1
|
Caputo A, Pisapia P, L'Imperio V. Current role of cytopathology in the molecular and computational era: The perspective of young pathologists. Cancer Cytopathol 2024; 132:678-685. [PMID: 38748507 DOI: 10.1002/cncy.22832] [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: 03/29/2024] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 11/03/2024]
Abstract
Cytopathology represents a well established diagnostic approach because of its limited cost, reliability, and minimal invasiveness with respect to other methodologies. The evolving complexity of the different classifications systems and the implementation of ancillary techniques to refine the diagnosis is progressively helping in the risk of malignancy stratification, and the adoption of next-generation sequencing techniques contributes to enrich this valuable tool with predictive information, which is always more essential in the tailored medicine era. The recent introduction of digital and computational pathology is further boosting the potentialities of cytopathology, aiding in the interpretation of samples to improve the cost effectiveness of large screening programs and the diagnostic efficiency within intermediate/atypical categories. Moreover, the adoption of artificial intelligence tools is promising to complement molecular investigations, representing a stimulating perspective in the cytopathology field. In this work, the authors tried to summarize the multifaceted nature of this complex and evolving field of pathology, synthesizing the most recent advances and providing the young pathologists' perspective on this fascinating world.
Collapse
Affiliation(s)
- Alessandro Caputo
- Department of Medicine and Surgery, University of Salerno, Fisciano, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo dei Tintori, University of Milano-Bicocca, Milan, Italy
| |
Collapse
|
2
|
Xiaojian Y, Zhanbo Q, Jian C, Zefeng W, Jian L, Jin L, Yuefen P, Shuwen H. Deep learning application in prediction of cancer molecular alterations based on pathological images: a bibliographic analysis via CiteSpace. J Cancer Res Clin Oncol 2024; 150:467. [PMID: 39422817 PMCID: PMC11489169 DOI: 10.1007/s00432-024-05992-z] [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: 04/06/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND The advancements in artificial intelligence (AI) technology for image recognition were propelling molecular pathology research into a new era. OBJECTIVE To summarize the hot spots and research trends in the field of molecular pathology image recognition. METHODS Relevant articles from January 1st, 2010, to August 25th, 2023, were retrieved from the Web of Science Core Collection. Subsequently, CiteSpace was employed for bibliometric and visual analysis, generating diverse network diagrams illustrating keywords, highly cited references, hot topics, and research trends. RESULTS A total of 110 relevant articles were extracted from a pool of 10,205 articles. The overall publication count exhibited a rising trend each year. The leading contributors in terms of institutions, countries, and authors were Maastricht University (11 articles), the United States (38 articles), and Kather Jacob Nicholas (9 articles), respectively. Half of the top ten research institutions, based on publication volume, were affiliated with Germany. The most frequently cited article was authored by Nicolas Coudray et al. accumulating 703 citations. The keyword "Deep learning" had the highest frequency in 2019. Notably, the highlighted keywords from 2022 to 2023 included "microsatellite instability", and there were 21 articles focusing on utilizing algorithms to recognize microsatellite instability (MSI) in colorectal cancer (CRC) pathological images. CONCLUSION The use of DL is expected to provide a new strategy to effectively solve the current problem of time-consuming and expensive molecular pathology detection. Therefore, further research is needed to address issues, such as data quality and standardization, model interpretability, and resource and infrastructure requirements.
Collapse
Affiliation(s)
- Yu Xiaojian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Qu Zhanbo
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Chu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Wang Zefeng
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jian
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Liu Jin
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China
| | - Pan Yuefen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, No.1558, Sanhuan North Road, Wuxing District, Huzhou, 313000, Zhejiang Province, China.
- Key Laboratory of Multiomics Research and Clinical Transformation of Digestive Cancer of Huzhou, Huzhou, China.
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Huzhou, China.
- ASIR(Institute - Association of intelligent systems and robotics), Rueil-Malmaison, France.
| |
Collapse
|
3
|
Gong D, Arbesfeld-Qiu JM, Perrault E, Bae JW, Hwang WL. Spatial oncology: Translating contextual biology to the clinic. Cancer Cell 2024; 42:1653-1675. [PMID: 39366372 DOI: 10.1016/j.ccell.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/01/2024] [Accepted: 09/06/2024] [Indexed: 10/06/2024]
Abstract
Microscopic examination of cells in their tissue context has been the driving force behind diagnostic histopathology over the past two centuries. Recently, the rise of advanced molecular biomarkers identified through single cell profiling has increased our understanding of cellular heterogeneity in cancer but have yet to significantly impact clinical care. Spatial technologies integrating molecular profiling with microenvironmental features are poised to bridge this translational gap by providing critical in situ context for understanding cellular interactions and organization. Here, we review how spatial tools have been used to study tumor ecosystems and their clinical applications. We detail findings in cell-cell interactions, microenvironment composition, and tissue remodeling for immune evasion and therapeutic resistance. Additionally, we highlight the emerging role of multi-omic spatial profiling for characterizing clinically relevant features including perineural invasion, tertiary lymphoid structures, and the tumor-stroma interface. Finally, we explore strategies for clinical integration and their augmentation of therapeutic and diagnostic approaches.
Collapse
Affiliation(s)
- Dennis Gong
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jeanna M Arbesfeld-Qiu
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Ella Perrault
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Jung Woo Bae
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - William L Hwang
- Center for Systems Biology, Department of Radiation Oncology, Center for Cancer Research, Massachusetts General Hospital, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Harvard University, Graduate School of Arts and Sciences, Cambridge, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
4
|
Cazzaniga G, Del Carro F, Eccher A, Becker JU, Gambaro G, Rossi M, Pieruzzi F, Fraggetta F, Pagni F, L'Imperio V. Improving the Annotation Process in Computational Pathology: A Pilot Study with Manual and Semi-automated Approaches on Consumer and Medical Grade Devices. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01248-x. [PMID: 39231887 DOI: 10.1007/s10278-024-01248-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/06/2024]
Abstract
The development of reliable artificial intelligence (AI) algorithms in pathology often depends on ground truth provided by annotation of whole slide images (WSI), a time-consuming and operator-dependent process. A comparative analysis of different annotation approaches is performed to streamline this process. Two pathologists annotated renal tissue using semi-automated (Segment Anything Model, SAM)) and manual devices (touchpad vs mouse). A comparison was conducted in terms of working time, reproducibility (overlap fraction), and precision (0 to 10 accuracy rated by two expert nephropathologists) among different methods and operators. The impact of different displays on mouse performance was evaluated. Annotations focused on three tissue compartments: tubules (57 annotations), glomeruli (53 annotations), and arteries (58 annotations). The semi-automatic approach was the fastest and had the least inter-observer variability, averaging 13.6 ± 0.2 min with a difference (Δ) of 2%, followed by the mouse (29.9 ± 10.2, Δ = 24%), and the touchpad (47.5 ± 19.6 min, Δ = 45%). The highest reproducibility in tubules and glomeruli was achieved with SAM (overlap values of 1 and 0.99 compared to 0.97 for the mouse and 0.94 and 0.93 for the touchpad), though SAM had lower reproducibility in arteries (overlap value of 0.89 compared to 0.94 for both the mouse and touchpad). No precision differences were observed between operators (p = 0.59). Using non-medical monitors increased annotation times by 6.1%. The future employment of semi-automated and AI-assisted approaches can significantly speed up the annotation process, improving the ground truth for AI tool development.
Collapse
Affiliation(s)
- Giorgio Cazzaniga
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Via Pergolesi, 33, 20900, Monza, Italy
| | - Fabio Del Carro
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Via Pergolesi, 33, 20900, Monza, Italy
| | - Albino Eccher
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Jan Ulrich Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Giovanni Gambaro
- Division of Nephrology, Department of Medicine, University of Verona, Verona, Italy
| | - Mattia Rossi
- Division of Nephrology, Department of Medicine, University of Verona, Verona, Italy
| | - Federico Pieruzzi
- Clinical Nephrology, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- School of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy
| | - Filippo Fraggetta
- Pathology Unit, Azienda Sanitaria Provinciale (ASP) Catania, "Gravina" Hospital, Caltagirone, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Via Pergolesi, 33, 20900, Monza, Italy
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo Dei Tintori, University of Milano-Bicocca, Via Pergolesi, 33, 20900, Monza, Italy.
| |
Collapse
|
5
|
Guerini-Rocco E, Venetis K, Cursano G, Mane E, Frascarelli C, Pepe F, Negrelli M, Olmeda E, Vacirca D, Ranghiero A, Trapani D, Criscitiello C, Curigliano G, Rolfo C, Malapelle U, Fusco N. Standardized molecular pathology workflow for ctDNA-based ESR1 testing in HR+/HER2- metastatic breast cancer. Crit Rev Oncol Hematol 2024; 201:104427. [PMID: 38917944 DOI: 10.1016/j.critrevonc.2024.104427] [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: 04/23/2024] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 06/27/2024] Open
Abstract
Mutations in the estrogen receptor alpha gene (ESR1) can lead to resistance to endocrine therapy (ET) in hormone receptor-positive (HR+)/ HER2- metastatic breast cancer (MBC). ESR1 mutations can be detected in up to 40 % of patients pretreated with ET in circulating tumor DNA (ctDNA). Data from prospective randomized trials highlight those patients with HR+/HER2- MBC with detectable ESR1 mutations experience better outcomes when receiving novel selective estrogen receptor degraders (SERDs). There is a high need for optimizing ESR1 testing strategies on liquid biopsy samples in HR+/HER2- MBC, including a hugh quality workflow implementation and molecular pathology reporting standardization. Our manuscript aims to elucidate the clinical and biological rationale for ESR1 testing in MBC, while critically examining the currently available guidelines and recommendations for this specific type of molecular testing on ctDNA. The objective will extend to the critical aspects of harmonization and standardization, specifically focusing on the pathology laboratory workflow. Finally, we propose a clear and comprehensive model for reporting ESR1 testing results on ctDNA in HR+/HER2- MBC.
Collapse
Affiliation(s)
- Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Giulia Cursano
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Eltjona Mane
- Division of Pathology, European Institute of Oncology IRCCS, 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
| | - Francesco Pepe
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | - Mariachiara Negrelli
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; School of Pathology, University of Milan, Milan, Italy
| | - Edoardo Olmeda
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; School of Pathology, University of Milan, Milan, Italy
| | - Davide Vacirca
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Alberto Ranghiero
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy
| | - Dario Trapani
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | - Christian Rolfo
- Center for Thoracic Oncology, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Naples, Italy
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
6
|
Venetis K, Pescia C, Cursano G, Frascarelli C, Mane E, De Camilli E, Munzone E, Dellapasqua S, Criscitiello C, Curigliano G, Guerini Rocco E, Fusco N. The Evolving Role of Genomic Testing in Early Breast Cancer: Implications for Diagnosis, Prognosis, and Therapy. Int J Mol Sci 2024; 25:5717. [PMID: 38891906 PMCID: PMC11172282 DOI: 10.3390/ijms25115717] [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: 04/15/2024] [Revised: 05/13/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Multigene prognostic genomic assays have become indispensable in managing early breast cancer (EBC), offering crucial information for risk stratification and guiding adjuvant treatment strategies in conjunction with traditional clinicopathological parameters. The American Society of Clinical Oncology (ASCO) guidelines endorse these assays, though some clinical contexts still lack definitive recommendations. The dynamic landscape of EBC management demands further refinement and optimization of genomic assays to streamline their incorporation into clinical practice. The breast cancer community is poised at the brink of transformative advances in enhancing the clinical utility of genomic assays, aiming to significantly improve the precision and effectiveness of both diagnosis and treatment for women with EBC. This article methodically examines the testing methodologies, clinical validity and utility, costs, diagnostic frameworks, and methodologies of the established genomic tests, including the Oncotype Dx Breast Recurrence Score®, MammaPrint, Prosigna®, EndoPredict®, and Breast Cancer Index (BCI). Among these tests, Prosigna and EndoPredict® have at present been validated only on a prognostic level, while Oncotype Dx, MammaPrint, and BCI hold both a prognostic and predictive role. Oncologists and pathologists engaged in the management of EBC will find in this review a thorough comparison of available genomic assays, as well as strategies to optimize the utilization of the information derived from them.
Collapse
Affiliation(s)
- Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Carlo Pescia
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- School of Pathology, University of Milan, 20122 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Eltjona Mane
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Elisa De Camilli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
| | - Elisabetta Munzone
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.M.); (S.D.)
| | - Silvia Dellapasqua
- Division of Medical Senology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (E.M.); (S.D.)
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Elena Guerini Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy; (K.V.); (C.P.); (G.C.); (C.F.); (E.M.); (E.D.C.); (E.G.R.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; (C.C.); (G.C.)
| |
Collapse
|
7
|
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
|
8
|
Piga I, Magni F, Smith A. The journey towards clinical adoption of MALDI-MS-based imaging proteomics: from current challenges to future expectations. FEBS Lett 2024; 598:621-634. [PMID: 38140823 DOI: 10.1002/1873-3468.14795] [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: 11/03/2023] [Revised: 12/06/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023]
Abstract
Among the spatial omics techniques available, mass spectrometry imaging (MSI) represents one of the most promising owing to its capability to map the distribution of hundreds of peptides and proteins, as well as other classes of biomolecules, within a complex sample background in a multiplexed and relatively high-throughput manner. In particular, matrix-assisted laser desorption/ionisation (MALDI-MSI) has come to the fore and established itself as the most widely used technique in clinical research. However, the march of this technique towards clinical utility has been hindered by issues related to method reproducibility, appropriate biocomputational tools, and data storage. Notwithstanding these challenges, significant progress has been achieved in recent years regarding multiple facets of the technology and has rendered it more suitable for a possible clinical role. As such, there is now more robust and extensive evidence to suggest that the technology has the potential to support clinical decision-making processes under appropriate circumstances. In this review, we will discuss some of the recent developments that have facilitated this progress and outline some of the more promising clinical proteomics applications which have been developed with a clear goal towards implementation in mind.
Collapse
Affiliation(s)
- Isabella Piga
- Department of Medicine and Surgery, Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, Italy
| | - Fulvio Magni
- Department of Medicine and Surgery, Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, Italy
| | - Andrew Smith
- Department of Medicine and Surgery, Proteomics and Metabolomics Unit, University of Milano-Bicocca, Vedano al Lambro, Italy
| |
Collapse
|
9
|
Ivanova M, Porta FM, D'Ercole M, Pescia C, Sajjadi E, Cursano G, De Camilli E, Pala O, Mazzarol G, Venetis K, Guerini-Rocco E, Curigliano G, Viale G, Fusco N. Standardized pathology report for HER2 testing in compliance with 2023 ASCO/CAP updates and 2023 ESMO consensus statements on HER2-low breast cancer. Virchows Arch 2024; 484:3-14. [PMID: 37770765 PMCID: PMC10791807 DOI: 10.1007/s00428-023-03656-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/03/2023] [Accepted: 09/13/2023] [Indexed: 09/30/2023]
Abstract
Since the release of the DESTINY-Breast04 (DB-04) trial findings in June 2022, the field of pathology has seen a renaissance of HER2 as a predictive biomarker in breast cancer. The trial focused on patients with metastatic breast cancer who were classified as "HER2-low," i.e., those with immunohistochemistry (IHC) HER2 1 + or 2 + and negative in situ hybridization (ISH) results. The study revealed that treating these patients with trastuzumab deruxtecan (T-DXd) instead of the oncologist's chosen chemotherapy led to outstanding improvements in survival. This has challenged the existing binary HER2 pathological classification system, which categorized tumors as either positive (overexpression/amplification) or negative, as per the ASCO/CAP 2018 guideline reaffirmed by ASCO/CAP 2023 guideline update. Given that DB-04 excluded patients with HER2 IHC score 0 status, the results of the ongoing DB-06 trial may shed further light on the potential benefits of T-DXd therapy for these patients. Roughly half of all breast cancers are estimated to belong to the HER2-low category, which does not represent a distinct or specific subtype of cancer. Instead, it encompasses a diverse group of tumors that exhibit clinical, morphological, immunohistochemical, and molecular variations. However, HER2-low offers a distinctive biomarker status that identifies a specific therapeutic regimen (i.e., T-DXd) linked to a favorable prognosis in breast cancer. This unique association emphasizes the importance of accurately identifying these tumors. Differentiating between a HER2 IHC score 0 and score 1 + has not been clinically significant until now. To ensure accurate classification and avoid misdiagnosis, it is necessary to adopt standardized procedures, guidelines, and specialized training for pathologists in interpreting HER2 expression in the lower spectrum. Additionally, the utilization of artificial intelligence holds promise in supporting this endeavor. Here, we address the current state of the art and unresolved issues in assessing HER2-low status, with a particular emphasis on the score 0. We explore the dilemma surrounding the exclusion of HER2-zero patients from potentially beneficial therapy based on traditional HER2 testing. Additionally, we examine the clinical context, considering that DB-04 primarily involved heavily pretreated late-stage metastatic breast cancers. We also delve into emerging evidence suggesting that extrapolating HER2-low status from the original diagnosis may lead to misleading results. Finally, we provide recommendations for conducting high-quality testing and propose a standardized pathology report in compliance with 2023 ASCO/CAP updates and 2023 ESMO consensus statements on HER2-low breast cancer.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Francesca Maria Porta
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Marianna D'Ercole
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Carlo Pescia
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Elisa De Camilli
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Oriana Pala
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Giovanni Mazzarol
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy
- Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Giuseppe Viale
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO European Institute of Oncology IRCCS, 20141, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, 20122, Milan, Italy.
| |
Collapse
|
10
|
Pescia C, Guerini-Rocco E, Viale G, Fusco N. Advances in Early Breast Cancer Risk Profiling: From Histopathology to Molecular Technologies. Cancers (Basel) 2023; 15:5430. [PMID: 38001690 PMCID: PMC10670146 DOI: 10.3390/cancers15225430] [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: 10/15/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/26/2023] Open
Abstract
Early breast cancer (BC) is the definition applied to breast-confined tumors with or without limited involvement of locoregional lymph nodes. While risk stratification is essential for guiding clinical decisions, it can be a complex endeavor in these patients due to the absence of comprehensive guidelines. Histopathological analysis and biomarker assessment play a pivotal role in defining patient outcomes. Traditional histological criteria such as tumor size, lymph node involvement, histological type and grade, lymphovascular invasion, and immune cell infiltration are significant prognostic indicators. In addition to the hormone receptor, HER2, and-in specific scenarios-BRCA1/2 testing, molecular subtyping through gene expression profiling provides valuable insights to tailor clinical decision-making. The emergence of "omics" technologies, applicable to both tissue and liquid biopsy samples, has broadened our arsenal for evaluating the risk of early BC. However, a pressing need remains for standardized methodologies and integrated pathological models that encompass multiple analytical dimensions. In this study, we provide a detailed examination of the existing strategies for early BC risk stratification, intending to serve as a practical guide for histopathologists and molecular pathologists.
Collapse
Affiliation(s)
- Carlo Pescia
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- School of Pathology, University of Milan, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| | - Giuseppe Viale
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
| | - Nicola Fusco
- Division of Pathology, European Institute of Oncology IRCCS, 20141 Milan, Italy; (C.P.); (E.G.-R.); (G.V.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20141 Milan, Italy
| |
Collapse
|
11
|
Fusco N, Ivanova M, Frascarelli C, Criscitiello C, Cerbelli B, Pignataro MG, Pernazza A, Sajjadi E, Venetis K, Cursano G, Pagni F, Di Bella C, Accardo M, Amato M, Amico P, Bartoli C, Bogina G, Bortesi L, Boldorini R, Bruno S, Cabibi D, Caruana P, Dainese E, De Camilli E, Dell'Anna V, Duda L, Emmanuele C, Fanelli GN, Fernandes B, Ferrara G, Gnetti L, Gurrera A, Leone G, Lucci R, Mancini C, Marangi G, Mastropasqua MG, Nibid L, Orrù S, Pastena M, Peresi M, Perracchio L, Santoro A, Vezzosi V, Zambelli C, Zuccalà V, Rizzo A, Costarelli L, Pietribiasi F, Santinelli A, Scatena C, Curigliano G, Guerini-Rocco E, Martini M, Graziano P, Castellano I, d'Amati G. Advancing the PD-L1 CPS test in metastatic TNBC: Insights from pathologists and findings from a nationwide survey. Crit Rev Oncol Hematol 2023; 190:104103. [PMID: 37595344 DOI: 10.1016/j.critrevonc.2023.104103] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/20/2023] Open
Abstract
Pembrolizumab has received approval as a first-line treatment for unresectable/metastatic triple-negative breast cancer (mTNBC) with a PD-L1 combined positive score (CPS) of ≥ 10. However, assessing CPS in mTNBC poses challenges. Firstly, it represents a novel analysis for breast pathologists. Secondly, the heterogeneity of PD-L1 expression in mTNBC further complicates the assessment. Lastly, the lack of standardized assays and staining platforms adds to the complexity. In KEYNOTE trials, PD-L1 expression was evaluated using the IHC 22C3 pharmDx kit as a companion diagnostic test. However, both the 22C3 pharmDx and VENTANA PD-L1 (SP263) assays are validated for CPS assessment. Consequently, assay-platform choice, staining conditions, and scoring methods can significantly impact the testing outcomes. This consensus paper aims to discuss the intricacies of PD-L1 CPS testing in mTNBC and provide practical recommendations for pathologists. Additionally, we present findings from a nationwide Italian survey elucidating the state-of-the-art in PD-L1 CPS testing in mTNBC.
Collapse
Affiliation(s)
- Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Carmen Criscitiello
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Bruna Cerbelli
- Department of Medical-Surgical Sciences and Biotechnologies Sapienza University of Rome, Rome, Italy
| | - Maria Gemma Pignataro
- Department of Medical-Surgical Sciences and Biotechnologies Sapienza University of Rome, Rome, Italy
| | - Angelina Pernazza
- Department of Medical-Surgical Sciences and Biotechnologies Sapienza University of Rome, Rome, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, University Milan Bicocca, Monza (MB), Italy; Department of Pathology, IRCCS San Gerardo Hospital, Monza (MB), Italy
| | - Camillo Di Bella
- Department of Pathology, IRCCS San Gerardo Hospital, Monza (MB), Italy
| | - Marina Accardo
- Pathology Unit, Department of Mental and Physical Health and Preventive Medicine, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Michelina Amato
- Department of Pathology, San Giovanni-Addolorata Hospital, Rome Italy
| | - Paolo Amico
- Department of Pathology, Ospedale Maria Paternò Arezzo, Ragusa, Italy
| | - Caterina Bartoli
- Morphological Diagnostic and Biomolecular Characterization Area, Complex Unit of Pathological Anatomy Empoli-Prato, Oncological Department Azienda USL Toscana Centro, Italy
| | - Giuseppe Bogina
- Pathology Unit, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | - Laura Bortesi
- Pathology Unit, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | - Renzo Boldorini
- Pathology Unit, University of Eastern Piedmont, Novara, Italy
| | - Sara Bruno
- Division of Pathology, ASL2 Savona, Liguria, Italy
| | - Daniela Cabibi
- Department of Sciences for the Promotion of Health and Mother and Child Care, Anatomic Pathology, University of Palermo, Palermo, Italy
| | - Pietro Caruana
- Pathology Unit, Department of Medicine and Surgery, University Hospital of Parma, Parma, Italy
| | - Emanuele Dainese
- Surgical Pathology Division, Department of Oncology, ASST Lecco, "A. Manzoni" Hospital, Lecco, Italy
| | - Elisa De Camilli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Loren Duda
- Department of Clinical and Experimental Medicine, Pathology Unit, University of Foggia, Foggia, Italy
| | - Carmela Emmanuele
- Division of Pathology, Umberto I Hospital Presidium, Enna Provincial Health Department (ASP), Enna, Italy
| | - Giuseppe Nicolò Fanelli
- Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | | | - Gerardo Ferrara
- Department of Anatomic Pathology and Cytopathology, G. Pascale National Cancer Institute Foundation (IRCCS) Naples, Italy
| | - Letizia Gnetti
- Division of Pathology, Umberto I Hospital Presidium, Enna Provincial Health Department (ASP), Enna, Italy
| | | | - Giorgia Leone
- Division of Pathology, Clinical Institute Humanitas Catania Cubba, Misterbianco (Catania), Italy
| | - Raffaella Lucci
- Pathology Unit, Monaldi Hospital, A.O. dei Colli of Naples, Naples, Italy
| | - Cristina Mancini
- Division of Pathology, Umberto I Hospital Presidium, Enna Provincial Health Department (ASP), Enna, Italy
| | - Grazia Marangi
- Anatomic Pathology Unit, SS. Annunziata Hospital, Taranto, Italy
| | - Mauro G Mastropasqua
- Department of Precision and Regenerative Medicine and Jonian Area, School of Medicine, University of Bari "Aldo Moro", Bari, Italy
| | - Lorenzo Nibid
- Research Unit of Anatomical Pathology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy; Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Via Alvaro del Portillo, Rome, Italy
| | - Sandra Orrù
- Businco Oncologic Hospital, ARNAS Brotzu, Cagliari, Italy
| | - Maria Pastena
- IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Monica Peresi
- Pathology and Cytopathology Diagnostic Unit, Ospedale Villa Scassi di Genova, Genoa, Italy
| | - Letizia Perracchio
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Angela Santoro
- General Pathology Unit, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Vania Vezzosi
- Histopathology and Molecular Diagnostics Unit, Careggi Hospital, Firenze, Italy
| | | | - Valeria Zuccalà
- Pathology Unit, Pugliese-Ciaccio Hospital Catanzaro, Catanzaro, Italy
| | - Antonio Rizzo
- Division of Pathology, Clinical Institute Humanitas Catania Cubba, Misterbianco (Catania), Italy
| | | | | | - Alfredo Santinelli
- Anatomic Pathology, Azienda Sanitaria Territoriale di Pesaro-Urbino, Pesaro, Italy
| | - Cristian Scatena
- Division of Pathology, Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy; Division of New Drugs and Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - 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
| | - Maurizio Martini
- Department of Human and Developmental Pathology, University of Messina, Messina, Italy
| | - Paolo Graziano
- Pathology Unit, Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy
| | | | - Giulia d'Amati
- Department of Medical-Surgical Sciences and Biotechnologies Sapienza University of Rome, Rome, Italy
| |
Collapse
|
12
|
Sajjadi E, Frascarelli C, Venetis K, Bonizzi G, Ivanova M, Vago G, Guerini-Rocco E, Fusco N. Computational pathology to improve biomarker testing in breast cancer: how close are we? Eur J Cancer Prev 2023; 32:460-467. [PMID: 37038997 DOI: 10.1097/cej.0000000000000804] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/12/2023]
Abstract
The recent advancements in breast cancer precision medicine have highlighted the urgency for the precise and reproducible characterization of clinically actionable biomarkers. Despite numerous standardization efforts, biomarker testing by conventional methodologies is challenged by several issues such as high inter-observer variabilities, the spatial heterogeneity of biomarkers expression, and technological heterogeneity. In this respect, artificial intelligence-based digital pathology approaches are being increasingly recognized as promising methods for biomarker testing and subsequently improved clinical management. Here, we provide an overview on the most recent advances for artificial intelligence-assisted biomarkers testing in breast cancer, with a particular focus on tumor-infiltrating lymphocytes, programmed death-ligand 1, phosphatidylinositol-3 kinase catalytic alpha, and estrogen receptor 1. Challenges and solutions for this integrative analysis in pathology laboratories are also provided.
Collapse
Affiliation(s)
- Elham Sajjadi
- Department of Oncology and Hemato-Oncology, University of Milan
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Frascarelli
- Department of Oncology and Hemato-Oncology, University of Milan
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Giuseppina Bonizzi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Gianluca Vago
- Department of Oncology and Hemato-Oncology, University of Milan
| | - Elena Guerini-Rocco
- Department of Oncology and Hemato-Oncology, University of Milan
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| |
Collapse
|
13
|
Porta FM, Sajjadi E, Venetis K, Frascarelli C, Cursano G, Guerini-Rocco E, Fusco N, Ivanova M. Immune Biomarkers in Triple-Negative Breast Cancer: Improving the Predictivity of Current Testing Methods. J Pers Med 2023; 13:1176. [PMID: 37511789 PMCID: PMC10381494 DOI: 10.3390/jpm13071176] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 07/17/2023] [Accepted: 07/20/2023] [Indexed: 07/30/2023] Open
Abstract
Triple-negative breast cancer (TNBC) poses a significant challenge in terms of prognosis and disease recurrence. The limited treatment options and the development of resistance to chemotherapy make it particularly difficult to manage these patients. However, recent research has been shifting its focus towards biomarker-based approaches for TNBC, with a particular emphasis on the tumor immune landscape. Immune biomarkers in TNBC are now a subject of great interest due to the presence of tumor-infiltrating lymphocytes (TILs) in these tumors. This characteristic often coincides with the presence of PD-L1 expression on both neoplastic cells and immune cells within the tumor microenvironment. Furthermore, a subset of TNBC harbor mismatch repair deficient (dMMR) TNBC, which is frequently accompanied by microsatellite instability (MSI). All of these immune biomarkers hold actionable potential for guiding patient selection in immunotherapy. To fully capitalize on these opportunities, the identification of additional or complementary biomarkers and the implementation of highly customized testing strategies are of paramount importance in TNBC. In this regard, this article aims to provide an overview of the current state of the art in immune-related biomarkers for TNBC. Specifically, it focuses on the various testing methodologies available and sheds light on the immediate future perspectives for patient selection. By delving into the advancements made in understanding the immune landscape of TNBC, this study aims to contribute to the growing body of knowledge in the field. The ultimate goal is to pave the way for the development of more personalized testing strategies, ultimately improving outcomes for TNBC patients.
Collapse
Affiliation(s)
- Francesca Maria Porta
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, 20122 Milan, Italy
| |
Collapse
|
14
|
Piga I, L'Imperio V, Capitoli G, Denti V, Smith A, Magni F, Pagni F. Paving the path toward multi-omics approaches in the diagnostic challenges faced in thyroid pathology. Expert Rev Proteomics 2023; 20:419-437. [PMID: 38000782 DOI: 10.1080/14789450.2023.2288222] [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: 09/12/2023] [Accepted: 11/22/2023] [Indexed: 11/26/2023]
Abstract
INTRODUCTION Despite advancements in diagnostic methods, the classification of indeterminate thyroid nodules still poses diagnostic challenges not only in pre-surgical evaluation but even after histological evaluation of surgical specimens. Proteomics, aided by mass spectrometry and integrated with artificial intelligence and machine learning algorithms, shows great promise in identifying diagnostic markers for thyroid lesions. AREAS COVERED This review provides in-depth exploration of how proteomics has contributed to the understanding of thyroid pathology. It discusses the technical advancements related to immunohistochemistry, genetic and proteomic techniques, such as mass spectrometry, which have greatly improved sensitivity and spatial resolution up to single-cell level. These improvements allowed the identification of specific protein signatures associated with different types of thyroid lesions. EXPERT COMMENTARY Among all the proteomics approaches, spatial proteomics stands out due to its unique ability to capture the spatial context of proteins in both cytological and tissue thyroid samples. The integration of multi-layers of molecular information combining spatial proteomics, genomics, immunohistochemistry or metabolomics and the implementation of artificial intelligence and machine learning approaches, represent hugely promising steps forward toward the possibility to uncover intricate relationships and interactions among various molecular components, providing a complete picture of the biological landscape whilst fostering thyroid nodule diagnosis.
Collapse
Affiliation(s)
- Isabella Piga
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano - Bicocca, Monza, Italy
| | - Vincenzo L'Imperio
- Department of Medicine and Surgery, Pathology, Fondazione IRCCS San Gerardo dei Tintori, University of Milan-Bicocca, Monza, Italy
| | - Giulia Capitoli
- Department of Medicine and Surgery, Bicocca Bioinformatics Biostatistics and Bioimaging B4 Center, University of Milan - Bicocca (UNIMIB), Monza, Italy
| | - Vanna Denti
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano - Bicocca, Monza, Italy
| | - Andrew Smith
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano - Bicocca, Monza, Italy
| | - Fulvio Magni
- Department of Medicine and Surgery, Clinical Proteomics and Metabolomics Unit, University of Milano - Bicocca, Monza, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, Fondazione IRCCS San Gerardo dei Tintori, University of Milan-Bicocca, Monza, Italy
| |
Collapse
|
15
|
Malapelle U, Angerilli V, Pepe F, Fontanini G, Lonardi S, Scartozzi M, Memeo L, Pruneri G, Marchetti A, Perrone G, Fassan M. The ideal reporting of RAS testing in colorectal adenocarcinoma: a pathologists' perspective. Pathologica 2023; 115:1-11. [PMID: 37314870 PMCID: PMC10462993 DOI: 10.32074/1591-951x-895] [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: 05/24/2023] [Accepted: 05/24/2023] [Indexed: 06/16/2023] Open
Abstract
RAS gene mutational status represents an imperative predictive biomarker to be tested in the clinical management of metastatic colorectal adenocarcinoma. Even if it is one of the most studied biomarkers in the era of precision medicine, several pre-analytical and analytical factors may still impasse an adequate reporting of RAS status in clinical practice, with significant therapeutic consequences. Thus, pathologists should be aware on the main topics related to this molecular evaluation: (i) adopt diagnostic limit of detections adequate to avoid the interference of sub-clonal cancer cell populations; (ii) choose the most adequate diagnostic strategy according to the available sample and its qualification for molecular testing; (iii) provide all the information regarding the mutation detected, since many RAS mutation-specific targeted therapeutic approaches are in development and will enter into routine clinical practice. In this review, we give a comprehensive description of the current scenario about RAS gene mutational testing in the clinic focusing on the pathologist's role in patient selection for targeted therapies.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples (NA), Italy
| | | | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples (NA), Italy
| | - Gabriella Fontanini
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa (PI), Italy
| | - Sara Lonardi
- Medical Oncology 3, Veneto Institute of Oncology IOV-IRCCS, Padua (PD), Italy
| | - Mario Scartozzi
- Medical Oncology, University Hospital and University of Cagliari, Cagliari (CA), Italy
| | - Lorenzo Memeo
- Department of Experimental Oncology, Mediterranean Institute of Oncology, Viagrande, Catania (CT), Italy
| | - Gianfranco Pruneri
- Department of Advanced Diagnostics, Fondazione IRCCS Istituto Nazionale Tumori and University of Milan, Milan (MI), Italy
| | - Antonio Marchetti
- Center for Advanced Studies and Technology (CAST), University Chieti-Pescara, Chieti (CH), Italy
- Diagnostic Molecular Pathology, Unit of Anatomic Pathology, SS Annunziata Hospital, Chieti (CH), Italy and Department of Medical, Oral, and Biotechnological Sciences University “G. D’Annunzio” of Chieti-Pescara, Chieti (CH), Italy
| | - Giuseppe Perrone
- Department of Medicine and Surgery, Research Unit of Anatomical Pathology, Università Campus Bio-Medico di Roma, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), University of Padua, Padua (PD), Italy
- Veneto Institute of Oncology (IOV-IRCCS), Padua (PD), Italy
| |
Collapse
|
16
|
Lazure P, Sireci A, Subbiah V, Murray S, Grohé C, Sherman SI, Kelly E, Bubach P, Péloquin S. Challenges in diagnosis and biomarker testing for RET-altered lung and thyroid cancer care: an international mixed-method study. BMC MEDICAL EDUCATION 2023; 23:410. [PMID: 37277734 DOI: 10.1186/s12909-023-04396-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 05/25/2023] [Indexed: 06/07/2023]
Abstract
BACKGROUND The introduction of new targeted therapies for RET-altered lung and thyroid cancers (LC/TC) has impacted pathologists' practice by making genomic testing more relevant. Variations in health systems and treatment access result in distinct clinical challenges and barriers. This study aimed to assess practice gaps and challenges experienced by pathologists involved in the diagnosis of RET-altered LC/TC, including biomarker testing, to inform educational solutions. METHODS Pathologists in Germany, Japan, the UK, and US participated in this ethics-approved mixed-methods study, which included interviews and surveys (data collected January-March 2020). Qualitative data was thematically analysed, quantitative data was analysed with chi-square and Kruskal-Wallis H-tests, and both were triangulated. RESULTS A total of 107 pathologists took part in this study. Knowledge gaps were reported regarding genomic testing for LC/TC in Japan (79/60%), the UK (73/66%), and the US (53/30%). Skill gaps were reported when selecting genomic biomarker tests to diagnose TC in Japan (79%), the UK (73%) and US (57%) and when performing specific biomarker tests, especially in Japan (82% for RET) and in the UK (75% for RET). Japanese participants (80%) reported uncertainty about what information to share with the multidisciplinary team to ensure optimal patient-centered care. At the time of data collection, pathologists in Japan faced access barriers to using RET biomarker tests: only 28% agreed that there are relevant RET genomic biomarker tests available in Japan, versus 67% to 90% in other countries. CONCLUSIONS This study identified areas where pathologists need additional continuing professional development opportunities to enhance their competencies and better support delivery of care to patients with RET-altered lung or thyroid tumours. Addressing identified gaps and improving competencies of pathologists in this field should be emphasised in continuing medical education curricula and through quality improvement initiatives. Strategies deployed on an institutional and health system level should aim to improve interprofessional communication and genetic biomarker testing expertise.
Collapse
Affiliation(s)
- Patrice Lazure
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada.
| | | | - Vivek Subbiah
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suzanne Murray
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada
| | | | - Steven I Sherman
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | | | - Sophie Péloquin
- AXDEV Group Inc., 8, Place du Commerce, Suite 210, Brossard, Québec, J4W 3H2, Canada
| |
Collapse
|
17
|
Caputo A, L’Imperio V, Merolla F, Girolami I, Leoni E, Mea VD, Pagni F, Fraggetta F. The slow-paced digital evolution of pathology: lights and shadows from a multifaceted board. Pathologica 2023; 115:127-136. [PMID: 37387439 PMCID: PMC10462988 DOI: 10.32074/1591-951x-868] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 07/01/2023] Open
Abstract
Objective The digital revolution in pathology represents an invaluable resource fto optimise costs, reduce the risk of error and improve patient care, even though it is still adopted in a minority of laboratories. Barriers include concerns about initial costs, lack of confidence in using whole slide images for primary diagnosis, and lack of guidance on transition. To address these challenges and develop a programme to facilitate the introduction of digital pathology (DP) in Italian pathology departments, a panel discussion was set up to identify the key points to be considered. Methods On 21 July 2022, an initial conference call was held on Zoom to identify the main issues to be discussed during the face-to-face meeting. The final summit was divided into four different sessions: (I) the definition of DP, (II) practical applications of DP, (III) the use of AI in DP, (IV) DP and education. Results Essential requirements for the implementation of DP are a fully tracked and automated workflow, selection of the appropriate scanner based on the specific needs of each department, and a strong commitment combined with coordinated teamwork (pathologists, technicians, biologists, IT service and industries). This could reduce human error, leading to the application of AI tools for diagnosis, prognosis and prediction. Open challenges are the lack of specific regulations for virtual slide storage and the optimal storage solution for large volumes of slides. Conclusion Teamwork is key to DP transition, including close collaboration with industry. This will ease the transition and help bridge the gap that currently exists between many labs and full digitisation. The ultimate goal is to improve patient care.
Collapse
Affiliation(s)
- Alessandro Caputo
- Department of Pathology, Ruggi University Hospital, Salerno, Italy
- Pathology Unit, Gravina Hospital Caltagirone ASP, Catania, Italy
| | - Vincenzo L’Imperio
- Department of Medicine and Surgery, Pathology, University of Milan-Bicocca, IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
| | - Francesco Merolla
- Department of Medicine and Health Sciences “V. Tiberio”, University of Molise, Campobasso, Italy
| | - Ilaria Girolami
- Department of Pathology, Provincial Hospital of Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy; Lehrkrankenhaus der Paracelsus Medizinischen Privatuniversität
| | - Eleonora Leoni
- Pathology Unit, Busto Arsizio Hospital, Busto Arsizio, Italy
| | - Vincenzo Della Mea
- Department of Mathematics, Computer Science and Physics, University of Udine, Udine, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, University of Milan-Bicocca, IRCCS Fondazione San Gerardo dei Tintori, Monza, Italy
| | | |
Collapse
|
18
|
Angerilli V, Fornaro L, Pepe F, Rossi SM, Perrone G, Malapelle U, Fassan M. FGFR2 testing in cholangiocarcinoma: translating molecular studies into clinical practice. Pathologica 2023; 115:71-82. [PMID: 37017301 PMCID: PMC10462997 DOI: 10.32074/1591-951x-859] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 03/06/2023] [Indexed: 04/06/2023] Open
Abstract
Cholangiocarcinoma (CCA) is a heterogeneous group of neoplasms burdened by a dismal prognosis. Several studies have investigated the genomic profile of CCA and identified numerous druggable genetic alterations, including FGFR2 fusions/rearrangements. Approximately 5-7% of CCAs and 10-20% of intrahepatic iCCAs harbor FGFR2 fusions. With the recent advent of FGFR-targeting therapies into clinical practice, a standardization of molecular testing for FGFR2 alterations in CCA will be necessary. In this review, we describe the technical aspects and challenges related to FGFR2 testing in routine practice, focusing on the comparison between Next-Generation Sequencing (NGS) and FISH assays, the best timing to perform the test, and on the role of liquid biopsy.
Collapse
Affiliation(s)
- Valentina Angerilli
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua (PD), Italy
| | - Lorenzo Fornaro
- Medical Oncology, Azienda Ospedaliero-Universitaria Pisana, Pisa (PI), Italy
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, Naples (NA), Italy
| | - Silvia Maria Rossi
- Department of Medicine and Surgery, Research Unit of Anatomical Pathology, Università Campus Bio-Medico di Roma, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Giuseppe Perrone
- Department of Medicine and Surgery, Research Unit of Anatomical Pathology, Università Campus Bio-Medico di Roma, Roma, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples (NA), Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University of Padua, Padua (PD), Italy
- Veneto Institute of Oncology, IOV - IRCCS, Padua (PD), Italy
| |
Collapse
|
19
|
Iaccarino A, Nacchio M, Acanfora G, Pisapia P, Malapelle U, Bellevicine C, Troncone G, Vigliar E. Multiple predictive biomarker testing in melanoma: Another challenge in identifying the optimal approach on cytological samples. Cytopathology 2023; 34:198-203. [PMID: 36658094 DOI: 10.1111/cyt.13211] [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: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 01/21/2023]
Abstract
BACKGROUND The management of cutaneous melanoma has changed dramatically in recent years thanks to the development of tyrosine kinase and immune-checkpoint inhibitors (ICIs). Thus, multiple biomarker testing is becoming ever more important for the identification of patients who are potentially eligible for these treatments. One reliable approach to the molecular evaluation of metastatic melanoma is fine needle cytology (FNC). To examine the utility of this approach for assessing PD-L1 expression levels, we evaluated the cellular adequacy of residual cell block (CB) material from metastatic melanomas that were previously tested for BRAF and NRAS mutations. METHODS We retrieved from our internal archives a series of FNC samples of metastatic melanoma that had been subjected to molecular testing on residual CB material or a dedicated needle rinse between January 2016 and July 2022. Real-time polymerase chain reaction was used to assess BRAF and NRAS status, and an SP263 assay was employed to ascertain PD-L1 expression levels. RESULTS Overall, n = 19 cases were selected. Of these, 11 (57.9%) cases revealed a BRAF exon 15 p.V600E mutation, one case (5.3%) revealed NRAS mutation, and seven cases (36.8%) showed no mutations. Regarding PD-L1 assessment, 16/19 (84.2%) cases were deemed adequate, meaning they contained at least 100 viable cells. CONCLUSIONS We highlighted the feasibility of assessing PD-L1 expression levels in residual CB material from metastatic melanomas previously tested for BRAF and NRAS mutations. Moreover, we pointed out that FNC needle rinses may be an alternative source of nucleic acids for molecular testing, preserving CB material for immunocytochemistry evaluation.
Collapse
Affiliation(s)
- Antonino Iaccarino
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Mariantonia Nacchio
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Gennaro Acanfora
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Pasquale Pisapia
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Claudio Bellevicine
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| | - Elena Vigliar
- Department of Public Health, University of Naples "Federico II", Naples, Italy
| |
Collapse
|
20
|
Fusco N, Malapelle U, Criscitiello C. Editorial: Diagnosis and Treatment of Breast Cancer in 2022: The Rise of Novel Molecular Biomarkers. Front Mol Biosci 2023; 9:1117323. [PMID: 36660432 PMCID: PMC9845888 DOI: 10.3389/fmolb.2022.1117323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 12/16/2022] [Indexed: 01/06/2023] Open
Affiliation(s)
- Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy,Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy,*Correspondence: Nicola Fusco, ; Umberto Malapelle, ; Carmen Criscitiello,
| | - Umberto Malapelle
- Department of Public Health, Federico II University of Naples, Naples, Italy,*Correspondence: Nicola Fusco, ; Umberto Malapelle, ; Carmen Criscitiello,
| | - Carmen Criscitiello
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy,Division of Early Drug Development for Innovative Therapy, IEO, European Institute of Oncology, Milan, Italy,*Correspondence: Nicola Fusco, ; Umberto Malapelle, ; Carmen Criscitiello,
| |
Collapse
|
21
|
Malapelle U, Borralho P, Wang L, Schmitt F. Editorial: Cancer diagnostics in solid tumors-from pathology to precision oncology. Front Mol Biosci 2023; 10:1150641. [PMID: 36895807 PMCID: PMC9990903 DOI: 10.3389/fmolb.2023.1150641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Pedro Borralho
- Novartis Farma - Produtos Farmacêuticos, Porto Salvo, Portugal
| | - Liang Wang
- Department of Tumor Biology, Moffitt Cancer Center & Research Institute Tampa, Tampa, FL, United States
| | - Fernando Schmitt
- Department of Pathology, Faculty of Medicine of University of Porto, Porto, Portugal
| |
Collapse
|
22
|
Ivanova M, Venetis K, Guerini-Rocco E, Bottiglieri L, Mastropasqua MG, Garrone O, Fusco N, Ghidini M. HER2 in Metastatic Colorectal Cancer: Pathology, Somatic Alterations, and Perspectives for Novel Therapeutic Schemes. Life (Basel) 2022; 12:1403. [PMID: 36143438 PMCID: PMC9502498 DOI: 10.3390/life12091403] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 12/03/2022] Open
Abstract
HER2 is an emerging biomarker in colorectal cancer (CRC). This oncogene plays an essential role in regulating cell proliferation, differentiation, migration, and, more in general, tumorigenesis and tumor progression. The most frequent types of HER2 alterations in CRC include gene amplification and missense mutations in 7-8% of CRC, often being mirrored by HER2 protein overexpression, representing founder events in solid tumors, including CRC. There are currently no approved HER2-targeted therapy guidelines for CRC; however, several studies have shown that HER2 can be effectively targeted in meta-static CRC settings. In this review, we discuss the current knowledge of HER2 testing in CRC and the immediate future perspectives for HER2 targeting in the metastatic setting.
Collapse
Affiliation(s)
- Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Luca Bottiglieri
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Mauro Giuseppe Mastropasqua
- Department of Emergency and Organ Transplantation, School of Medicine, University of Bari “Aldo Moro”, Piazza G Cesare, 11, 70124 Bari, Italy
| | - Ornella Garrone
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141 Milan, Italy
| | - Michele Ghidini
- Medical Oncology Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy
| |
Collapse
|