taking sections from mucinous tumours of the appendix, among others, are also discussed.
Collapse
Affiliation(s)
- Monika Vyas
- Department of Pathology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Dipti M Karamchandani
- Department of Pathology, Division of Anatomic Pathology, UT Southwestern Medical School, Dallas, Texas, USA
| |
Collapse
|
18
|
Mohamed WT, Jahagirdar V, Jaber F, Ahmed MK, Fatima I, Bierman T, Fu Z, Jones PG, Hassan AF, Faber E, Clarkston WK, Ghoz H, Tawfik OW, Jonnalagadda S. Endoscopic Ultrasound-Guided Fine-Needle Biopsy Versus Aspiration for Tissue Sampling Adequacy for Molecular Testing in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2024; 16:761. [PMID: 38398152 PMCID: PMC10886941 DOI: 10.3390/cancers16040761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 02/06/2024] [Accepted: 02/10/2024] [Indexed: 02/25/2024] Open
Abstract
BACKGROUND AND AIMS There is limited literature on sample adequacy for molecular testing in pancreatic ductal adenocarcinoma obtained via endoscopic ultrasound (EUS) fine-needle aspiration (FNA) versus EUS fine-needle biopsy (FNB). We aimed to compare these two modalities regarding sample adequacy for molecular and genomic sequencing. METHODS We reviewed all patients with pancreatic ductal adenocarcinoma who underwent EUS at Saint Luke's Hospital from 2018 to 2021. The patients were categorized based on the method of EUS tissue acquisition, specifically FNA or FNB. A comprehensive evaluation was conducted for all cases by cytotechnologists. RESULTS Out of 132 patients who underwent EUS-guided biopsies, 76 opted for FNA, 48 opted for FNB, and 8 opted for a combination of both. The average number of passes required for FNB and FNA was 2.58 ± 1.06 and 2.49 ± 1.07, respectively (p = 0.704), indicating no significant difference. Interestingly, 71.4% (35) of FNB-obtained samples were deemed adequate for molecular testing, surpassing the 32.1% (26) adequacy observed with FNA (p < 0.001). Additionally, 46.4% (26) of FNB-obtained samples were considered adequate for genomic testing, a notable improvement over the 23.8% (20) adequacy observed with FNA (p = 0.005). CONCLUSION Although the number of passes required for cytologic diagnosis did not differ significantly between EUS-FNB and EUS-FNA, the former demonstrated superiority in obtaining samples adequate for molecular testing. Tumor surface area and cellularity were crucial parameters in determining sample adequacy for molecular testing, irrespective of the chosen tissue acquisition modality.
Collapse
Affiliation(s)
- Wael T. Mohamed
- Department of Transplant Hepatology, Cleveland Clinic, Cleveland, OH 44114, USA
| | - Vinay Jahagirdar
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (V.J.); (I.F.)
| | - Fouad Jaber
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (V.J.); (I.F.)
| | - Mohamed K. Ahmed
- Department of Gastroenterology, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (M.K.A.); (W.K.C.); (H.G.)
| | - Ifrah Fatima
- Department of Internal Medicine, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (V.J.); (I.F.)
| | - Thomas Bierman
- Department of Gastroenterology, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (M.K.A.); (W.K.C.); (H.G.)
| | - Zhuxuan Fu
- Department of Cardiovascular Research, Saint Luke’s Health System, Kansas City, MO 64108, USA; (Z.F.); (P.G.J.)
| | - Philip G. Jones
- Department of Cardiovascular Research, Saint Luke’s Health System, Kansas City, MO 64108, USA; (Z.F.); (P.G.J.)
| | - Amira F. Hassan
- Department of Pathology, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (A.F.H.); (E.F.)
| | - Erin Faber
- Department of Pathology, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (A.F.H.); (E.F.)
- MAWD Pathology Group, Lenexa, KS 66215, USA;
| | - Wendell K. Clarkston
- Department of Gastroenterology, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (M.K.A.); (W.K.C.); (H.G.)
- Department of Gastroenterology, Saint Luke’s Health System of Kansas City, Kansas City, MO 64108, USA;
| | - Hassan Ghoz
- Department of Gastroenterology, University of Missouri-Kansas City, Kansas City, MO 64108, USA; (M.K.A.); (W.K.C.); (H.G.)
| | - Ossama W. Tawfik
- MAWD Pathology Group, Lenexa, KS 66215, USA;
- Department of Pathology, Saint Luke’s Health System of Kansas City, Kansas City, MO 64108, USA
| | - Sreeni Jonnalagadda
- Department of Gastroenterology, Saint Luke’s Health System of Kansas City, Kansas City, MO 64108, USA;
| |
Collapse
|
19
|
Charania AA, Pokal AG, Zuaiter DR, Islam M, Kim A, Budhwani KI. Preanalytical protocol for fresh solid tumor biospecimens. RESEARCH SQUARE 2024:rs.3.rs-3911742. [PMID: 38352336 PMCID: PMC10862947 DOI: 10.21203/rs.3.rs-3911742/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/23/2024]
Abstract
Nearly seventy percent of diagnostic lab test errors occur due to variability in preanalytical factors. 1-4 These are the parameters involved with all aspects of tissue processing, starting from the time tissue is collected from the patient in the operating room, until it is received and tested in the laboratory. While there are several protocols for transporting fixed tissue, organs, and liquid biopsies, such protocols are lacking for transport and handling of live solid tumor tissue specimens. There is a critical need to establish preanalytical protocols to reduce variability in biospecimen integrity and improve diagnostics for personalized medicine. 2,5 Here, we provide a comprehensive protocol for the standard collection, handling, packaging, cold-chain logistics, and receipt of solid tumor tissue biospecimens to preserve tissue viability.
Collapse
|
20
|
Kerr KM, Bubendorf L, Lopez-Rios F, Khalil F, Roy-Chowdhuri S, Joubert P, Hartmann A, Guerini-Rocco E, Yatabe Y, Hofman P, Cooper WA, Dacic S. Optimizing tissue stewardship in non-small cell lung cancer to support molecular characterization and treatment selection: statement from a working group of thoracic pathologists. Histopathology 2024; 84:429-439. [PMID: 37957137 DOI: 10.1111/his.15078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 11/15/2023]
Abstract
Many patients with non-small cell lung cancer do not receive guideline-recommended, biomarker-directed therapy, despite the potential for improved clinical outcomes. Access to timely, accurate, and comprehensive molecular profiling, including targetable protein overexpression, is essential to allow fully informed treatment decisions to be taken. In turn, this requires optimal tissue management to protect and maximize the use of this precious finite resource. Here, a group of leading thoracic pathologists recommend factors to consider for optimal tissue management. Starting from when lung cancer is first suspected, keeping predictive biomarker testing in the front of the mind should drive the development of practices and procedures that conserve tissue appropriately to support molecular characterization and treatment selection.
Collapse
Affiliation(s)
- Keith M Kerr
- Department of Pathology, Aberdeen Royal Infirmary and Aberdeen University Medical School, Aberdeen, UK
| | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, Basel, Switzerland
| | - Fernando Lopez-Rios
- Department of Pathology, 12 de Octubre University Hospital-CIBERONC, Research Institute 12 de Octubre University Hospital (i+12), Universidad Complutense, Madrid, Spain
| | | | | | - Philippe Joubert
- Québec Heart and Lung Institute-Laval University (IUCPQ-UL), Quebec, QC, Canada
| | - Arndt Hartmann
- Institute of Pathology, University Erlangen-Nürnberg, Erlangen, Germany
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Paul Hofman
- Nice University Hospital, FHU OncoAge, BB-0033-00025, University Côte d'Azur, Nice, France
| | - Wendy A Cooper
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia
- The University of Sydney, Sydney, NSW, Australia
- Western Sydney University, Campbelltown, NSW, Australia
| | | |
Collapse
|
21
|
Noda Y, Sakagami T, Suzuki K, Fujisawa T, Yagi M, Iwai H, Tsuta K. Direct specimen collection during routine operation improves nucleic acid quality in genetic analysis samples for head and neck tumors: A retrospective study. Pathol Res Pract 2023; 252:154943. [PMID: 37977036 DOI: 10.1016/j.prp.2023.154943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/07/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
The nucleic acid integrity of head and neck squamous cell carcinoma (HNSCC) samples is poor, and the material available for genetic analysis is limited. Therefore, to expand the effectiveness of personalized medicine in patients with HNSCC, a new sampling method is needed. In total, 128 samples from 44 patients with HNSCC were studied: 32 genetic analysis samples (GASs) collected as 5 × 5 × 5 mm tissue fragments from resected large tumors and immediately embedded in a small formalin bottle within 10 min (i.e., the ischemic time), 43 primary tumor components (primary), 14 decalcified tumor (DC) samples, 32 metastatic tumors in lymph nodes (LNs), and 7 parakeratinized components (PKCs). The nucleic acid quality in the GAS, primary, DC, LN, and PKC groups was compared and next-generation sequencing (NGS) was performed. DNA integrity number and percentage of RNA fragments with > 200 nucleotides were significantly higher in the GAS group than those in the other groups. RNA integrity number decreased first in LN, followed by GAS, primary, and DC. No significant differences were observed in DIN, RIN and DV200 among the PKC, primary and LN. Following methyl green-pyronin staining, preserved DNA and RNA were not visualized in DC samples. Most NGS metrics did not differ significantly among primary, LN, and PKC samples. In conclusion, GASs should be collected during routine hospital activities. When the volume of viable materials is limited, PKCs should be considered for genetic analysis.
Collapse
Affiliation(s)
- Yuri Noda
- Department of Pathology and Laboratory Medicine, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan; Department of Pathology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan.
| | - Tomofumi Sakagami
- Department of Otolaryngology, Head and Neck Surgery, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Kensuke Suzuki
- Department of Otolaryngology, Head and Neck Surgery, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Takuo Fujisawa
- Department of Otolaryngology, Head and Neck Surgery, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Masao Yagi
- Department of Otolaryngology, Head and Neck Surgery, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Hiroshi Iwai
- Department of Otolaryngology, Head and Neck Surgery, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Koji Tsuta
- Department of Pathology and Laboratory Medicine, Kansai Medical University Hospital, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan; Department of Pathology, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
22
|
Mandarano M, Pelliccia C, Tomasello L, Caselli E, Floridi C, Loreti E, Barberini F, Rulli A, Gili A, Potenza R, Puma F, Rosati E, Donini A, Petrina A, Baccari P, Del Sordo R, Colella R, Bellezza G, Sidoni A. A New Medium (HistoCold) for Surgical Specimens Preserving to Improve the Preanalytic Issues in Histopathological Samples Handling: Morphologic and Antigenic Analysis. Biopreserv Biobank 2023; 21:610-623. [PMID: 37192479 DOI: 10.1089/bio.2022.0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Introduction: The onset of precision medicine has led to the integration of traditional morphologic tissues evaluation with biochemical and molecular data for a more appropriate pathological diagnosis. The preanalytic phase and, particularly, timing of cold ischemia are crucial to guarantee high-quality biorepositories of formalin-fixed paraffin-embedded (FFPE) tissues for patients' needs and scientific research. However, delayed fixation using the gold-standard and carcinogenic fixative neutral-buffered formalin (NBF) can be a significant limitation to diagnosis and biopathological characterization. HistoCold (patented; Bio-Optica Milano S.p.A., Milano, Italy) is a nontoxic, stable, and refrigerated preservative solution for tissue handling. This study examined HistoCold's potential role in improving the preanalytic phase of the pathological diagnostic process. Materials and Methods: Breast, lung, or colorectal cancers (20, 25, and 10 cases, respectively) that were to be surgically resected were recruited between 2019 and 2021. Once specimens were surgically removed, three residual samples for each patient were first promptly immersed into HistoCold for 24, 48, and 72 hours and then FFPE. These were compared with routine specimens regarding morphologic features (hematoxylin and eosin) and tissue antigenicity (immunohistochemical stains). Results: Good concordance regarding both the morphologic characteristics of the neoplasms and their proteins expression between the routine and HistoCold handled tissues were found. The tissue handling with the solution never affected the histopathological diagnosis. Conclusions: The use of HistoCold for samples transporting is easy, allows for improving the management of cold ischemia time, and monitoring the fixation times in NBF, resulting in good quality tissue blocks for biobanking. Moreover, it could be a candidate to eliminate formalin from operating theaters. HistoCold looks very promising for the preanalytic phase of human tissues handling in the era of precision medicine, to provide the best service to patients, and to scientific research.
Collapse
Affiliation(s)
- Martina Mandarano
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Cristina Pelliccia
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Laura Tomasello
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emanuele Caselli
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Claudia Floridi
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Elisabetta Loreti
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Barberini
- Breast Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Antonio Rulli
- Breast Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Alessio Gili
- Section of Public Health, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Rossella Potenza
- Thoracic Surgery Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Francesco Puma
- Thoracic Surgery Unit, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Emanuele Rosati
- Section of General and Emergency Surgery, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Annibale Donini
- Section of General and Emergency Surgery, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Adolfo Petrina
- General Surgery Unit, S.M. Misericordia Hospital, Perugia, Italy
| | - Paolo Baccari
- General Surgery Unit, S.M. Misericordia Hospital, Perugia, Italy
| | - Rachele Del Sordo
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Renato Colella
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Guido Bellezza
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Angelo Sidoni
- Section of Anatomic Pathology and Histology, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| |
Collapse
|
23
|
Dorca E, Velasco A, Varela M, Gatius S, Villatoro S, Fullana N, Cuevas D, Vaquero M, Birnbaum A, Neumann K, Matias-Guiu X. Validation of Modaplex POLE mutation assay in endometrial carcinoma. Virchows Arch 2023; 483:787-794. [PMID: 37874375 DOI: 10.1007/s00428-023-03636-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 08/11/2023] [Accepted: 08/24/2023] [Indexed: 10/25/2023]
Abstract
The TCGA-based molecular classification of endometrial cancer has emerged as an important tool to stratify patients according to prognosis. A simplified scheme has been proposed, by using immunohistochemistry for p53, MSH6, and PMS2 and a molecular test for POLE mutations (NGS or Sanger sequencing, techniques that are not available in many centers worldwide). In this study, we validate a novel method that allows simultaneous analysis of multiple pathogenic POLE mutations. The Modaplex technology integrates polymerase chain reaction and capillary electrophoresis. The design of this study encompassed 4 different steps: (1) a retrospective-pilot phase, with 80 tumors, balancing the four molecular subgroups. (2) A retrospective phase of 25 tumors obtained between 2016 and 2020, and 30 tumors obtained between 2000 and 2015. (3) An inter-laboratory corssavalidation step with 19 cases (belonging to phases 1 and 2). (4) A prospective cohort of 123 tumors, of unknown POLE status, with simultaneous validation by Sanger sequencing. A total of 258 samples were analyzed. In the first and second phases, the test showed positive/negative predictive values of 100%, by correctly identifying POLE mutation status in 79/79 and 55/55 cases. Phase 3 showed 100% of inter-laboratory consistency. Phase 4 showed 16 positive samples out of the 123 prospective cases. Overall, the test has revealed sensitivity and specificity of 100%, identifying a total of 47 POLE-mutated tumors. We have shown that this technique allows faster and easier identification of multiple pathogenic POLE mutations with high robustness and confidence when comparing to other tests such as Sanger sequencing.
Collapse
Affiliation(s)
- Eduard Dorca
- Pathology Department, Hospital Universitari de Bellvitge, Universitat de Barcelona, IDIBELL, Feixa Llarga SN, 08907, L'Hospitalet de Llobregat, Spain
| | - Ana Velasco
- Pathology Department, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, CIBERONC, Av Rovira Roure 80, 25198, Lleida, Spain
| | - Mar Varela
- Pathology Department, Hospital Universitari de Bellvitge, Universitat de Barcelona, IDIBELL, Feixa Llarga SN, 08907, L'Hospitalet de Llobregat, Spain
| | - Sonia Gatius
- Pathology Department, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, CIBERONC, Av Rovira Roure 80, 25198, Lleida, Spain
| | - Sergio Villatoro
- Pathology Department, Hospital Universitari de Bellvitge, Universitat de Barcelona, IDIBELL, Feixa Llarga SN, 08907, L'Hospitalet de Llobregat, Spain
| | - Neus Fullana
- Pathology Department, Hospital Universitari de Bellvitge, Universitat de Barcelona, IDIBELL, Feixa Llarga SN, 08907, L'Hospitalet de Llobregat, Spain
| | - Dolors Cuevas
- Pathology Department, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, CIBERONC, Av Rovira Roure 80, 25198, Lleida, Spain
| | - Marta Vaquero
- Pathology Department, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, CIBERONC, Av Rovira Roure 80, 25198, Lleida, Spain
| | - Astrid Birnbaum
- Städtisches Klinikum Dessau Institut für Pathologie, Dessau-Roßlau, Germany
| | - Karsten Neumann
- Städtisches Klinikum Dessau Institut für Pathologie, Dessau-Roßlau, Germany
| | - Xavier Matias-Guiu
- Pathology Department, Hospital Universitari de Bellvitge, Universitat de Barcelona, IDIBELL, Feixa Llarga SN, 08907, L'Hospitalet de Llobregat, Spain.
- Pathology Department, Hospital Universitari Arnau de Vilanova, Universitat de Lleida, IRBLleida, CIBERONC, Av Rovira Roure 80, 25198, Lleida, Spain.
| |
Collapse
|
24
|
Grillo F, Ali M, Paudice M, Pigozzi S, Anselmi G, Scabini S, Sciallero S, Piol N, Mastracci L. Impact of formalin fixation on mismatch repair protein evaluation by immunohistochemistry. Virchows Arch 2023; 483:677-685. [PMID: 37773452 PMCID: PMC10673985 DOI: 10.1007/s00428-023-03661-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 08/29/2023] [Accepted: 09/15/2023] [Indexed: 10/01/2023]
Abstract
Mismatch repair/microsatellite instability (MMR/MSI) status in colorectal cancer (CRC) has become fundamental as a diagnostic, prognostic, and predictive factor. MMR immunohistochemistry (IHC) is considered a simple and reliable approach; however, its effectiveness depends on pre-analytic factors. Aim of this study was to investigate the impact of different fixation times/protocols on MMR protein IHC quality. Left over tissue from surgically resected CRC samples (cold ischemia time < 30 min) where fixed as follows: standard formalin fixation (24-48 h); hypo-fixation (<20 h); hyper-fixation (>90 h); cold (4°C) fixation (24-48 h); standard fixation for small sample size (0.5×0.5 cm). Samples for each group were collected from 30 resected CRC and the following parameters were evaluated on 600 immunohistochemical stains: intensity of expression; patchiness of staining; presence of central artefact. Forty-six immunoreactions were inadequate (score 0 intensity), the majority regarding MLH1 or PMS2 in the hypo-fixation group (47.8%), followed by the hyper-fixation group (28.1%); cold formalin fixation showed the least inadequate cases. Patchiness and central artefact were more frequent in hypo-fixation and standard fixation group compared to the others. MLH1 (closely followed by PMS2) performed worse with regard to immunostaining intensity (p=0.0002) in the standard and in the hypo-fixation group (p< 0.00001). Using a small sample size improved patchiness/central artefacts. This is the first study specifically created to evaluate the impact of fixation on MMR protein IHC, showing that both formalin hypo- and hyper-fixation can cause problems; 24-h formalin fixation as well as cold (4°C) formalin fixation are recommended for successful IHC MMR evaluation.
Collapse
Affiliation(s)
- Federica Grillo
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy.
| | - Murad Ali
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Michele Paudice
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Simona Pigozzi
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Giorgia Anselmi
- Molecular Diagnostic Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefano Scabini
- Oncological Surgical Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Stefania Sciallero
- Medical Oncology Unit 1, IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Nataniele Piol
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Luca Mastracci
- Pathology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Pathology Unit, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genoa and IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| |
Collapse
|
25
|
Monaghan JF, Cullen D, Wynne C, Lyng FM, Meade AD. Effect of pre-analytical variables on Raman and FTIR spectral content of lymphocytes. Analyst 2023; 148:5422-5434. [PMID: 37750362 DOI: 10.1039/d3an00686g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The use of Fourier transform infrared (FTIR) and Raman spectroscopy (RS) for the analysis of lymphocytes in clinical applications is increasing in the field of biomedicine. The pre-analytical phase, which is the most vulnerable stage of the testing process, is where most errors and sample variance occur; however, it is unclear how pre-analytical variables affect the FTIR and Raman spectra of lymphocytes. In this study, we evaluated how pre-analytical procedures undertaken before spectroscopic analysis influence the spectral integrity of lymphocytes purified from the peripheral blood of male volunteers (n = 3). Pre-analytical variables investigated were associated with (i) sample preparation, (blood collection systems, anticoagulant, needle gauges), (ii) sample storage (fresh or frozen), and (iii) sample processing (inter-operator variability, time to lymphocyte isolation). Although many of these procedural pre-analytical variables did not alter the spectral signature of the lymphocytes, evidence of spectral effects due to the freeze-thaw cycle, in vitro culture inter-operator variability and the time to lymphocyte isolation was observed. Although FTIR and RS possess clinical potential, their translation into a clinical environment is impeded by a lack of standardisation and harmonisation of protocols related to the preparation, storage, and processing of samples, which hinders uniform, accurate, and reproducible analysis. Therefore, further development of protocols is required to successfully integrate these techniques into current clinical workflows.
Collapse
Affiliation(s)
- Jade F Monaghan
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Central Quad, City Campus, Grangegorman, D07 XT95, Ireland.
- Radiation and Environmental Science Centre, Focas Research Institute, Technological University Dublin, Aungier Street, D02 HW71, Ireland
| | - Daniel Cullen
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Central Quad, City Campus, Grangegorman, D07 XT95, Ireland.
- Radiation and Environmental Science Centre, Focas Research Institute, Technological University Dublin, Aungier Street, D02 HW71, Ireland
| | - Claire Wynne
- School of Biological, Health and Sports Sciences, Technological University Dublin, Central Quad, City Campus, Grangegorman, D07 XT95, Ireland
| | - Fiona M Lyng
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Central Quad, City Campus, Grangegorman, D07 XT95, Ireland.
- Radiation and Environmental Science Centre, Focas Research Institute, Technological University Dublin, Aungier Street, D02 HW71, Ireland
| | - Aidan D Meade
- School of Physics, Clinical and Optometric Sciences, Technological University Dublin, Central Quad, City Campus, Grangegorman, D07 XT95, Ireland.
- Radiation and Environmental Science Centre, Focas Research Institute, Technological University Dublin, Aungier Street, D02 HW71, Ireland
| |
Collapse
|
26
|
Amaral Duarte F, Aguiar Junior PN, Dienstmann R, Ferreira CG. Precision medicine in Thoracic Oncology: understanding disparities to tackle inequities in access. Expert Rev Pharmacoecon Outcomes Res 2023; 23:981-987. [PMID: 37750550 DOI: 10.1080/14737167.2023.2260563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/12/2023] [Indexed: 09/27/2023]
Abstract
INTRODUCTION Precision medicine is defined as personalized interventions fitted to patients' or tumors' characteristics. Patients diagnosed with different neoplasms have benefited from a personalized therapeutic approach in terms of response and survival. However, several challenges must be addressed for precision oncology to become a global reality. Access to genomic testing that allows biomarker identification is a main issue. AREAS COVERED A nonsystematic literature review about inequities in access to molecular genetic testing, focusing on lung cancer as the prominent example, was performed by a group of expert clinical oncologists. EXPERT OPINION Access to molecular tests and their matched treatments differ between regions of the world and even among diverse populations in the same country. Socioeconomic characteristics are often strongly correlated with this disparity. Furthermore, although the cost is a determinant factor for inequality, other issues have been recognized. Advances in the education of healthcare professionals, patient advocacy initiatives, building local laboratory workstreams, and promoting favorable regulatory environment are vital factors in promoting equal access.
Collapse
Affiliation(s)
| | - Pedro Nazareth Aguiar Junior
- Department of Clinical Oncology, Grupo Oncoclínicas, São Paulo, SP, Brazil
- Department of Clinical Oncology, Faculdade de Medicina do ABC, Santo André, SP, Brazil
| | - Rodrigo Dienstmann
- Department of Clinical Oncology, Grupo Oncoclínicas, São Paulo, SP, Brazil
| | | |
Collapse
|
27
|
Maloberti T, De Leo A, Coluccelli S, Sanza V, Gruppioni E, Altimari A, Zagnoni S, Giunchi F, Vasuri F, Fiorentino M, Mollica V, Ferrari S, Miccoli S, Visani M, Turchetti D, Massari F, Tallini G, de Biase D. Multi-Gene Next-Generation Sequencing Panel for Analysis of BRCA1/ BRCA2 and Homologous Recombination Repair Genes Alterations Metastatic Castration-Resistant Prostate Cancer. Int J Mol Sci 2023; 24:ijms24108940. [PMID: 37240284 DOI: 10.3390/ijms24108940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Revised: 05/11/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023] Open
Abstract
Despite significant therapeutic advances, metastatic CRPC (mCRPC) remains a lethal disease. Mutations in homologous recombination repair (HRR) genes are frequent in mCRPC, and tumors harboring these mutations are known to be sensitive to PARP inhibitors. The aim of this study was to verify the technical effectiveness of this panel in the analysis of mCRPC, the frequency and type of mutations in the BRCA1/BRCA2 genes, as well as in the homologous recombination repair (HRR) genes. A total of 50 mCRPC cases were analyzed using a multi-gene next-generation sequencing panel evaluating a total of 1360 amplicons in 24 HRR genes. Of the 50 cases, 23 specimens (46.0%) had an mCRPC harboring a pathogenic variant or a variant of uncertain significance (VUS), whereas in 27 mCRPCs (54.0%), no mutations were detected (wild-type tumors). BRCA2 was the most commonly mutated gene (14.0% of samples), followed by ATM (12.0%), and BRCA1 (6.0%). In conclusion, we have set up an NGS multi-gene panel that is capable of analyzing BRCA1/BRCA2 and HRR alterations in mCRPC. Moreover, our clinical algorithm is currently being used in clinical practice for the management of patients with mCRPC.
Collapse
Affiliation(s)
- Thais Maloberti
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Antonio De Leo
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Sara Coluccelli
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Viviana Sanza
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Elisa Gruppioni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Annalisa Altimari
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Stefano Zagnoni
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesca Giunchi
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Vasuri
- Pathology Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Michelangelo Fiorentino
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- Pathology Unit, Maggiore Hospital, AUSL Bologna, 40133 Bologna, Italy
| | - Veronica Mollica
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Simona Ferrari
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Sara Miccoli
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Michela Visani
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| | - Daniela Turchetti
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- Unit of Medical Genetics, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Francesco Massari
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
| | - Giovanni Tallini
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40138 Bologna, Italy
| | - Dario de Biase
- Solid Tumor Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Pharmacy and Biotechnology, University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
28
|
Kanesvaran R, Chia PL, Chiong E, Chua MLK, Ngo NT, Ow S, Sim HG, Tan MH, Tay KH, Wong ASC, Wong SW, Tan PH. An approach to genetic testing in patients with metastatic castration-resistant prostate cancer in Singapore. ANNALS OF THE ACADEMY OF MEDICINE, SINGAPORE 2023; 52:135-148. [PMID: 38904491 DOI: 10.47102/annals-acadmedsg.2022372] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/22/2024]
Abstract
Introduction There has been a rapid evolution in the treatment strategies for metastatic castration-resistant prostate cancer (mCRPC) following the identification of targetable mutations, making genetic testing essential for patient selection. Although several international guidelines recommend genetic testing for patients with mCRPC, there is a lack of locally endorsed clinical practice guidelines in Singapore. Method A multidisciplinary specialist panel with representation from medical and radiation oncology, urology, pathology, interventional radiology, and medical genetics discussed the challenges associated with patient selection, genetic counselling and sample processing in mCRPC. Results A clinical model for incorporating genetic testing into routine clinical practice in Singapore was formulated. Tumour testing with an assay that is able to detect both somatic and germline mutations should be utilised. The panel also recommended the "mainstreaming" approach for genetic counselling in which pre-test counselling is conducted by the managing clinician and post-test discussion with a genetic counsellor, to alleviate the bottlenecks at genetic counselling stage in Singapore. The need for training of clinicians to provide pre-test genetic counselling and educating the laboratory personnel for appropriate sample processing that facilitates downstream genetic testing was recognised. Molecular tumour boards and multidisciplinary discussions are recommended to guide therapeutic decisions in mCRPC. The panel also highlighted the issue of reimbursement for genetic testing to reduce patient-borne costs and increase the reach of genetic testing among this patient population. Conclusion This article aims to provide strategic and implementable recommendations to overcome the challenges in genetic testing for patients with mCRPC in Singapore.
Collapse
Affiliation(s)
| | - Puey Ling Chia
- Department of Medical Oncology, Tan Tock Seng Hospital, Singapore
| | - Edmund Chiong
- Department of Urology, National University Hospital, Singapore
- Department of Surgery, National University of Singapore, Singapore
| | | | - Nye Thane Ngo
- Division of Pathology, Singapore General Hospital, Singapore
| | - Samuel Ow
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Hong Gee Sim
- Ravenna Urology Clinic, Gleneagles Medical Centre, Singapore
| | | | - Kiang Hiong Tay
- Department of Vascular and Interventional Radiation, Singapore General Hospital, Singapore
| | | | | | - Puay Hoon Tan
- Division of Pathology, Singapore General Hospital, Singapore
| |
Collapse
|
29
|
Dameri M, Cirmena G, Ravera F, Ferrando L, Cuccarolo P, Stabile M, Fanelli GN, Nuzzo PV, Calabrese M, Tagliafico A, Ballestrero A, Zoppoli G. Standard Operating Procedures (SOPs) for non-invasive multiple biomarkers detection in an academic setting: a critical review of the literature for the RENOVATE study protocol. Crit Rev Oncol Hematol 2023; 185:103963. [PMID: 36931614 DOI: 10.1016/j.critrevonc.2023.103963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 02/06/2023] [Accepted: 02/13/2023] [Indexed: 03/17/2023] Open
Abstract
Liquid biopsy has the potential to drastically change clinical practice, paving the way to a novel non-invasive approach for cancer diagnosis and treatment. One of the limitations for the implementation of liquid biopsy in clinical practice is the lack of shared and reproducible standard operating procedures (SOPs) for sample collection, processing and storage. Here, we present a critical review of the literature focusing on the available SOPs to guide liquid biopsy management in research settings and describe SOPs that our laboratory developed and employed in the context of a prospective clinical-translational trial (RENOVATE, NCT04781062). The main aim of this manuscript is to address common issues, towards the implementation of interlaboratory shared protocols for optimized preanalytical handling of blood and urine samples. To our knowledge, this work is one of the few up-to-date, freely available comprehensive reports on trial-level procedures for the handling of liquid biopsy.
Collapse
Affiliation(s)
- Martina Dameri
- Department of Internal Medicine and Medical Specialties DiMI, University of Genoa, 16132, Genoa, Italy
| | | | - Francesco Ravera
- Department of Internal Medicine and Medical Specialties DiMI, University of Genoa, 16132, Genoa, Italy; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 10044, New York, NY, USA
| | | | - Paola Cuccarolo
- Department of Internal Medicine and Medical Specialties DiMI, University of Genoa, 16132, Genoa, Italy
| | - Mario Stabile
- Department of Internal Medicine and Medical Specialties DiMI, University of Genoa, 16132, Genoa, Italy
| | - Giuseppe Nicolò Fanelli
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, 10021, New York, NY, USA; First Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Pier Vitale Nuzzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, 10044, New York, NY, USA
| | | | - Alberto Tagliafico
- IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy; Department of Health Sciences DISSAL, University of Genoa, 16132, Genoa, Italy
| | - Alberto Ballestrero
- Department of Internal Medicine and Medical Specialties DiMI, University of Genoa, 16132, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy
| | - Gabriele Zoppoli
- Department of Internal Medicine and Medical Specialties DiMI, University of Genoa, 16132, Genoa, Italy; IRCCS Ospedale Policlinico San Martino, 16132, Genoa, Italy.
| |
Collapse
|
30
|
Naso J, Lo YC, Sholl LM. Updates in pathology and molecular diagnostics to inform the evolving landscape of thoracic surgery and oncology. J Surg Oncol 2023; 127:244-257. [PMID: 36630101 DOI: 10.1002/jso.27184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 01/12/2023]
Abstract
The pathologic assessment of lung cancers provides essential guidance to the surgeon and oncologist who are considering the best treatment strategies for patients with both early and advanced-stage disease. The management of patients with lung cancer is predicated first and foremost on access to an accurate diagnosis, even when the sample size is limited, as is often the case with use of modern, minimally invasive sampling techniques. Once the diagnosis and disease stage are established, predictive biomarker testing may be essential, particularly for those patients with nonsmall cell lung carcinoma (NSCLC) being considered for immunotherapy or genomic biomarker-driven targeted therapy. This review will discuss the best practices for the diagnosis of NSCLC using morphology and immunohistochemistry, thus providing the surgeon with needed information to understand and critically evaluate pathology reports. Controversial and evolving topics including tumor spread through airspaces, evaluation of multiple tumors, and staging based on invasive tumor size will be addressed. Clinical genomic profiling in NSCLC is driven by published guidelines and reflects evidence based on clinical trials and regulatory approvals. In this fast-moving space, surgeons should be aware of the critical immunohistochemical and genomic biomarkers that drive systemic therapy decisions and anticipate when such testing will be required, both to ensure adequate sampling and to advise the pathologist when tumor material will be required for biomarker analysis. The basic approaches to and sample requirements for molecular biomarker testing will be addressed. As biomarker testing moves exclusively from advanced-stage patients into earlier stage disease, the surgeon should be aware of the relevant markers and work with the pathologist and oncologist to ensure that this information is available to facilitate timely access to therapies not just in the advanced setting, but in consideration of neoadjuvant and adjuvant care.
Collapse
Affiliation(s)
- Julia Naso
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Ying-Chun Lo
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lynette M Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts, USA
| |
Collapse
|
31
|
Bauer DR, Chafin DR. Assessing Tissue Fixation Time and Quality with Label-free Mid Infrared Spectroscopy and Machine Learning. Biopreserv Biobank 2022; 21:208-216. [PMID: 36516138 PMCID: PMC10125394 DOI: 10.1089/bio.2022.0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objectives: This work investigates whether changes in a biospecimen's molecular composition from formaldehyde fixation drive changes in the mid infrared (MID-IR) spectrum. Our ultimate goal was to develop an analytical metrology that could be used to accurately determine the fixation time of a tissue sample as a surrogate to overall tissue quality. Methods: Multiple unstained formalin-fixed paraffin-embedded tissue samples were scanned with an MID-IR microscope to identify a molecular fingerprint of formaldehyde fixation. The fixation specific patterns were then mined to develop a predictive model. A multiple tissue experiment using greater than 100 samples was designed to train the algorithm and validate the accuracy of predicting fixation status. Results: We present data that formaldehyde crosslinking results in alterations to multiple bands of the MID-IR spectra. The impact was most dramatic in the Amide I band, which is sensitive to the conformational state of proteins. The spectroscopic fixation signature was used to train a machine-learning model that could predict fixation time of unknown tissues with an average accuracy of 1.4 hours. Results were validated by histological stain quality for bcl-2, FOXP3, and ki-67. Further, two-dimensional imaging was used to visualize the spatial dependence of fixation, as demonstrated by multiple features in the tissue's vibrational spectra. Conclusions: This work demonstrates that it is possible to predict the fixation status of tissues for which the preanalytics are unknown. This novel capability could help standardize clinical tissue diagnostics and ensure every patient gets the absolutely best treatment based on the highest quality tissue sample.
Collapse
Affiliation(s)
- Daniel R Bauer
- Roche Diagnostics Solutions, Pathology Research and Early Development (Ventana Medical Systems, Inc.), Tucson, Arizona, USA
| | - David R Chafin
- Roche Diagnostics Solutions, Pathology Research and Early Development (Ventana Medical Systems, Inc.), Tucson, Arizona, USA
| |
Collapse
|
32
|
S. Charantimath V, S. Bubanale V. Effect of centrifugation force and time on the analysis of lactate dehydrogenase and potassium in the serum samples. Biomedicine (Taipei) 2022. [DOI: 10.51248/.v42i5.1915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Introduction and Aim: Any imperfection that occurs during any stage of the testing process is described as laboratory error. Increasing requirements of biochemical tests, numerous patient samples and automation has forced laboratory work to be carried out at a faster speed. Few studies are shown to investigate the influence of settings of centrifugation of less than 10 minutes on the laboratory result in serum. Thus, our study was aimed to see the effect of centrifugation force and time on the analysis of lactate dehydrogenase(LDH) and potassium from serum samples.
Methodology: Samples were collected from 61 healthy volunteers. 5ml was taken in two separate BD vacutainer serum tubes. Tube 1 was centrifuged for 2000g for 10 minutes, tube 2 for 5 minutes 3000g, and analysed for LDH and potassium.
Results: A significant difference was observed between 5 min (U/L) (3000g) and 10 min (U/L) (2000g) with LDH and 5 min (mmol/l) 3000g and 10 min (mmol/l) 2000g with potassium.
Conclusion: LDH and potassium levels were found to be raised by increasing the centrifugal force to 3000g. Hence, the standard centrifugation protocol of 10 min at 2000 or 2500 rpm is to be followed to get the accurate results.
Collapse
|
33
|
Chiriboga L, Callis GM, Wang Y, Chlipala E. Guide for collecting and reporting metadata on protocol variables and parameters from slide-based histotechnology assays to enhance reproducibility. J Histotechnol 2022; 45:132-147. [DOI: 10.1080/01478885.2022.2134022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Luis Chiriboga
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- NYULH Center for Biospecimen Research and Development, New York, NY, USA
| | | | - Yongfu Wang
- Stowers Institute for Medical Research, Kansas, MO, USA
| | | |
Collapse
|
34
|
Gobbi H, Carvalho FM, Rahal RMS, Klôh MI, Pitol DL. Optimizing pathological assessment of breast cancer in Brazil: recommendations from a multidisciplinary working group on the tumor-tissue journey. SURGICAL AND EXPERIMENTAL PATHOLOGY 2022. [DOI: 10.1186/s42047-022-00121-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
AbstractTimely and correct assessment of histopathological, immunohistochemical and molecular features of biopsy and surgical specimens is of paramount importance in the provision of care to patients with breast cancer, particularly in the current era of precision oncology. In order to ensure that tissue samples are obtained, processed, analyzed and reported in an optimal way, a concerted effort is required by institutions and individuals, taking into account state-of-the-art scientific and technical knowledge and circumventing logistic and operational constraints. This may be particularly challenging in some settings due to several sources of economic, structural, organizational and communication inefficiencies. In the current article, we present a brief review of breast cancer epidemiology and challenges in the disease diagnosis, especially in Brazil, and report the results of a multidisciplinary working group convened in May 2020 in an expert panel to identify and discuss the barriers and challenges related to the journey of breast cancer samples in Brazil. Following the identification of the issues, the working group also discussed and proposed recommendations for improving the journey and quality of breast cancer samples based on their professional experience and the current scientific literature, including guidelines of national and international health organizations (e.g. World Health Organization), consensus of medical societies and other published literature on the topic. We outline the most salient issues related to that journey in Brazilian public and private medical institutions, based on the experts’ clinical experience, since all of them are actively working at both sectors, and discuss current recommendations to address these issues aiming at mitigating and preventing preanalytical and analytical issues affecting diagnostic and therapeutic decisions. Such issues are grouped under four headings pertaining to education, communication, procedures in the operating room and sample transportation, and procedures in the pathology laboratory. Selected recommendations based on the current literature and discussed by the group of Brazilian experts are reviewed, which may mitigate the issues identified and optimize diagnostic and therapeutic decisions for patients with breast cancer, currently the most frequent malignant tumor worldwide and in Brazil. This paper has been submitted and published jointly, upon invitation and consent, in both the Surgical and Experimental Pathology and the Mastology journals.
Collapse
|
35
|
Bapat PR, Epari S, Joshi PV, Dhanavade DS, Rumde RH, Gurav MY, Shetty OA, Desai SB. Comparative Assessment of DNA Extraction Techniques From Formalin-Fixed, Paraffin-Embedded Tumor Specimens and Their Impact on Downstream Analysis. Am J Clin Pathol 2022; 158:739-749. [PMID: 36197908 DOI: 10.1093/ajcp/aqac122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 09/05/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES Good-quality nucleic acid extraction from formalin-fixed, paraffin-embedded (FFPE) specimens remains a challenge in molecular-oncopathology practice. This study evaluates the efficacy of an in-house developed FFPE extraction buffer compared with other commercially available kits. METHODS Eighty FFPE specimens processed in different surgical pathology laboratories formed the study sample. DNA extraction was performed using three commercial kits and the in-house developed FFPE extraction buffer. DNA yield was quantified by a NanoDrop spectrophotometer and Qubit Fluorometer, and its purity was measured by the 260/280-nm ratio. A fragment analyzer system was used for accurate sizing of DNA fragments of FFPE DNA. The downstream effects of all extraction methods were evaluated by polymerase chain reaction (PCR) and Sanger sequencing. RESULTS In comparison with the commercial kits, the in-house buffer yielded higher DNA quantity and quality number (P < .0001). In addition, DNA integrity and fragment size were preserved in a significantly greater number of samples isolated with the in-house buffer (P < .05). The target PCR amplification rate with the in-house buffer extracted samples was also significantly higher, with 98% of the samples showing interpretable sequencing results. CONCLUSIONS The in-house developed FFPE extraction buffer performed superior to other methods in terms of suitability for downstream applications, time, cost-efficiency, and ease of performance.
Collapse
Affiliation(s)
- Prachi R Bapat
- Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sridhar Epari
- Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Pradnya V Joshi
- Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Dipika S Dhanavade
- Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Rachna H Rumde
- Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Mamta Y Gurav
- Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Omshree A Shetty
- Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| | - Sangeeta B Desai
- Molecular Pathology, Tata Memorial Hospital, Homi Bhabha National Institute, Mumbai, India
| |
Collapse
|
36
|
Dobritoiu F, Baltan A, Chefani A, Billingham K, Chenard MP, Vaziri R, Lacroix-Triki M, Waydelich A, Erb G, Andersson E, Cañamero M, Toro P, Wedden S, D’Arrigo C. Tissue Selection for PD-L1 Testing in Triple Negative Breast Cancer (TNBC). Appl Immunohistochem Mol Morphol 2022; 30:549-556. [PMID: 36036647 PMCID: PMC9444286 DOI: 10.1097/pai.0000000000001053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 06/13/2022] [Indexed: 11/26/2022]
Abstract
Atezolizumab in combination with nab-paclitaxel has been introduced for the treatment of locally advanced or recurrent triple negative breast cancer (TNBC). Patient selection relies on the use of immunohistochemistry using a specific monoclonal PD-L1 antibody (clone SP142) in a tightly controlled companion diagnostic test (CDx) with a defined interpretative algorithm. Currently there are no standardized recommendations for selecting the optimal tissue to be tested and there is limited data to support decision making, raising the possibility that tissue selection may bias test results. We compared PD-L1 SP142 assessment in a collection of 73 TNBC cases with matched core biopsies and excision samples. There was good correlation between PD-L1-positive core biopsy and subsequent excision, but we found considerable discrepancy between PD-L1 negative core biopsy and matched excision, with a third of cases found negative on core biopsies converting to positive upon examination of the excision tissue. In view of these findings, we developed a workflow for the clinical testing of TNBC for PD-L1 and implemented it in a central referral laboratory. We present audit data from the clinical PD-L1 testing relating to 2 years of activities, indicating that implementation of this workflow results in positivity rates in our population of TNBC similar to those of IMpassion130 clinical trial. We also developed an online atlas with a precise numerical annotation to aid pathologists in the interpretation of PD-L1 scoring in TNBC.
Collapse
Affiliation(s)
- Florin Dobritoiu
- Poundbury Cancer Institute
- Department of Pathology, University Emergency Hospital, Bucharest, Romania
| | | | | | - Kim Billingham
- Department of Pathology, Great Western Hospital, Swindon, UK
| | | | - Reza Vaziri
- Department of Pathology, Worcestershire Acute Hospitals, Worcester, UK
| | | | - Anne Waydelich
- Oncology Medical and Government Affairs Roche Diagnostics EMEA-LATAM Region, Rotkreuz, Switzerland
| | - Gilles Erb
- Global Medical Affairs/PDMA, Roche Basel, Switzerland
| | - Emilia Andersson
- Oncology Medical and Government Affairs Roche Diagnostics EMEA-LATAM Region, Rotkreuz, Switzerland
| | - Marta Cañamero
- Oncology Medical and Government Affairs Roche Diagnostics EMEA-LATAM Region, Rotkreuz, Switzerland
| | - Paula Toro
- Clinical Development & Medical Affairs, Roche Diagnostics Solutions, Tucson
| | | | | |
Collapse
|
37
|
De Martino E, Medeot C, D'Amico L, Stanta G, Bonin S. Impact of standardization in tissue processing: the performance of different fixatives. N Biotechnol 2022; 71:30-36. [PMID: 35878783 DOI: 10.1016/j.nbt.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 10/31/2022]
Abstract
Most tissues in clinical practice are formalin-fixed and paraffin-embedded for histological as well as molecular analyses. The reproducibility and uniformity of molecular analyses is strictly dependent on the quality of the biomolecules, which is highly influenced by pre-analytical processes. In this study, the effect of different fixatives was compared, including formalin, Bouin's solution, RCL2® and TAG-1™ fixatives, by stringent application of ISO standards in mouse liver tissue processing, including formalin-free transport of tissues and tissue grossing in a refrigerated environment. The effect of fixatives was studied in terms of nucleic acid quality at the time of tissue processing and after one year of tissue storage at room temperature in the dark. Furthermore, a microcomputed tomography (CT) scan analysis was applied to investigate the paraffin embedding. The results show that the application of ISO standards in tissue processing allows analysis of 400 bases amplicons from RNA and 1000 bases from DNA, even in extracts from formalin-fixed and paraffin-embedded tissues. However, after one year storage at room temperature in the dark, a degradation of the nucleic acids was observed. Nevertheless, extracts can still be analyzed, but for metachronous tests it is highly recommended to repeat the quantitation of housekeeping genes in order to standardize the extent of nucleic acid degradation.
Collapse
Affiliation(s)
| | - Caterina Medeot
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Lorenzo D'Amico
- Department of Physics, University of Trieste, Trieste, Italy; Elettra-Sincrotrone Trieste S.C.p.A., Area Science Park, Trieste, Italy
| | - Giorgio Stanta
- Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Serena Bonin
- Department of Medical Sciences, University of Trieste, Trieste, Italy.
| |
Collapse
|
38
|
Zhang Y, Blomquist TM, Kusko R, Stetson D, Zhang Z, Yin L, Sebra R, Gong B, Lococo JS, Mittal VK, Novoradovskaya N, Yeo JY, Dominiak N, Hipp J, Raymond A, Qiu F, Arib H, Smith ML, Brock JE, Farkas DH, Craig DJ, Crawford EL, Li D, Morrison T, Tom N, Xiao W, Yang M, Mason CE, Richmond TA, Jones W, Johann DJ, Shi L, Tong W, Willey JC, Xu J. Deep oncopanel sequencing reveals within block position-dependent quality degradation in FFPE processed samples. Genome Biol 2022; 23:141. [PMID: 35768876 PMCID: PMC9241261 DOI: 10.1186/s13059-022-02709-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 06/15/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Clinical laboratories routinely use formalin-fixed paraffin-embedded (FFPE) tissue or cell block cytology samples in oncology panel sequencing to identify mutations that can predict patient response to targeted therapy. To understand the technical error due to FFPE processing, a robustly characterized diploid cell line was used to create FFPE samples with four different pre-tissue processing formalin fixation times. A total of 96 FFPE sections were then distributed to different laboratories for targeted sequencing analysis by four oncopanels, and variants resulting from technical error were identified. RESULTS Tissue sections that fail more frequently show low cellularity, lower than recommended library preparation DNA input, or target sequencing depth. Importantly, sections from block surfaces are more likely to show FFPE-specific errors, akin to "edge effects" seen in histology, while the inner samples display no quality degradation related to fixation time. CONCLUSIONS To assure reliable results, we recommend avoiding the block surface portion and restricting mutation detection to genomic regions of high confidence.
Collapse
Affiliation(s)
- Yifan Zhang
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Thomas M Blomquist
- (Formerly) Department of Pathology, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
- Lucas County Coroner's Office, 2595 Arlington Ave, Toledo, OH, 43614, USA
| | - Rebecca Kusko
- Immuneering Corporation, 245 Main St, Cambridge, MA, 02142, USA
| | - Daniel Stetson
- Astrazeneca Pharmaceuticals, 35 Gatehouse Dr, Waltham, MA, 02451, USA
| | - Zhihong Zhang
- Research and Development, Burning Rock Biotech, Shanghai, 201114, China
| | - Lihui Yin
- (Formerly) Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Robert Sebra
- Icahn Institute and Department of Genetics and Genomic Sciences Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Binsheng Gong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | | | - Vinay K Mittal
- Thermo Fisher Scientific, 110 Miller Ave, Ann Arbor, MI, 48104, USA
| | | | - Ji-Youn Yeo
- Department of Pathology, University of Toledo, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Nicole Dominiak
- Department of Pathology, University of Toledo, 3000 Arlington Ave, Toledo, OH, 43614, USA
| | - Jennifer Hipp
- Department of Pathology, Strata Oncology, Inc., Ann Arbor, MI, 48103, USA
| | - Amelia Raymond
- Astrazeneca Pharmaceuticals, 35 Gatehouse Dr, Waltham, MA, 02451, USA
| | - Fujun Qiu
- Research and Development, Burning Rock Biotech, Shanghai, 201114, China
| | - Hanane Arib
- Icahn Institute and Department of Genetics and Genomic Sciences Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Melissa L Smith
- Icahn Institute and Department of Genetics and Genomic Sciences Icahn School of Medicine at Mount Sinai, 1425 Madison Ave, New York, NY, 10029, USA
| | - Jay E Brock
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Daniel H Farkas
- Pathology and Laboratory Medicine Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Daniel J Craig
- Department of Medicine, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Erin L Crawford
- Department of Medicine, College of Medicine and Life Sciences, The University of Toledo, Toledo, OH, 43614, USA
| | - Dan Li
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - Tom Morrison
- Accugenomics, Inc., 1410 Commonwealth Drive, Suite 105, Wilmington, NC, 20403, USA
| | - Nikola Tom
- Center of Molecular Medicine, Central European Institute of Technology, Masaryk University, Kamenice 5, 625 00, Brno, Czech Republic
- EATRIS ERIC- European Infrastructure for Translational Medicine, De Boelelaan 1118, 1081 HZ, Amsterdam, The Netherlands
| | - Wenzhong Xiao
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, 94304, USA
| | - Mary Yang
- Department of Information Science, University of Arkansas at Little Rock, 2801 S. Univ. Ave, Little Rock, AR, 72204, USA
| | - Christopher E Mason
- Department of Physiology and Biophysics, Weill Cornell Medicine, Cornell University, New York, NY, 10065, USA
| | - Todd A Richmond
- Market & Application Development Bioinformatics, Roche Sequencing Solutions Inc., 4300 Hacienda Dr, Pleasanton, CA, 94588, USA
| | - Wendell Jones
- Q2 Solutions - EA Genomics, 5927 S Miami Blvd, Morrisville, NC, 27560, USA
| | - Donald J Johann
- Winthrop P Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W Markham St, Little Rock, AR, 72205, USA
| | - Leming Shi
- State Key Laboratory of Genetic Engineering, School of Life Sciences and Shanghai Cancer Hospital/Cancer Institute, Fudan University, Shanghai, 200438, China
- Human Phenome Institute, Fudan University, Shanghai, 201203, China
- Fudan-Gospel Joint Research Center for Precision Medicine, Fudan University, Shanghai, 200438, China
| | - Weida Tong
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA
| | - James C Willey
- Departments of Medicine, Pathology, and Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Sciences Campus, 3000 Arlington Ave, Toledo, OH, 43614, USA.
| | - Joshua Xu
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, U.S. Food and Drug Administration, Jefferson, AR, 72079, USA.
| |
Collapse
|
39
|
Velu PD, Cushman-Vokoun A, Ewalt MD, Feilotter H, Gastier-Foster JM, Goswami RS, Laudadio J, Olsen RJ, Johnson R, Schlinsog A, Douglas A, Sandersfeld T, Kaul KL. Alignment of Fellowship Training with Practice Patterns for Molecular Pathologists: A Report of the Association for Molecular Pathology Training and Education Committee. J Mol Diagn 2022; 24:825-840. [PMID: 35690309 DOI: 10.1016/j.jmoldx.2022.04.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 04/12/2022] [Accepted: 04/26/2022] [Indexed: 12/19/2022] Open
Abstract
In the two decades since Accreditation Council for Graduate Medical Education-accredited Molecular Genetic Pathology fellowships began, the field of clinical molecular pathology has evolved considerably. The American Board of Pathology gathered data from board-certified molecular genetic pathologists assessing the alignment of skills and knowledge gained during fellowship with current needs on the job. The Association of Molecular Pathology conducted a parallel survey of program directors, and included questions on how various topics were taught during fellowship, as well as ranking their importance. Both surveys showed that most training aligned well with the practice needs of former trainees. Genomic profiling of tumors by next-generation sequencing, bioinformatics, laboratory management, and regulatory issues were topics thought to require increased emphasis in training. Topics related to clinical genetics and microbiology were deemed less important by those in practice, perhaps reflecting the increasing subspecialization of molecular pathologists. Program directors still viewed these topics as important to provide foundational knowledge. Parentage, identity, and human leukocyte antigen testing were less important to both survey audiences. These data may be helpful in guiding future adjustments to the Molecular Genetic Pathology curriculum and Accreditation Council for Graduate Medical Education program requirements.
Collapse
Affiliation(s)
- Priya D Velu
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York
| | - Allison Cushman-Vokoun
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Mark D Ewalt
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Harriet Feilotter
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Julie M Gastier-Foster
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; Departments of Pediatrics and Pathology and Immunology, Baylor College of Medicine, Houston, Texas; Department of Pathology, Ohio State University College of Medicine, Columbus, Ohio
| | - Rashmi S Goswami
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Laboratory Medicine and Molecular Diagnostics/Department of Laboratory Medicine and Pathobiology, Sunnybrook Health Sciences Centre/University of Toronto, Toronto, Ontario, Canada
| | - Jennifer Laudadio
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Randall J Olsen
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York; Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Houston, Texas
| | | | | | | | | | - Karen L Kaul
- Molecular Genetic Pathology Curriculum Update Working Group of the Training and Education Committee, Association for Molecular Pathology, Rockville, Maryland; NorthShore University HealthSystem, University of Chicago Pritzker School of Medicine, Evanston, Illinois.
| |
Collapse
|
40
|
Grafodatskaya D, O'Rielly DD, Bedard K, Butcher DT, Howlett CJ, Lytwyn A, McCready E, Parboosingh J, Spriggs EL, Vaags AK, Stockley TL. Practice guidelines for BRCA1/2 tumour testing in ovarian cancer. J Med Genet 2022; 59:727-736. [PMID: 35393334 PMCID: PMC9340048 DOI: 10.1136/jmedgenet-2021-108238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 02/24/2022] [Indexed: 12/26/2022]
Abstract
The purpose of this document is to provide pre-analytical, analytical and post-analytical considerations and recommendations to Canadian clinical laboratories developing, validating and offering next-generation sequencing (NGS)-based BRCA1 and BRCA2 (BRCA1/2) tumour testing in ovarian cancers. This document was drafted by the members of the Canadian College of Medical Geneticists (CCMG) somatic BRCA Ad Hoc Working Group, and representatives from the Canadian Association of Pathologists. The document was circulated to the CCMG members for comment. Following incorporation of feedback, this document has been approved by the CCMG board of directors. The CCMG is a Canadian organisation responsible for certifying medical geneticists and clinical laboratory geneticists, and for establishing professional and ethical standards for clinical genetics services in Canada. The current CCMG Practice Guidelines were developed as a resource for clinical laboratories in Canada; however, they are not inclusive of all information laboratories should consider in the validation and use of NGS for BRCA1/2 tumour testing in ovarian cancers.
Collapse
Affiliation(s)
- Daria Grafodatskaya
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Hamilton Regional Laboratory Medicine Program, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Darren D O'Rielly
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, Newfoundland, Canada.,Centre for Translational Genomes & Division of Genetics, Eastern Regional Health Authority, St. John's, Newfoundland, Canada
| | - Karine Bedard
- Département de Pathologie et Biologie cellulaire, Université de Montréal, Montreal, Québec, Canada.,Laboratoire de Diagnostic Moléculaire, Centre hospitalier de l'Université de Montréal, Montreal, Québec, Canada
| | - Darci T Butcher
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Hamilton Regional Laboratory Medicine Program, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Christopher J Howlett
- Department of Pathology and Laboratory Medicine, Schulich School of Medicine & Dentistry, Wester University, London, Ontario, Canada
| | - Alice Lytwyn
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Hamilton Regional Laboratory Medicine Program, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Elizabeth McCready
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada.,Hamilton Regional Laboratory Medicine Program, Hamilton Health Sciences, Hamilton, Ontario, Canada
| | - Jillian Parboosingh
- Department of Medical Genetics, Alberta Children's Hospital Research Institute for Child and Maternal Health, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Genetics and Genomics, Alberta Precision Laboratories, Calgary, Alberta, Canada
| | - Elizabeth L Spriggs
- Genomics, Diagnostic Services, Shared Health Manitoba, Winnipeg, Manitoba, Canada.,Department of Biochemistry and Medical Genetics, Max Rady College of Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrea K Vaags
- Laboratory Medicine and Genetics, Trillium Health Partners, Mississauga, Ontario, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Tracy L Stockley
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada .,Department of Clinical Laboratory Genetics, Laboratory Medicine Program, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
41
|
Marshall JL, Peshkin BN, Yoshino T, Vowinckel J, Danielsen HE, Melino G, Tsamardinos I, Haudenschild C, Kerr DJ, Sampaio C, Rha SY, FitzGerald KT, Holland EC, Gallagher D, Garcia-Foncillas J, Juhl H. The Essentials of Multiomics. Oncologist 2022; 27:272-284. [PMID: 35380712 PMCID: PMC8982374 DOI: 10.1093/oncolo/oyab048] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 11/05/2021] [Indexed: 11/13/2022] Open
Abstract
Within the last decade, the science of molecular testing has evolved from single gene and single protein analysis to broad molecular profiling as a standard of care, quickly transitioning from research to practice. Terms such as genomics, transcriptomics, proteomics, circulating omics, and artificial intelligence are now commonplace, and this rapid evolution has left us with a significant knowledge gap within the medical community. In this paper, we attempt to bridge that gap and prepare the physician in oncology for multiomics, a group of technologies that have gone from looming on the horizon to become a clinical reality. The era of multiomics is here, and we must prepare ourselves for this exciting new age of cancer medicine.
Collapse
Affiliation(s)
- John L Marshall
- Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC, USA
| | - Beth N Peshkin
- Georgetown University, Lombardi Comprehensive Cancer Center, Washington, DC, USA
| | | | | | - Håvard E Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, Radiumhospitalet, Montebello, Oslo, Norway
| | - Gerry Melino
- Department of Experimental Medicine, TOR, University of Rome Tor Vergata, Rome, Italy
| | - Ioannis Tsamardinos
- JADBio Gnosis DA, N. Plastira 100, Science and Technology Park of Crete and Institute of Applied and Computational Mathematics, Foundation for Research and Technology Hellas, Heraklion, GR, Greece
| | | | - David J Kerr
- Nuffield Division of Clinical and Laboratory Sciences, Level 4, Academic Block, John Radcliffe Infirmary, Headington, Oxford, UK
| | | | - Sun Young Rha
- Yonsei Cancer Center, Yonsei University College of Medicine, Seodaemun-Ku, Seoul, Korea
| | - Kevin T FitzGerald
- Department of Medical Humanities in the School of Medicine, Creighton University, Omaha, NE, USA
| | - Eric C Holland
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - David Gallagher
- St. James’s Hospital/Trinity College Dublin, St. Raphael’s House, Dublin, Ireland
| | - Jesus Garcia-Foncillas
- Cancer Institute, Fundacion Jimenez Diaz University Hospital, Autonomous University, Madrid, Spain
| | | |
Collapse
|
42
|
Monteiro CRDA, Korkes F, Krutman-Zveibil D, Glina S. Fibroblast growth factor receptor 3 gene (FGFR3) mutations in high-grade muscle-invasive urothelial bladder cancer in a Brazilian population: evaluation and prevalence. EINSTEIN-SAO PAULO 2022; 20:eAO6450. [PMID: 35384983 PMCID: PMC8967311 DOI: 10.31744/einstein_journal/2022ao6450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Accepted: 06/10/2021] [Indexed: 11/05/2022] Open
Abstract
Objective To understand the feasibility of FGFR3 tests in the Brazilian public health context, and to sample the mutational burden of this receptor in high-grade muscle invasive bladder cancer. Methods A total of 31 patients with high-grade muscle-invasive bladder cancer were included in the present study. Either transurethral resection of bladder tumor or radical cystectomy specimens were analyzed. Formalin-fixed paraffin-embedded tissue blocks were sectioned, hematoxylin and eosin stained, and histologic sections were reviewed. Total RNA was extracted using the RNeasy DSP formalin-fixed paraffin-embedded kit. Qualitative results were displayed in Rotor-Gene AssayManager software. Results Six patients were excluded. From the samples analyzed, four (16.7%) were considered inadequate and could not have their RNA extracted. Two patients presented FGFR3 mutations, accounting for 9.5% of material available for adequate analysis. The two mutations detected included a Y373C mutation in a male patient and a S249C mutation in a female patient. Conclusion FGFR3 mutations could be analyzed in 84% of our cohort and occurred in 9.5% of patients with high-grade muscle invasive bladder cancer in this Brazilian population. FGFR3 gene mutations are targets for therapeutic drugs in muscle-invasive bladder cancer. For this reason, know the frequency of these mutations can have a significant impact on public health policies and costs provisioning.
Collapse
|
43
|
Conde E, Rojo F, Gómez J, Enguita AB, Abdulkader I, González A, Lozano D, Mancheño N, Salas C, Salido M, Salido-Ruiz E, de Álava E. Molecular diagnosis in non-small-cell lung cancer: expert opinion on ALK and ROS1 testing. J Clin Pathol 2022; 75:145-153. [PMID: 33875457 PMCID: PMC8862096 DOI: 10.1136/jclinpath-2021-207490] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/22/2021] [Accepted: 03/24/2021] [Indexed: 01/09/2023]
Abstract
The effectiveness of targeted therapies with tyrosine kinase inhibitors in non-small-cell lung cancer (NSCLC) depends on the accurate determination of the genomic status of the tumour. For this reason, molecular analyses to detect genetic rearrangements in some genes (ie, ALK, ROS1, RET and NTRK) have become standard in patients with advanced disease. Since immunohistochemistry is easier to implement and interpret, it is normally used as the screening procedure, while fluorescence in situ hybridisation (FISH) is used to confirm the rearrangement and decide on ambiguous immunostainings. Although FISH is considered the most sensitive method for the detection of ALK and ROS1 rearrangements, the interpretation of results requires detailed guidelines. In this review, we discuss the various technologies available to evaluate ALK and ROS1 genomic rearrangements using these techniques. Other techniques such as real-time PCR and next-generation sequencing have been developed recently to evaluate ALK and ROS1 gene rearrangements, but some limitations prevent their full implementation in the clinical setting. Similarly, liquid biopsies have the potential to change the treatment of patients with advanced lung cancer, but further research is required before this technology can be applied in routine clinical practice. We discuss the technical requirements of laboratories in the light of quality assurance programmes. Finally, we review the recent updates made to the guidelines for the determination of molecular biomarkers in patients with NSCLC.
Collapse
Affiliation(s)
- Esther Conde
- Department of Pathology and Laboratory of Therapeutic Targets & CIBERONC, HM Hospitales, Madrid, Spain
| | - Federico Rojo
- Department of Pathology, Hospital Universitario Fundacion Jiménez Díaz, Madrid, Spain
| | - Javier Gómez
- Department of Pathology, Hospital Universitario Marques de Valdecilla, Santander, Cantabria, Spain
- Instituto de Investigación Sanitaria Valdecilla IDIVAL, Universidad de Cantabria, Santander, Cantabria, Spain
| | - Ana Belén Enguita
- Department of Pathology, Clínica Dermatológica Internacional, Madrid, Spain
| | - Ihab Abdulkader
- Department of Pathology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Ana González
- Department of Pathology, Hospital Álvaro Cunqueiro, Vigo, Spain
| | - Dolores Lozano
- Department of Pathology, Clinica Universidad de Navarra, Pamplona, Navarra, Spain
| | - Nuria Mancheño
- Department of Pathology, La Fe University and Polytechnic Hospital, Valencia, Comunidad Valenciana, Spain
| | - Clara Salas
- Department of Pathology, Hospital Universitario Puerta del Hierro Majadahonda, Majadahonda, Madrid, Spain
| | - Marta Salido
- Department of Pathology, Hospital del Mar, Barcelona, Spain
| | - Eduardo Salido-Ruiz
- Department of Pathology, Hospital Universitario de Canarias, La Laguna, Canarias, Spain
| | - Enrique de Álava
- Department of Pathology, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| |
Collapse
|
44
|
Kazdal D, Hofman V, Christopoulos P, Ilié M, Stenzinger A, Hofman P. Fusion-positive non-small cell lung carcinoma: Biological principles, clinical practice, and diagnostic implications. Genes Chromosomes Cancer 2022; 61:244-260. [PMID: 34997651 DOI: 10.1002/gcc.23022] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
Based on superior efficacy and tolerability, targeted therapy is currently preferred over chemotherapy and/or immunotherapy for actionable gene fusions that occur in late-stage non-small cell lung carcinoma (NSCLC). Consequently, current clinical practice guidelines mandate testing for ALK, ROS1, NTRK, and RET gene fusions in all patients with newly diagnosed advanced non-squamous NSCLC (NS-NSCLC). Gene fusions can be detected using different approaches, but today RNA next-generation sequencing (NGS) or combined DNA/RNA NGS is the method of choice. The discovery of other gene fusions (involving, eg, NRG1, NUT, FGFR1, FGFR2, MET, BRAF, EGFR, SMARC fusions) and their partners has increased progressively in recent years, leading to the development of new and promising therapies and mandating the development and implementation of comprehensive detection methods. The purpose of this review is to focus on recent data concerning the main gene fusions identified in NSCLC, followed by the discussion of major challenges in this domain.
Collapse
Affiliation(s)
- Daniel Kazdal
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC) Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany
| | - Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, FHU OncoAge, Nice, France.,Centre Antoine Lacassagne Cancer Center, Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France.,Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Petros Christopoulos
- Translational Lung Research Center (TLRC) Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany.,Thoraxklinik and National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, FHU OncoAge, Nice, France.,Centre Antoine Lacassagne Cancer Center, Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France.,Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,German Center for Lung Research (DZL), Heidelberg, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Pasteur Hospital, Université Côte d'Azur, FHU OncoAge, Nice, France.,Centre Antoine Lacassagne Cancer Center, Université Côte d'Azur, CNRS, INSERM, IRCAN, FHU OncoAge, Nice, France.,Hospital-Integrated Biobank BB-0033-00025, Université Côte d'Azur, CHU Nice, FHU OncoAge, Nice, France
| |
Collapse
|
45
|
Genestie C, Gladieff L, Frère-Belda MAL, Lortholary A, Vaur D, Treilleux I, Lyonnet DS. Diagnostic histologique et moléculaire des cancers de l'ovaire - recommandations pour la pratique clinique Saint-Paul 2021: Histological and molecular diagnosis of ovarian. Bull Cancer 2021; 108:S33-S38. [PMID: 34955160 DOI: 10.1016/s0007-4551(21)00585-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Oncogenetic testing is now part of standard management in high grade ovarian cancer, including at least mutational status of BRCA1/BRCA2 genes. If necessary, tumor genetic testing is followed by constitutional testing to either confirm the constitutional origin of variants identified in BRCA1/2 genes or detect variants in other predisposition genes. The whole process including prescription of tumoral testing, retrieval of analysis report and communication of results must be formalized, as well as information on possible consequences of the results for the patient and her family. Tumor material must meet criteria of size and cellularity to allow high-quality analysis. These samples are processed during the preanalytical phase with two major steps : time of cold ischemia and fixation. Only pathogenic (Class V) and likely pathogenic (Class IV) variants shown in tumor tissue are mentioned in the report. Currently, only BRCA1 and BRCA2 genes are routinely studied but, in the future, analysis will be extended to other genes involved in homologous recombination repair. In patients without BRCA mutation, other biomarkers reflecting sensitivity to PARP inhibitors, such as HRD scores (homologous recombination deficiency) that appeared recently, will have to be implemented in routine practice in order to better select patients for these treatments and choose optimal therapy.
Collapse
Affiliation(s)
| | - Laurence Gladieff
- Département d'oncologie médicale et unité d'oncogénétique, institut Claudius Regaud, IUCT-Oncopole, Toulouse, France
| | | | | | - Dominique Vaur
- Laboratoire de biologie et de génétique du cancer, Centre François-Baclesse, Caen, France
| | | | | |
Collapse
|
46
|
Pathology: Hub and Integrator of Modern, Multidisciplinary [Precision] Oncology. Clin Cancer Res 2021; 28:265-270. [PMID: 34893516 DOI: 10.1158/1078-0432.ccr-21-1206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 10/13/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022]
Abstract
Every profession has an essential, behind-the-scenes component without which it cannot function. Medicine is no exception. Pathologists are one of medicine's group of specialists whose work as diagnosticians is critical for patient care. This article describes the multifaceted role that pathologists play in oncology practice and cancer research. To do so, we will highlight the role of pathologists in a typical "day-in-the-life" of a patient's journey in cancer care. Clinicians, clinical trialists, radiologists, researchers, and pathologists, all work together to provide optimal, multidisciplinary care for patients with cancer.
Collapse
|
47
|
Making a science out of preanalytics: An analytical method to determine optimal tissue fixation in real-time. PLoS One 2021; 16:e0258495. [PMID: 34648597 PMCID: PMC8516200 DOI: 10.1371/journal.pone.0258495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 09/28/2021] [Indexed: 11/19/2022] Open
Abstract
Modern histopathology is built on the cornerstone principle of tissue fixation, however there are currently no analytical methods of detecting fixation and as a result, in clinical practice fixation is highly variable and a persistent source of error. We have previously shown that immersion in cold formalin followed by heated formalin is beneficial for preservation of histomorphology and have combined two-temperature fixation with ultra-sensitive acoustic monitoring technology that can actively detect formalin diffusing into a tissue. Here we expand on our previous work by developing a predictive statistical model to determine when a tissue is properly diffused based on the real-time acoustic signal. We trained the model based on the morphology and characteristic diffusion curves of 30 tonsil cores. To test our model, a set of 87 different tonsil samples were fixed with four different protocols: dynamic fixation according to our predictive algorithm (C/H:Dynamic, N = 18), gold-standard 24 hour room temperature (RT:24hr, N = 24), 6 hours in cold formalin followed by 1 hour in heated formalin (C/H:6+1, N = 21), and 2 hours in cold formalin followed by 1 hour in heated formalin (C/H:2+1, N = 24). Digital pathology analysis revealed that the C/H:Dynamic samples had FOXP3 staining that was spatially uniform and statistically equivalent to RT:24hr and C/H:6+1 fixation protocols. For comparison, the intentionally underfixed C/H:2+1 samples had significantly suppressed FOXP3 staining (p<0.002). Furthermore, our dynamic fixation protocol produced bcl-2 staining concordant with standard fixation techniques. The dynamically fixed samples were on average only submerged in cold formalin for 4.2 hours, representing a significant workflow improvement. We have successfully demonstrated a first-of-its-kind analytical method to assess the quality of fixation in real-time and have confirmed its performance with quantitative analysis of downstream staining. This innovative technology could be used to ensure high-quality and standardized staining as part of an expedited and fully documented preanalytical workflow.
Collapse
|
48
|
Aggarwal C, Bubendorf L, Cooper WA, Illei P, Borralho Nunes P, Ong BH, Tsao MS, Yatabe Y, Kerr KM. Molecular testing in stage I-III non-small cell lung cancer: Approaches and challenges. Lung Cancer 2021; 162:42-53. [PMID: 34739853 DOI: 10.1016/j.lungcan.2021.09.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 09/02/2021] [Accepted: 09/08/2021] [Indexed: 02/08/2023]
Abstract
Precision medicine in non-small cell lung cancer (NSCLC) is a rapidly evolving area, with the development of targeted therapies for advanced disease and concomitant molecular testing to inform clinical decision-making. In contrast, routine molecular testing in stage I-III disease has not been required, where standard of care comprises surgery with or without adjuvant or neoadjuvant chemotherapy, or concurrent chemoradiotherapy for unresectable stage III disease, without the integration of targeted therapy. However, the phase 3 ADAURA trial has recently shown that the epidermal growth factor receptor (EGFR) tyrosine kinase inhibitor (TKI), osimertinib, reduces the risk of disease recurrence by 80% versus placebo in the adjuvant setting for patients with stage IB-IIIA EGFR mutation-positive NSCLC following complete tumor resection with or without adjuvant chemotherapy, according to physician and patient choice. Treatment with adjuvant osimertinib requires selection of patients based on the presence of an EGFR-TKI sensitizing mutation. Other targeted agents are currently being evaluated in the adjuvant and neoadjuvant settings. Approval of at least some of these other agents is highly likely in the coming years, bringing with it in parallel, a requirement for comprehensive molecular testing for stage I-III disease. In this review, we consider the implications of integrating molecular testing into practice when managing patients with stage I-III non-squamous NSCLC. We discuss best practices, approaches and challenges from pathology, surgical and oncology perspectives.
Collapse
Affiliation(s)
- Charu Aggarwal
- Abramson Cancer Center and Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Lukas Bubendorf
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Switzerland
| | - Wendy A Cooper
- Department of Tissue Pathology and Diagnostic Oncology, Royal Prince Alfred Hospital and NSW Health Pathology, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Western Sydney University, Campbelltown, NSW, Australia
| | - Peter Illei
- Department of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Paula Borralho Nunes
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal; Hospital CUF Descobertas, Lisbon, Portugal
| | - Boon-Hean Ong
- Department of Cardiothoracic Surgery, National Heart Centre Singapore, Singapore
| | - Ming-Sound Tsao
- Department of Pathology, University Health Network, Princess Margaret Cancer Centre, Toronto, Canada
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center, Tokyo, Japan
| | - Keith M Kerr
- Department of Pathology, Aberdeen University, Medical School and Aberdeen Royal Infirmary, Foresterhill, Aberdeen, UK.
| |
Collapse
|
49
|
Tawfik OW, Subramanian J, Caughron S, Mana P, Ewing E, Aboudara M, Borsa J, Schafer L, Saettele T, Jonnalagadda S. Challenges in Pathology Specimen Processing in the New Era of Precision Medicine. Arch Pathol Lab Med 2021; 146:603-610. [PMID: 34424953 DOI: 10.5858/arpa.2021-0089-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/15/2021] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Precision therapies for patients with driver mutations can offer deep and durable responses that correlate with diagnosis, metastasis prognosis, and improvement in survival. Such targeted therapies will continue to increase, pushing us to change our traditional approaches. We needed to search for new tools to effectively integrate technological advancements into our practices because of their capability to improve the efficiency and accuracy of our diagnostic and treatment approaches. Perhaps nothing is as relevant as identifying and implementing new workflows for processing pathologic specimens and for improving communication of critical laboratory information to and from clinicians for appropriate care of patients in an efficient and timely manner. OBJECTIVES.— To define the gold standard in delivering the best care for patients, to identify gaps in the process, and to identify potential solutions that would improve our process, including gaps related to knowledge, skills, attitudes, and practices. DESIGN.— We assembled a team across disciplines to systematically perform a gap analysis study to clarify the discrepancy between the current reality in pathology specimen processing and the desired optimal situation to deliver the results intended for patient care. RESULTS.— A practical collaborative workflow for specimen management seeking the cooperation of the stakeholders in each medical discipline to provide guidelines in specimen collection, delivery, processing, and reporting of results with the ultimate goal of improving patients' outcomes is provided. CONCLUSIONS.— New tools are required to effectively integrate data-driven approaches in specimen processing to meet the new demands.
Collapse
Affiliation(s)
- Ossama William Tawfik
- The Department of Pathology (Tawfik, Caughron, Mana, Ewing), Saint Luke's Health System, Kansas City, Missouri.,MAWD Pathology Group, Kansas City, Kansas (Tawfik, Caughron, Mana, Ewing)
| | - Janakiraman Subramanian
- From the Division of Oncology (Subramanian, Schafer), Saint Luke's Health System, Kansas City, Missouri.,The Department of Medicine (Subramanian), niversity of Missouri, Kansas City
| | - Samuel Caughron
- The Department of Pathology (Tawfik, Caughron, Mana, Ewing), Saint Luke's Health System, Kansas City, Missouri.,MAWD Pathology Group, Kansas City, Kansas (Tawfik, Caughron, Mana, Ewing)
| | - Pradip Mana
- The Department of Pathology (Tawfik, Caughron, Mana, Ewing), Saint Luke's Health System, Kansas City, Missouri.,MAWD Pathology Group, Kansas City, Kansas (Tawfik, Caughron, Mana, Ewing)
| | - Eric Ewing
- The Department of Pathology (Tawfik, Caughron, Mana, Ewing), Saint Luke's Health System, Kansas City, Missouri.,MAWD Pathology Group, Kansas City, Kansas (Tawfik, Caughron, Mana, Ewing)
| | - Matthew Aboudara
- Division of Pulmonology (Aboudara, Saettele), Saint Luke's Health System, Kansas City, Missouri
| | - John Borsa
- Department of Radiology (Borsa), Saint Luke's Health System, Kansas City, Missouri.,Department of Radiology (Borsa), niversity of Missouri, Kansas City
| | - Liudmila Schafer
- From the Division of Oncology (Subramanian, Schafer), Saint Luke's Health System, Kansas City, Missouri
| | - Timothy Saettele
- Division of Pulmonology (Aboudara, Saettele), Saint Luke's Health System, Kansas City, Missouri
| | - Sreeni Jonnalagadda
- Division of Gastroenterology, in the Department of Medicine, (Jonnalagadda), Saint Luke's Health System, Kansas City, Missouri
| |
Collapse
|
50
|
Hofman P. EGFR Status Assessment for Better Care of Early Stage Non-Small Cell Lung Carcinoma: What Is Changing in the Daily Practice of Pathologists? Cells 2021; 10:2157. [PMID: 34440926 PMCID: PMC8392580 DOI: 10.3390/cells10082157] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 02/08/2023] Open
Abstract
The recent emergence of novel neoadjuvant and/or adjuvant therapies for early stage (I-IIIA) non-small cell lung carcinoma (NSCLC), mainly tyrosine kinase inhibitors (TKIs) targeting EGFR mutations and immunotherapy or chemo-immunotherapy, has suddenly required the evaluation of biomarkers predictive of the efficacy of different treatments in these patients. Currently, the choice of one or another of these treatments mainly depends on the results of immunohistochemistry for PD-L1 and of the status of EGFR and ALK. This new development has led to the setup of different analyses for clinical and molecular pathology laboratories, which have had to rapidly integrate a number of new challenges into daily practice and to establish new organization for decision making. This review outlines the impact of the management of biological samples in laboratories and discusses perspectives for pathologists within the framework of EGFR TKIs in early stage NSCLC.
Collapse
Affiliation(s)
- Paul Hofman
- Laboratory of Clinical and Experimental Pathology, CHU Nice, FHU OncoAge, Pasteur Hospital, Université Côte d’Azur, 06108 Nice, France; ; Tel.: +33-492-038-855; Fax: +33-492-8850
- CHU Nice, FHU OncoAge, Hospital-Integrated Biobank BB-0033-00025, Université Côte d’Azur, 06000 Nice, France
| |
Collapse
|