1
|
Ende TVD, Kuijper SC, Widaatalla Y, Noortman WA, van Velden FHP, Woodruff HC, van der Pol Y, Moldovan N, Pegtel DM, Derks S, Bijlsma MF, Mouliere F, de Geus-Oei LF, Lambin P, van Laarhoven HWM. Integrating Clinical Variables, Radiomics, and Tumor-derived Cell-Free DNA for Enhanced Prediction of Resectable Esophageal Adenocarcinoma Outcomes. Int J Radiat Oncol Biol Phys 2025; 121:963-974. [PMID: 39424077 DOI: 10.1016/j.ijrobp.2024.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 09/13/2024] [Accepted: 10/06/2024] [Indexed: 10/21/2024]
Abstract
PURPOSE The value of integrating clinical variables, radiomics, and tumor-derived cell-free DNA (cfDNA) for the prediction of survival and response to chemoradiation of patients with resectable esophageal adenocarcinoma is not yet known. Our aim was to investigate if radiomics and cfDNA metrics combined with clinical variables can improve personalized predictions. METHODS AND MATERIALS A cohort of 111 patients with resectable esophageal adenocarcinoma from 2 centers treated with neoadjuvant chemoradiation therapy was used for exploratory retrospective analyses. Models combining the clinical variables of the SOURCE survival model with radiomic features and cfDNA were built using elastic net regression and internally validated using 5-fold cross-validation. Model performance for overall survival (OS) and time to progression (TTP) were evaluated with the C-index and the area under the curve for pathologic complete response. RESULTS The best-performing baseline models for OS and TTP were based on the combination of SOURCE-cfDNA that reached a C-index of 0.55 and 0.59 compared with 0.44 to 0.45 with SOURCE alone. The addition of restaging positron emission tomography radiomics to SOURCE was the most promising addition for predicting OS (C-index: 0.65) and TTP (C-index: 0.60). Baseline risk stratification was achieved for OS and TTP by combining SOURCE with radiomics or cfDNA, log-rank P < .01. The best-performing combination model for the prediction of pathologic complete response reached an area under the curve of 0.61 compared with 0.47 with SOURCE variables alone. CONCLUSIONS The addition of radiomics and cfDNA can improve the performance of an established survival model. External validity needs to be further assessed in future studies together with the optimization of radiomic pipelines.
Collapse
Affiliation(s)
- Tom van den Ende
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Steven C Kuijper
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands
| | - Yousif Widaatalla
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Wyanne A Noortman
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands; TechMed Centre, University of Twente, Enschede, The Netherlands
| | - Floris H P van Velden
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - Henry C Woodruff
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Ymke van der Pol
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Norbert Moldovan
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - D Michiel Pegtel
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Sarah Derks
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands
| | - Maarten F Bijlsma
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Oncode Institute, Utrecht, The Netherlands; Laboratory for Experimental Oncology and Radiobiology, Center for Experimental and Molecular Medicine, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Florent Mouliere
- Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands; Department of Pathology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lioe-Fee de Geus-Oei
- Department of Radiology, Section of Nuclear Medicine, Leiden University Medical Center, Leiden, The Netherlands; TechMed Centre, University of Twente, Enschede, The Netherlands; Department of Radiation Science & Technology, Delft University of Technology, Delft., The Netherlands
| | - Philippe Lambin
- The D-Lab, Department of Precision Medicine, GROW-School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands; Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Hanneke W M van Laarhoven
- Department of Medical Oncology, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands; Cancer Center Amsterdam, Imaging and Biomarkers, Amsterdam, The Netherlands.
| |
Collapse
|
2
|
Wang Q, Chen J, Wang Y, Li X, Ping X, Shen J, Yang S, Shen L. The profiles of immunosuppressive microenvironment in the Lauren intestinal-type gastric adenocarcinoma. Cancer Immunol Immunother 2025; 74:82. [PMID: 39891785 PMCID: PMC11787096 DOI: 10.1007/s00262-024-03938-5] [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: 11/22/2024] [Accepted: 12/30/2024] [Indexed: 02/03/2025]
Abstract
BACKGROUND Gastric adenocarcinoma (GAC), particularly the Lauren intestinal-type GAC (IGAC), leads to significant mortality in China due to the limited effectiveness of current treatments. This study aims to investigate the mechanisms of immune suppression in IGAC to identify potential targets for enhancing immunotherapy outcomes. METHODS Performing an extensive collection and re-analysis of single-cell RNA sequencing (scRNA-seq) of tumor tissues and the corresponding noncancerous mucosae from 15 Chinese patients diagnosed with IGAC, we identified cell subpopulations involved in immune suppression within the tumor microenvironment (TME). We further validated our findings using spatially resolved transcriptomics (SRT), immunofluorescence (IF), and flow cytometry (FCM) on tissues from IGAC patients. RESULTS We demonstrated that the TME of IGAC harbors CD8+ exhausted T cells (Texs) and various subtypes that mediate immunity. We identified specific subpopulations of Texs (HAVCR2+VCAM1+) and regulatory T cells (Tregs) (LAYN+TNFRSF4+) contributing to immune suppression. Furthermore, TNFRSF12A+ cancer-associated fibroblasts (CAFs), CTSB+ macrophages, and SOD2+ monocytes were found to be involved in maintaining the immunosuppressive milieu. SRT and IF assays confirmed the presence and colocalization of these cell types within the tumor tissues, highlighting their functional interactions. FCM assays indicated that the prevalence of HAVCR2+VCAM1+ Texs and LAYN+TNFRSF4+ Tregs in tumor tissues was positively associated with IGAC progression. CONCLUSIONS Detailed profiles of immunosuppressive cell subpopulations in IGAC provide valuable insights into the complexity and heterogeneity of immunosuppression. These findings underscore the necessity for targeted strategies that disrupt specific immunosuppressive pathways, potentially enhancing the efficacy of immunotherapeutic interventions in IGAC.
Collapse
Affiliation(s)
- Qingyuan Wang
- Departemtn of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jia Chen
- Departemtn of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
- Department of General Surgery, Northern Jiangsu People's Hospital, Yangzhou, 225001, Jiangsu, China
| | - Yaohui Wang
- Department of Pathology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Xiang Li
- Department of Surgical Oncology, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210029, Jiangsu, China
| | - Xiaochun Ping
- Departemtn of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jiajia Shen
- Departemtn of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| | - Sheng Yang
- Department of Biostatistics, Center for Global Health, School of Public Health, Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
| | - Lizong Shen
- Departemtn of General Surgery, the First Affiliated Hospital, Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
3
|
Hill SL, Sugiyarto G, Harrington J, James E, Underwood TJ, Elliott T. High proportion of PD-1 and CD39 positive CD8+ tissue resident T lymphocytes correlates with better clinical outcome in resected human oesophageal adenocarcinoma. Cancer Immunol Immunother 2024; 73:213. [PMID: 39235606 PMCID: PMC11377377 DOI: 10.1007/s00262-024-03799-y] [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: 04/05/2024] [Accepted: 08/01/2024] [Indexed: 09/06/2024]
Abstract
OBJECTIVE To understand the CD8+ tumour infiltrating lymphocyte (TIL) compartment of oesophageal adenocarcinoma (OAC) with regards to markers of lymphocyte exhaustion, tissue residency and to identify possible reasons behind differential responses to therapy. DESIGN Tumour samples from 44 patients undergoing curative resection for OAC were assessed by flow cytometry for presence of antigen-experienced TILs and markers of activation and exhaustion. Populations of PD-1 and CD39 positive OAC TILs were sorted, and bulk RNA sequencing undertaken using a modified SmartSeq2 protocol. Flow cytometric assessment of functionality was completed. RESULTS A higher proportion of antigen experienced CD8+ OAC TILs was associated with improved survival following surgery; while, high double positivity (DP) for PD-1 and CD39 among these TILs also correlated significantly with outcome. These DP TILs possess a minority population which is positive for the markers of exhaustion TIM3 and LAG3. Transcriptomic assessment of the PD-1 and CD39 DP TILs demonstrated enrichment for a tissue resident memory T lymphocyte (TRM) phenotype associated with improved survival in other cancers, reinforced by positivity for the canonical TRM marker CD103 by flow cytometry. This population demonstrated maintained functional capacity both in their transcriptomic profile, and on flow cytometric assessment, as well as preserved proliferative capacity. CONCLUSION Resected OAC are variably infiltrated by PD-1 and CD39 DP TILs, an abundance of which among lymphocytes is associated with improved survival. This DP population has an increased, but still modest, frequency of TIM3 and LAG3 positivity compared to DN, and is in keeping with a functionally competent TRM phenotype.
Collapse
Affiliation(s)
- Samuel L Hill
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
| | - Gessa Sugiyarto
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Jack Harrington
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Edward James
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Timothy J Underwood
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Tim Elliott
- Oxford Cancer Centre for Immuno-Oncology and CAMS-Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
4
|
He P, Ma L, Xu B, Wang Y, Li X, Chen H, Li Y. Research progress and future directions of immune checkpoint inhibitor combination therapy in advanced gastric cancer. Ther Adv Med Oncol 2024; 16:17588359241266156. [PMID: 39091604 PMCID: PMC11292724 DOI: 10.1177/17588359241266156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 06/18/2024] [Indexed: 08/04/2024] Open
Abstract
In recent years, with the continuous development of molecular immunology, immune checkpoint inhibitors (ICIs) have also been widely used in the treatment of gastric cancer, but they still face some challenges: The first is that only some people can benefit, the second is the treatment-related adverse events (TRAEs) that occur during treatment, and the third is the emergence of varying degrees of drug resistance with long-term use. How to overcome these challenges, combined therapy based on ICIs has become one of the important strategies. This article summarizes the clinical application of ICIs combined with chemotherapy, targeted therapy, radiotherapy, photodynamic therapy, thermotherapy, immune adjuvant, and dual immunotherapy and discusses the mechanism, and also summarizes the advantages and disadvantages of the current combination modalities and the potential research value. The aim of this study is to provide more and more optimized combination regimen for ICI combined therapy in patients with advanced gastric cancer and to provide reference for clinical and scientific research.
Collapse
Affiliation(s)
- Puyi He
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Long Ma
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Bo Xu
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Yunpeng Wang
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Xiaomei Li
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, China
| | - Hao Chen
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
- No. 82, Cuiyingmen, Chengguan, Lanzhou 730030, China
| | - Yumin Li
- The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Key Laboratory of Environmental Oncology, Lanzhou, China
- No. 82, Cuiyingmen, Chengguan, Lanzhou 730030, China
| |
Collapse
|
5
|
Prenen H, Deva S, Keam B, Lindsay CR, Lugowska I, Yang JC, Longo F, de Miguel M, Ponz-Sarvise M, Ahn MJ, Gumus M, Champiat S, Italiano A, Salas S, Perets R, Arslan C, Cho BC, Evers S, Boetsch C, Marbach D, Dejardin D, Sleiman N, Ardeshir C, Richard M, Charo J, Kraxner A, Keshelava N, Teichgräber V, Moreno V. Phase II Study to Determine the Antitumor Activity and Safety of Simlukafusp Alfa (FAP-IL2v) Combined with Atezolizumab in Esophageal Cancer. Clin Cancer Res 2024; 30:2945-2953. [PMID: 38709220 DOI: 10.1158/1078-0432.ccr-23-2677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/29/2024] [Accepted: 05/01/2024] [Indexed: 05/07/2024]
Abstract
PURPOSE In this study, we report the results from the esophageal squamous cell carcinoma (SCC) cohort of a phase II, noncomparative, basket study evaluating the antitumor activity and safety of fibroblast activation protein-IL2 variant (FAP-IL2v) plus atezolizumab in patients with advanced/metastatic solid tumors (NCT03386721). PATIENTS AND METHODS Eligible patients had an Eastern Cooperative Oncology Group performance status of 0 to 1; measurable metastatic, persistent, or recurrent esophageal SCC; progression on ≥1 prior therapy; and were checkpoint inhibitor-naïve. Patients received FAP-IL2v 10 mg plus atezolizumab 1,200 mg intravenously every 3 weeks, or FAP-IL2v weekly for 4 weeks and then every 2 weeks plus atezolizumab 840 mg intravenously every 2 weeks. The primary endpoint was investigator-assessed objective response rate (ORR). RESULTS In the response-evaluable population (N = 34), the best confirmed ORR was 20.6% [95% confidence interval (CI), 10.4-36.8], with a complete response seen in 1 patient and partial responses in 6 patients. The disease control rate was 44.1% (complete response = 2.9%; partial response = 17.6%; stable disease = 23.5%), and the median duration of response was 10.1 mon/ths (95% CI, 5.6-26.7). The median progression-free survival was 1.9 months (95% CI, 1.8-3.7). Analysis of response by PDL1 expression (Ventana SP263) resulted in an ORR of 26.7% for patients with PDL1-positive tumors (tumor area positivity cutoff ≥1%; n = 15) and 7.1% for patients with PDL1-negative tumors (tumor area positivity cutoff <1%; n = 14). Overall, the treatment combination was tolerable, and adverse events were consistent with the known safety profiles of each drug. CONCLUSIONS FAP-IL2v plus atezolizumab demonstrated clinical activity and was tolerable in patients with previously treated esophageal SCC.
Collapse
Affiliation(s)
- Hans Prenen
- Antwerp University Hospital, Edegem, Belgium
| | | | - Bhumsuk Keam
- Department of Internal Medicine, Seoul National University Hospital, Seoul, South Korea
| | - Colin R Lindsay
- The Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Iwona Lugowska
- Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - James C Yang
- National Taiwan University Hospital, National Taiwan University Cancer Center, Taipei City, Taiwan
| | - Federico Longo
- Ramón y Cajal University Hospital, IRYCIS, Universidad Alcala, CIBERONC, Madrid, Spain
| | - Maria de Miguel
- START-Madrid-FJD, Hospital Universitario HM Sanchinarro, Madrid, Spain
| | | | - Myung-Ju Ahn
- Samusung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | | | - Stephane Champiat
- Département d'Innovation Thérapeutique et d'Essais Précoces (DITEP), Gustave Roussy, Villejuif, France
| | | | | | - Ruth Perets
- Division of Oncology, Clinical Research Institute at Rambam, Rambam Medical Center, Haifa, Israel
| | - Cagatay Arslan
- Izmir Economy University Medical Point Medical Hospital, Izmir, Turkey
| | - Byoung C Cho
- Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Stefan Evers
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center, Basel, Switzerland
| | | | - Daniel Marbach
- Pharmaceutical Sciences, Roche Innovation Center, Basel, Switzerland
| | - David Dejardin
- Biostatistics, Product Development, Roche Innovation Center, Basel, Switzerland
| | - Nassim Sleiman
- Biostatistics, Product Development, Roche Innovation Center, Basel, Switzerland
| | | | - Muriel Richard
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center, Basel, Switzerland
| | - Jehad Charo
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Anton Kraxner
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center, Basel, Switzerland
| | - Nino Keshelava
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center Zurich, Schlieren, Switzerland
| | - Volker Teichgräber
- Roche Pharma Research and Early Development, Discovery and Translational Area Oncology, Roche Innovation Center, Basel, Switzerland
| | - Victor Moreno
- START-Madrid-FJD, Hospital Fundación Jimenez Diaz, Madrid, Spain
| |
Collapse
|
6
|
Nowak KM, Chetty R. Predictive and prognostic biomarkers in gastrointestinal tract tumours. Pathology 2024; 56:205-213. [PMID: 38238239 DOI: 10.1016/j.pathol.2023.12.412] [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: 11/16/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 02/18/2024]
Abstract
Tumours of the gastrointestinal tract represent nearly a quarter of all newly diagnosed tumours diagnosed in 2019. Various treatment modalities for gastrointestinal cancers exist, some of which may be guided by biomarkers. Biomarkers act as gauges of either normal or pathogenic processes or responses to an exposure or intervention. They come in many forms. This review explores established and potential molecular/immunohistochemical (IHC) predictive and prognostic biomarkers of the gastrointestinal tract.
Collapse
Affiliation(s)
- Klaudia M Nowak
- Laboratory Medicine Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
| | | |
Collapse
|
7
|
Angerilli V, Fassan M, Parente P, Gullo I, Campora M, Rossi C, Sacramento ML, Pennelli G, Vanoli A, Grillo F, Mastracci L. A practical approach for PD-L1 evaluation in gastroesophageal cancer. Pathologica 2023; 115:57-70. [PMID: 36537078 PMCID: PMC10462995 DOI: 10.32074/1591-951x-836] [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: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/24/2022] Open
Abstract
PD-L1 is an established predictive immunohistochemical biomarker of response to immune checkpoint inhibitors. At present, PD-L1 is routinely assessed on biopsy samples of advanced gastroesophageal cancer patients before initiating first-line treatment. However, PD-L1 is still a suboptimal biomarker, due to changing cut-off values and scoring systems, interobserver and interlaboratory variability. This practical illustrated review discusses the range of staining patterns of PD-L1 and the potential pitfalls and challenges that can be encountered when evaluating PD-L1, focusing on gastric and gastroesophageal adenocarcinoma (G/GEA) and esophageal squamous cell carcinoma (ESCC).
Collapse
Affiliation(s)
- Valentina Angerilli
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua (PD), Italy
| | - Paola Parente
- Unit of Pathology, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Irene Gullo
- Department of Pathology, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
- Department of Pathology, Faculty of Medicine of the University of Porto (FMUP), Portugal
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Portugal
| | - Michela Campora
- Public Healthcare Trust of the Autonomous Province of Trento, Santa Chiara Hospital, Department of Laboratory Medicine, Pathology Unit, Trento, Italy
| | - Chiara Rossi
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, and IRCCS San Matteo Hospital, Pavia, Italy
| | - Maria Luisa Sacramento
- Department of Pathology, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
| | - Gianmaria Pennelli
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, and IRCCS San Matteo Hospital, Pavia, Italy
| | - Federica Grillo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Italy
| | - Luca Mastracci
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Italy
| |
Collapse
|
8
|
Non-hereditary early onset gastric cancer: An unmet medical need. Curr Opin Pharmacol 2023; 68:102344. [PMID: 36608410 DOI: 10.1016/j.coph.2022.102344] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 11/28/2022] [Accepted: 12/03/2022] [Indexed: 01/06/2023]
Abstract
Gastric cancer (GC) is a lethal disease and the diagnosis in the young population is a major challenge from both individual and social point of views. Early-onset GC accounts for ∼5% of GC; among them, 3% are part of a hereditary syndrome and the majority are sporadic. However, even if the early-onset forms were less frequent in the past, the increasing number in the last decades has improved the interest and awareness of them in the society and in the scientific community. In particular, the different behaviour and characteristics of early-onset GC suggest that it is a completely different entity, which requires a tailored and personalized management. Here we provide an updated overview about non-hereditary early-onset GC, which is an unmet clinical need today, along with future perspectives in this field.
Collapse
|
9
|
Mastracci L, Grillo F, Parente P, Gullo I, Campora M, Angerilli V, Rossi C, Sacramento ML, Pennelli G, Vanoli A, Fassan M. PD-L1 evaluation in the gastrointestinal tract: from biological rationale to its clinical application. Pathologica 2022; 114:352-364. [PMID: 36305021 PMCID: PMC9614301 DOI: 10.32074/1591-951x-803] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Accepted: 09/20/2022] [Indexed: 11/06/2022] Open
Abstract
Immune-checkpoint inhibitors targeting the PD-1/PD-L1 axis have brought significant clinical benefit in many solid cancer types, including gastrointestinal malignancies. However, it has been estimated that only 20-40% of patients respond to treatment. The pattern of expression and potential predictive value of PD-L1 as an immunohistochemical biomarker has been extensively studied in gastrointestinal neoplasms. Until now, its predictive value has been demonstrated, and is currently in use only in upper gastrointestinal malignancies (gastroesophageal adenocarcinoma and esophageal squamous cell carcinoma). In this Review, we describe the technical aspects and challenges related to PD-L1 immunohistochemical assays, the current role of PD-L1 as a biomarker in clinical practice and we outline the main studies and clinical trials analyzing the prognostic and predictive value of PD-L1 in gastrointestinal cancers.
Collapse
Affiliation(s)
- Luca Mastracci
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Italy
| | - Federica Grillo
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
- Anatomic Pathology, Department of Surgical Sciences and Integrated Diagnostics (DISC), University of Genova, Italy
| | - Paola Parente
- Unit of Pathology, Fondazione IRCCS Ospedale Casa Sollievo della Sofferenza, San Giovanni Rotondo, FG, Italy
| | - Irene Gullo
- Department of Pathology, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
- Department of Pathology, Faculty of Medicine of the University of Porto (FMUP), Portugal
- i3S - Instituto de Investigação e Inovação em Saúde da Universidade do Porto, Portugal
| | - Michela Campora
- Public Healthcare Trust of the Autonomous Province of Trento, Santa Chiara Hospital, Department of Laboratory Medicine, Pathology Unit, Trento, Italy
| | - Valentina Angerilli
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
| | - Chiara Rossi
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, and IRCCS San Matteo Hospital, Pavia, Italy
| | - Maria Luisa Sacramento
- Department of Pathology, Centro Hospitalar Universitário de São João (CHUSJ), Porto, Portugal
| | - Gianmaria Pennelli
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
| | - Alessandro Vanoli
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia, and IRCCS San Matteo Hospital, Pavia, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology Unit, University Hospital of Padua, Padua (PD), Italy
- Veneto Institute of Oncology IOV - IRCCS, Padua (PD), Italy
| |
Collapse
|