1
|
Zhou KI, Hanks BA, Strickler JH. Management of Microsatellite Instability High (MSI-H) Gastroesophageal Adenocarcinoma. J Gastrointest Cancer 2024; 55:483-496. [PMID: 38133871 PMCID: PMC11186732 DOI: 10.1007/s12029-023-01003-5] [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] [Accepted: 12/17/2023] [Indexed: 12/23/2023]
Abstract
BACKGROUND Gastroesophageal cancer is a major cause of cancer-related mortality worldwide. Treatment of both early stage and advanced disease remains highly reliant on cytotoxic chemotherapy. About 4-24% of gastroesophageal cancers are microsatellite instability high (MSI-H). The MSI-H subtype is associated with favorable prognosis, resistance to cytotoxic chemotherapy, and sensitivity to immune checkpoint inhibitors (ICI). Recent studies have demonstrated promising activity of ICIs in the MSI-H subtype, resulting in fundamental changes in the management of MSI-H gastroesophageal adenocarcinoma. PURPOSE In this review, we discuss the prevalence, characteristics, prognosis, and management of MSI-H gastroesophageal adenocarcinoma, with a focus on recent and ongoing studies that have changed the landscape of treatment for the MSI-H subtype. We also discuss current challenges in the management of resectable and advanced MSI-H gastroesophageal cancer, including the need for more accurate biomarkers of response to ICI therapy.
Collapse
Affiliation(s)
- Katherine I Zhou
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA
| | - Brent A Hanks
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA
- Department of Pharmacology and Cancer Biology, Duke University, Durham, NC, USA
| | - John H Strickler
- Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC, USA.
| |
Collapse
|
2
|
Ottaiano A, Santorsola M, Ianniello M, Ceccarelli A, Casillo M, Sabbatino F, Petrillo N, Cascella M, Caraglia F, Picone C, Perri F, Sirica R, Zappavigna S, Nasti G, Savarese G, Caraglia M. Predictive significance of FGFR4 p.G388R polymorphism in metastatic colorectal cancer patients receiving trifluridine/tipiracil (TAS-102) treatment. J Transl Med 2024; 22:379. [PMID: 38650006 PMCID: PMC11036552 DOI: 10.1186/s12967-024-05184-w] [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: 03/15/2024] [Accepted: 04/08/2024] [Indexed: 04/25/2024] Open
Abstract
BACKGROUND TAS-102 (Lonsurf®) is an oral fluoropyrimidine consisting of a combination of trifluridine (a thymidine analog) and tipiracil (a thymidine phosphorylation inhibitor). The drug is effective in metastatic colorectal cancer (mCRC) patients refractory to fluorouracil, irinotecan and oxaliplatin. This study is a real-world analysis, investigating the interplay of genotype/phenotype in relation to TAS-102 sensitivity. METHODS Forty-seven consecutive mCRC patients were treated with TAS-102 at the National Cancer Institute of Naples from March 2019 to March 2021, at a dosage of 35 mg/m2, twice a day, in cycles of 28 days (from day 1 to 5 and from day 8 to 12). Clinical-pathological parameters were described. Activity was evaluated with RECIST criteria (v1.1) and toxicity with NCI-CTC (v5.0). Survival was depicted through the Kaplan-Meyer curves. Genetic features of patients were evaluated with Next Generation Sequencing (NGS) through the Illumina NovaSeq 6000 platform and TruSigt™Oncology 500 kit. RESULTS Median age of patients was 65 years (range: 46-77). Forty-one patients had 2 or more metastatic sites and 38 patients underwent to more than 2 previous lines of therapies. ECOG (Eastern Cooperative Oncology Group) Performance Status (PS) was 2 in 19 patients. The median number of TAS-102 cycles was 4 (range: 2-12). The most frequent toxic event was neutropenia (G3/G4 in 16 patients). There were no severe (> 3) non-haematological toxicities or treatment-related deaths. Twenty-six patients experienced progressive disease (PD), 21 stable disease (SD). Three patients with long-lasting disease control (DC: complete, partial responses or stable disease) shared an FGFR4 (p.Gly388Arg) mutation. Patients experiencing DC had more frequently a low tumour growth rate (P = 0.0306) and an FGFR4 p.G388R variant (P < 0.0001). The FGFR4 Arg388 genotype was associated with better survival (median: 6.4 months) compared to the Gly388 genotype (median: 4 months); the HR was 0.25 (95% CI 0.12- 0.51; P = 0.0001 at Log-Rank test). CONCLUSIONS This phenotype/genotype investigation suggests that the FGFR4 p.G388R variant may serve as a new marker for identifying patients who are responsive to TAS-102. A mechanistic hypothesis is proposed to interpret these findings.
Collapse
Affiliation(s)
- Alessandro Ottaiano
- Istituto Nazionale Tumori Di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy.
| | - Mariachiara Santorsola
- Istituto Nazionale Tumori Di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | - Monica Ianniello
- Centro Polidiagnostico Strumentale Srl, AMES, 80013, Naples, Italy
| | - Anna Ceccarelli
- Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Marika Casillo
- Centro Polidiagnostico Strumentale Srl, AMES, 80013, Naples, Italy
| | - Francesco Sabbatino
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081, Baronissi, Italy
| | - Nadia Petrillo
- Centro Polidiagnostico Strumentale Srl, AMES, 80013, Naples, Italy
| | - Marco Cascella
- Department of Medicine, Surgery and Dentistry, University of Salerno, 84081, Baronissi, Italy
| | - Francesco Caraglia
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via L. de Crecchio, 7, 80138, Naples, Italy
| | - Carmine Picone
- Istituto Nazionale Tumori Di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | - Francesco Perri
- Istituto Nazionale Tumori Di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | - Roberto Sirica
- Centro Polidiagnostico Strumentale Srl, AMES, 80013, Naples, Italy
| | - Silvia Zappavigna
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via L. de Crecchio, 7, 80138, Naples, Italy
- Laboratory of Precision and Molecular Oncology, Institute of Genetic Research, Biogem Scarl, Ariano Irpino, Italy
| | - Guglielmo Nasti
- Istituto Nazionale Tumori Di Napoli, IRCCS "G. Pascale", Via M. Semmola, 80131, Naples, Italy
| | | | - Michele Caraglia
- Department of Precision Medicine, University of Campania "L. Vanvitelli", Via L. de Crecchio, 7, 80138, Naples, Italy.
- Laboratory of Precision and Molecular Oncology, Institute of Genetic Research, Biogem Scarl, Ariano Irpino, Italy.
| |
Collapse
|
3
|
Nádorvári ML, Lotz G, Kulka J, Kiss A, Tímár J. Microsatellite instability and mismatch repair protein deficiency: equal predictive markers? Pathol Oncol Res 2024; 30:1611719. [PMID: 38655493 PMCID: PMC11036414 DOI: 10.3389/pore.2024.1611719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/27/2024] [Indexed: 04/26/2024]
Abstract
Current clinical guidelines recommend mismatch repair (MMR) protein immunohistochemistry (IHC) or molecular microsatellite instability (MSI) tests as predictive markers of immunotherapies. Most of the pathological guidelines consider MMR protein IHC as the gold standard test to identify cancers with MMR deficiency and recommend molecular MSI tests only in special circumstances or to screen for Lynch syndrome. However, there are data in the literature which suggest that the two test types may not be equal. For example, molecular epidemiology studies reported different rates of deficient MMR (dMMR) and MSI in various cancer types. Additionally, direct comparisons of the two tests revealed relatively frequent discrepancies between MMR IHC and MSI tests, especially in non-colorectal and non-endometrial cancers and in cases with unusual dMMR phenotypes. There are also scattered clinical data showing that the efficacy of immune checkpoint inhibitors is different if the patient selection was based on dMMR versus MSI status of the cancers. All these observations question the current dogma that dMMR phenotype and genetic MSI status are equal predictive markers of the immunotherapies.
Collapse
Affiliation(s)
| | | | | | | | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|
4
|
Lukomski L, Pisula J, Wirsik N, Damanakis A, Jung JO, Knipper K, Datta R, Schröder W, Gebauer F, Schmidt T, Quaas A, Bozek K, Bruns C, Popp F. Analyzing the Impact of Oncological Data at Different Time Points and Tumor Biomarkers on Artificial Intelligence Predictions for Five-Year Survival in Esophageal Cancer. MACHINE LEARNING AND KNOWLEDGE EXTRACTION 2024; 6:679-698. [DOI: 10.3390/make6010032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
Abstract
AIM: In this study, we use Artificial Intelligence (AI), including Machine (ML) and Deep Learning (DL), to predict the long-term survival of resectable esophageal cancer (EC) patients in a high-volume surgical center. Our objective is to evaluate the predictive efficacy of AI methods for survival prognosis across different time points of oncological treatment. This involves comparing models trained with clinical data, integrating either Tumor, Node, Metastasis (TNM) classification or tumor biomarker analysis, for long-term survival predictions. METHODS: In this retrospective study, 1002 patients diagnosed with EC between 1996 and 2021 were analyzed. The original dataset comprised 55 pre- and postoperative patient characteristics and 55 immunohistochemically evaluated biomarkers following surgical intervention. To predict the five-year survival status, four AI methods (Random Forest RF, XG Boost XG, Artificial Neural Network ANN, TabNet TN) and Logistic Regression (LR) were employed. The models were trained using three predefined subsets of the training dataset as follows: (I) the baseline dataset (BL) consisting of pre-, intra-, and postoperative data, including the TNM but excluding tumor biomarkers, (II) clinical data accessible at the time of the initial diagnostic workup (primary staging dataset, PS), and (III) the PS dataset including tumor biomarkers from tissue microarrays (PS + biomarkers), excluding TNM status. We used permutation feature importance for feature selection to identify only important variables for AI-driven reduced datasets and subsequent model retraining. RESULTS: Model training on the BL dataset demonstrated similar predictive performances for all models (Accuracy, ACC: 0.73/0.74/0.76/0.75/0.73; AUC: 0.78/0.82/0.83/0.80/0.79 RF/XG/ANN/TN/LR, respectively). The predictive performance and generalizability declined when the models were trained with the PS dataset. Surprisingly, the inclusion of biomarkers in the PS dataset for model training led to improved predictions (PS dataset vs. PS dataset + biomarkers; ACC: 0.70 vs. 0.77/0.73 vs. 0.79/0.71 vs. 0.75/0.69 vs. 0.72/0.63 vs. 0.66; AUC: 0.77 vs. 0.83/0.80 vs. 0.85/0.76 vs. 0.86/0.70 vs. 0.76/0.70 vs. 0.69 RF/XG/ANN/TN/LR, respectively). The AI models outperformed LR when trained with the PS datasets. The important features shared after AI-driven feature selection in all models trained with the BL dataset included histopathological lymph node status (pN), histopathological tumor size (pT), clinical tumor size (cT), age at the time of surgery, and postoperative tracheostomy. Following training with the PS dataset with biomarkers, the important predictive features included patient age at the time of surgery, TP-53 gene mutation, Mesothelin expression, thymidine phosphorylase (TYMP) expression, NANOG homebox protein expression, and indoleamine 2,3-dioxygenase (IDO) expressed on tumor-infiltrating lymphocytes, as well as tumor-infiltrating Mast- and Natural killer cells. CONCLUSION: Different AI methods similarly predict the long-term survival status of patients with EC and outperform LR, the state-of-the-art classification model. Survival status can be predicted with similar predictive performance with patient data at an early stage of treatment when utilizing additional biomarker analysis. This suggests that individual survival predictions can be made early in cancer treatment by utilizing biomarkers, reducing the necessity for the pathological TNM status post-surgery. This study identifies important features for survival predictions that vary depending on the timing of oncological treatment.
Collapse
Affiliation(s)
- Leandra Lukomski
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Juan Pisula
- Data science of Bioimages Lab, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital of Cologne, Robert-Koch-Straße 21, 50937 Cologne, Germany
| | - Naita Wirsik
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Alexander Damanakis
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Jin-On Jung
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Karl Knipper
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Rabi Datta
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Wolfgang Schröder
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Florian Gebauer
- Department of General, Visceral and Cancer Surgery, Helios University Hospital Wuppertal, University Witten/Herdecke, Heusnerstraße 40, 42283 Wuppertal, Germany
| | - Thomas Schmidt
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Katarzyna Bozek
- Data science of Bioimages Lab, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital of Cologne, Robert-Koch-Straße 21, 50937 Cologne, Germany
| | - Christiane Bruns
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Felix Popp
- Department of General, Visceral and Cancer Surgery, Faculty of Medicine and University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| |
Collapse
|
5
|
Mehta R, Sinnamon A, Dam A, Walko C, Palm R, Barton L, Lauwers G, Pimiento JM. Locally advanced mismatch repair-deficient gastroesophageal junction cancer: Diagnosis, treatment modifications, and monitoring. CA Cancer J Clin 2024; 74:123-131. [PMID: 37849051 DOI: 10.3322/caac.21813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/05/2023] [Indexed: 10/19/2023] Open
Affiliation(s)
- Rutika Mehta
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Andrew Sinnamon
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Aamir Dam
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Christine Walko
- Precision Medicine, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Russell Palm
- Radiation Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Laura Barton
- Personalized Medicine, H. Lee Moffitt Cancer Center, Florida, Tampa, USA
| | - Gregory Lauwers
- Pathology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| | - Jose M Pimiento
- Gastrointestinal Oncology, H. Lee Moffitt Cancer Center, Tampa, Florida, USA
| |
Collapse
|
6
|
Nádorvári ML, Kenessey I, Kiss A, Barbai T, Kulka J, Rásó E, Tímár J. Comparison of standard mismatch repair deficiency and microsatellite instability tests in a large cancer series. J Transl Med 2024; 22:150. [PMID: 38350968 PMCID: PMC10863158 DOI: 10.1186/s12967-024-04960-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: 10/17/2023] [Accepted: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND The tumor-agnostic indication of immune checkpoint inhibitors to treat cancers with mismatch repair deficiency (dMMR)/microsatellite instability (MSI) increased the demand for such tests beyond Lynch syndrome. International guideline recommendations accept immunohistochemistry (IHC) for dMMR or molecular techniques (PCR or NGS) for MSI status determinations considering the two tests are equal, although there are scattered reports contradicting to this presumption. MATERIALS AND METHODS Here we have directly compared four protein MMR immunohistochemistry (IHC) to MSI Pentaplex PCR test in a large cancer patient cohort (n = 1306) of our diagnostic center where the two tests have been run parallel in 703 cases. RESULTS In this study we have found a high discrepancy rate (19.3%) of the two tests which was independent of the tumor types. The MSI PCR sensitivity for MMR IHC status was found to be very low resulting in a relatively low positive and negative predicting values. As a consequence, the correlation of the two tests was low (kappa < 0.7). During analysis of the possible contributing factors of this poor performance, we have excluded low tumor percentage of the samples, but identified dMMR phenotypes (classic versus non-classic or unusual) as possible contributors. CONCLUSION Although our cohort did not include samples with identified technical errors, our data strongly support previous reports that unidentified preanalytical factors might have the major influence on the poor performance of the MSI PCR and MMR IHC. Furthermore, the case is open whether the two test types are equally powerful predictive markers of immunotherapies.
Collapse
Affiliation(s)
- Maja L Nádorvári
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - István Kenessey
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - András Kiss
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Tamás Barbai
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Erzsébet Rásó
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
7
|
Talari FF, Bozorg A, Zeinali S, Zali M, Mohsenifar Z, Asadzadeh Aghdaei H, Baghaei K. Low incidence of microsatellite instability in gastric cancers and its association with the clinicopathological characteristics: a comparative study. Sci Rep 2023; 13:21743. [PMID: 38065969 PMCID: PMC10709324 DOI: 10.1038/s41598-023-48157-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Gastric cancer is a complex heterogeneous disease with different molecular subtypes that have clinical implications. It is characterized by high mortality rates and limited effective therapies. Microsatellite instability (MSI) has been recognized as a subgroup with a good prognosis based on TCGA and ACRG categorizations. Besides its prognostic and predictive value, gastric cancers with high MSI exhibit different clinical behaviors. The prevalence of high MSI has been assessed in gastric cancer worldwide, especially in East Asia, but there is a lack of such information in the Middle East. Therefore, this study aimed to investigate the incidence and status of MSI in Iranian gastric cancer patients using 53 samples collected from 2015 to 2020 at Taleghani Hospital Medical Center. DNA from tumoral and normal tissues were extracted and assessed through multiplex-PCR based on five mononucleotide repeats panel. Clinicopathological variables, including age, sex, Lauren classification, lymph node involvement, TNM stage, differentiation, localization, and tumor size, were also analyzed. With 2 males and 2 females, high microsatellite instability represented a small subgroup of almost 7.5% of the samples with a median age of 60.5 years. High microsatellite instability phenotypes were significantly associated with patients aged 68 years and older (p‑value of 0.0015) and lower lymph node involvement (p‑value of 0.0004). Microsatellite instability was also more frequent in females, with distal gastric location, bigger tumor size, and in the intestinal type of gastric cancer rather than the diffuse type.
Collapse
Affiliation(s)
| | - Ali Bozorg
- Biotechnology Department, College of Science, University of Tehran, Tehran, Iran.
| | - Sirous Zeinali
- Dr. Zeinali's Medical Genetics Laboratory, Kawsar Human Genetics Research Center, Tehran, Iran
- Department of Molecular Medicine, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammadreza Zali
- Research Institute for Gastroenterology and Liver Diseases, Gastroenterology and Liver Diseases Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zhale Mohsenifar
- Department of Pathology, School of Medicine, Taleghani Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Asadzadeh Aghdaei
- Research Institute for Gastroenterology and Liver Diseases, Gastroenterology and Liver Diseases Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Kaveh Baghaei
- Research Institute for Gastroenterology and Liver Diseases, Gastroenterology and Liver Diseases Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Silva JR, Mascarenhas-Lemos L, Neto do Nascimento C, Sousa Marques D, Wen X, Pinho L, Maio R, Pontes P, Cirnes L, Cravo M, Carneiro F, Gullo I. Role of Endoscopic Biopsies and Morphologic Features in Predicting Microsatellite Instability Status in Gastric Cancer: A Multicenter Comparative Study of Endoscopic Biopsies and Surgical Specimens. Am J Surg Pathol 2023; 47:990-1000. [PMID: 37366224 DOI: 10.1097/pas.0000000000002085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Evaluation of mismatch repair (MMR) protein and microsatellite instability (MSI) status plays a pivotal role in the management of gastric cancer (GC) patients. In this study, we aimed to evaluate the accuracy of gastric endoscopic biopsies (EBs) in predicting MMR/MSI status and to uncover histopathologic features associated with MSI. A multicentric series of 140 GCs was collected retrospectively, in which EB and matched surgical specimens (SSs) were available. Laurén and WHO classifications were applied and detailed morphologic characterization was performed. EB/SS were analyzed by immunohistochemistry (IHC) for MMR status and by multiplex polymerase chain reaction (mPCR) for MSI status. IHC allowed accurate evaluation of MMR status in EB (sensitivity: 97.3%; specificity: 98.0%) and high concordance rates between EB and SS (Cohen κ=94.5%). By contrast, mPCR (Idylla MSI Test) showed lower sensitivity in evaluating MSI status (91.3% vs. 97.3%), while maintaining maximal specificity (100.0%). These results suggest a role of IHC as a screening method for MMR status in EB and the use of mPCR as a confirmatory test. Although Laurén/WHO classifications were not able to discriminate GC cases with MSI, we identified specific histopathologic features that are significantly associated with MMR/MSI status in GC, despite the morphologic heterogeneity of GC cases harboring this molecular phenotype. In SS, these features included the presence of mucinous and/or solid components ( P =0.034 and <0.001) and the presence of neutrophil-rich stroma, distant from tumor ulceration/perforation ( P <0.001). In EB, both solid areas and extracellular mucin lakes were also discriminating features for the identification of MSI-high cases ( P =0.002 and 0.045).
Collapse
Affiliation(s)
- João R Silva
- Faculty of Medicine of the University of Porto (FMUP)
| | - Luís Mascarenhas-Lemos
- Departments of Pathology
- Faculty of Medicine, Catholic University of Portugal
- NOVA Medical School, Universidade NOVA Lisbon
| | | | | | - Xiaogang Wen
- i3S (Instituto de Investigação e Inovação em Saúde) and Ipatimup (Institute of Molecular Pathology and Immunology of the University of Porto)
- Department of Pathology, Centro Hospitalar Universitário do Porto (CHUP)
| | - Lídia Pinho
- i3S (Instituto de Investigação e Inovação em Saúde) and Ipatimup (Institute of Molecular Pathology and Immunology of the University of Porto)
| | - Rui Maio
- Surgery
- NOVA Medical School, Universidade NOVA Lisbon
| | - Patrícia Pontes
- Department of Pathology, University Hospital Center of São João (CHUSJ)
| | - Luís Cirnes
- i3S (Instituto de Investigação e Inovação em Saúde) and Ipatimup (Institute of Molecular Pathology and Immunology of the University of Porto)
| | - Marília Cravo
- Gastroenterology, Hospital da Luz Lisbon
- Faculty of Medicine, University of Lisbon (FMUL), Lisbon
| | - Fátima Carneiro
- i3S (Instituto de Investigação e Inovação em Saúde) and Ipatimup (Institute of Molecular Pathology and Immunology of the University of Porto)
- Department of Pathology, University Hospital Center of São João (CHUSJ)
- Department of Pathology, FMUP, Porto
| | - Irene Gullo
- i3S (Instituto de Investigação e Inovação em Saúde) and Ipatimup (Institute of Molecular Pathology and Immunology of the University of Porto)
- Department of Pathology, University Hospital Center of São João (CHUSJ)
- Department of Pathology, FMUP, Porto
| |
Collapse
|
9
|
Hiltner T, Kohlruss M, Herz AL, Lorenzen S, Novotny A, Hapfelmeier A, Jesinghaus M, Slotta-Huspenina J, Sisic L, Gaida MM, Weichert W, Ott K, Keller G. Microsatellite instability and sex-specific differences of survival in gastric cancer after neoadjuvant chemotherapy without and with taxane: An observational study in real world patients. J Cancer Res Clin Oncol 2023; 149:7651-7662. [PMID: 37000259 PMCID: PMC10374811 DOI: 10.1007/s00432-023-04691-5] [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: 02/13/2023] [Accepted: 03/10/2023] [Indexed: 04/01/2023]
Abstract
PURPOSE To investigate the prognostic role of microsatellite instability (MSI) in association with sex of patients treated with platinum/fluoropyrimidine neoadjuvant chemotherapy (CTx) with or without a taxane-containing compound. METHODS Of the 505 retrospectively analyzed patients with gastric or gastroesophageal adenocarcinoma, 411 patients were treated without taxane and 94 patients with a taxane-containing compound. MSI was determined using standard assays. RESULTS Females demonstrated a better overall survival (OS) than males in the non-taxane group (HR, 0.59; 95% CI 0.41-0.86; p = 0.005), whereas no significant difference was found in the taxane group (HR 1.22; 95% CI 0.55-2.73, p = 0.630). MSI-High (-H) was associated with a better prognosis in both groups (without taxane: HR 0.56; 95% CI 0.33-0.97; p = 0.038; with taxane: HR 0.28; 95% CI 0.04-2.02, p = 0.204). In the non-taxane group, female MSI-H patients showed the best OS (HR 0.18, 95% CI 0.05-0.73; p = 0.016), followed by the female microsatellite stable (MSS) (HR 0.67, 95% CI 0.46-0.98, p = 0.040) and the male MSI-H group (HR 0.76; 95% CI 0.42-1.37, p = 0.760) taken the male MSS group as reference. In the taxane group, female and male MSI-H patients demonstrated the best OS (female MSI-H: HR 0.05, 95% CI 0.00-240.46; male MSI-H: HR 0.45, 95% CI 0.61-3.63, p = 0.438), whereas the female MSS group showed a decreased OS (HR 1.39 95% CI 0.62-3.12, p = 0.420) compared to male MSS patients. CONCLUSION OS in gastric/gastroesophageal cancer after CTx might depend on sex and MSI status and may differ between patients treated with or without a taxane compound in the chemotherapeutic regimen.
Collapse
Affiliation(s)
- Theresa Hiltner
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
| | - Meike Kohlruss
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
| | - Anna-Lina Herz
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
| | - Sylvie Lorenzen
- III. Medizinische Klinik and Poliklinik, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Novotny
- Department of Surgery, TUM School of Medicine, Technical University of Munich, Munich, Germany
| | - Alexander Hapfelmeier
- Institute of AI and Informatics in Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- Institute of General Practice and Health Services Research, School of Medicine, Technical University of Munich, Munich, Germany
| | - Moritz Jesinghaus
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
- Institute of Pathology, University of Marburg, Marburg, Germany
| | - Julia Slotta-Huspenina
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
| | - Leila Sisic
- Department of General, Visceral and Transplantation Surgery, University of Heidelberg, Heidelberg, Germany
| | - Matthias M Gaida
- Institute of Pathology, University of Heidelberg, Heidelberg, Germany
- Institute of Pathology, University Medical Center Mainz, JGU-Mainz, Mainz, Germany
- TRON-Translational, Oncology at The University Medical Center of The Johannes Gutenberg University gGmbH, Mainz, Germany
| | - Wilko Weichert
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
- Institute of Pathology, German Cancer Consortium [DKTK], Partner Site Munich, Munich, Germany
| | - Katja Ott
- Department of Surgery, Klinikum Rosenheim, Rosenheim, Germany
| | - Gisela Keller
- Institute of Pathology, TUM School of Medicine, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany.
| |
Collapse
|
10
|
Molnar A, Monroe H, Basri Aydin H, Arslan ME, Lightle A, Lee H, El Jabbour T. Tumors of the Digestive System: Comprehensive Review of Ancillary Testing and Biomarkers in the Era of Precision Medicine. Curr Oncol 2023; 30:2388-2404. [PMID: 36826143 PMCID: PMC9954843 DOI: 10.3390/curroncol30020182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 02/13/2023] [Accepted: 02/15/2023] [Indexed: 02/18/2023] Open
Abstract
Immunotherapy has remained at the vanguard of promising cancer therapeutic regimens due to its exceptionally high specificity for tumor cells and potential for significantly improved treatment-associated quality of life compared to other therapeutic approaches such as surgery and chemoradiation. This is especially true in the digestive system, where high rates of mutation give rise to a host of targetable tumor-specific antigens. Many patients, however, do not exhibit measurable improvements under immunotherapy due to intrinsic or acquired resistance, making predictive biomarkers necessary to determine which patients will benefit from this line of treatment. Many of these biomarkers are assessed empirically by pathologists according to nuanced scoring criteria and algorithms. This review serves to inform clinicians and pathologists of extant and promising upcoming biomarkers predictive of immunotherapeutic efficacy among digestive system malignancies and the ancillary testing required for interpretation by pathologists according to tumor site of origin.
Collapse
Affiliation(s)
- Attila Molnar
- Department of Pathology, Icahn School of Medicine at Mount Sinai, New York, NY 10025, USA
| | - Hunter Monroe
- Department of Pathology, West Virginia University, Morgantown, WV 26506, USA
| | - Hasan Basri Aydin
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Mustafa Erdem Arslan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Andrea Lightle
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Hwajeong Lee
- Department of Pathology, Albany Medical Center, Albany, NY 12208, USA
| | - Tony El Jabbour
- Department of Pathology, West Virginia University, Morgantown, WV 26506, USA
- Correspondence:
| |
Collapse
|
11
|
Röcken C. Predictive biomarkers in gastric cancer. J Cancer Res Clin Oncol 2023; 149:467-481. [PMID: 36260159 PMCID: PMC9889517 DOI: 10.1007/s00432-022-04408-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 02/04/2023]
Abstract
Predictive biomarkers are the mainstay of precision medicine. This review summarizes the advancements in tissue-based diagnostic biomarkers for gastric cancer, which is considered the leading cause of cancer-related deaths worldwide. A disease seen in the elderly, it is often diagnosed at an advanced stage, thereby limiting therapeutic options. In Western countries, neoadjuvant/perioperative (radio-)chemotherapy is administered, and adjuvant chemotherapy is administered in the East. The morpho-molecular classification of gastric cancer has opened novel avenues identifying Epstein-Barr-Virus (EBV)-positive, microsatellite instable, genomically stable and chromosomal instable gastric cancers. In chromosomal instable tumors, receptor tyrosine kinases (RKTs) (e.g., EGFR, FGFR2, HER2, and MET) are frequently overexpressed. Gastric cancers such as microsatellite instable and EBV-positive types often express immune checkpoint molecules, such as PD-L1 and VISTA. Genomically stable tumors show alterations in claudin 18.2. Next-generation sequencing is increasingly being used to search for druggable targets in advanced palliative settings. However, most tissue-based biomarkers of gastric cancer carry the risk of a sampling error due to intratumoral heterogeneity, and adequate tissue sampling is of paramount importance.
Collapse
Affiliation(s)
- C. Röcken
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, Haus U33, 24105 Kiel, Germany
| |
Collapse
|
12
|
EBV and MSI Status in Gastric Cancer: Does It Matter? Cancers (Basel) 2022; 15:cancers15010074. [PMID: 36612071 PMCID: PMC9817503 DOI: 10.3390/cancers15010074] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/13/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
We investigated the impactof microsatellite instability (MSI) and Epstein-Barr virus (EBV) status in gastric cancer (GC), regarding response to perioperative chemotherapy (POPChT), overall survival (OS), and progression-free survival (PFS). We included 137 cases of operated GC, 51 of which were submitted to POPChT. MSI status was determined by multiplex PCR and EBV status by EBV-encoded RNA in situ hybridization. Thirty-seven (27%) cases presented as MSI-high, and seven (5.1%) were EBV+. Concerning tumor regression after POPChT, no differences were observed between the molecular subtypes, but females were more likely to respond (p = 0.062). No significant differences were found in OS or PFS between different subtypes. In multivariate analysis, age (HR 1.02, IC 95% 1.002-1.056, p = 0.033) and positive lymph nodes (HR 1.82, IC 95% 1.034-3.211, p = 0.038) were the only prognostic factors for OS. However, females with MSI-high tumors treated with POPChT demonstrated a significantly increased OS compared to females with MSS tumors (p = 0.031). In conclusion, we found a high proportion of MSI-high cases. MSI and EBV status did not influence OS or PFS either in patients submitted to POPChT or surgery alone. However, superior survival of females with MSI-high tumors suggests that sex disparities and molecular classification may influence treatment options in GC.
Collapse
|
13
|
Mayr C, Kiesslich T, Modest DP, Stintzing S, Ocker M, Neureiter D. Chemoresistance and resistance to targeted therapies in biliary tract cancer: what have we learned? Expert Opin Investig Drugs 2022; 31:221-233. [PMID: 35098846 DOI: 10.1080/13543784.2022.2034785] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Biliary tract cancer (BTC), including intra- and extrahepatic cholangiocarcinoma and gallbladder cancer, is a rare and highly difficult to manage human malignancy. Besides late diagnosis and associated unresectability, frequently observed unresponsiveness toward and recurrence following chemotherapy or targeted therapy essentially contribute to the dismal prognosis of BTC patients. AREAS COVERED The review provides an update on individual mechanisms involved resistance of BTC toward conventional chemotherapy as well as targeted therapies. We review the distinct mechanisms of pharmacoresistance (MPRs) which have been defined in BTC cells on a molecular basis and examine the specific consequences for the various approaches of chemo-, targeted or immunomodulatory therapies. EXPERT OPINION Based on currently available experimental and clinical data, the present knowledge about these MPRs in BTCs are summarized. While some possible tactics for overcoming these mechanisms of resistance have been investigated, a BTC-specific and efficient approach based on comprehensive in vitro and in vivo experimental systems is not yet available. Additionally, a reliable monitoring of therapy-relevant cellular changes needs to be established which allows for choosing the optimal drug (combination) before and/or during pharmacological therapy.
Collapse
Affiliation(s)
- Christian Mayr
- Center for Physiology, Pathophysiology and Biophysics - Salzburg and Nuremberg, Institute for Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
- Department of Internal Medicine I, Paracelsus Medical University/University Hospital Salzburg (Salk), Salzburg, Austria
| | - Tobias Kiesslich
- Center for Physiology, Pathophysiology and Biophysics - Salzburg and Nuremberg, Institute for Physiology and Pathophysiology, Paracelsus Medical University, Salzburg, Austria
- Department of Internal Medicine I, Paracelsus Medical University/University Hospital Salzburg (Salk), Salzburg, Austria
| | - Dominik Paul Modest
- Medical Department, Division of Hematology,Oncology, and Tumor Immunology (Campus Charité Mitte), Charité University Medicine Berlin, Berlin, Germany
| | - Sebastian Stintzing
- Medical Department, Division of Hematology,Oncology, and Tumor Immunology (Campus Charité Mitte), Charité University Medicine Berlin, Berlin, Germany
| | - Matthias Ocker
- Charité University Medicine Berlin, Berlin, Germany
- Translational Medicine & Clinical Pharmacology, Boehringer Ingelheim Pharma GmbH & Co. Kg, Ingelheim, Germany
| | - Daniel Neureiter
- Institute of Pathology, Paracelsus Medical University/University Hospital Salzburg (Salk), Salzburg, Austria
- Cancer Cluster Salzburg, Austria
| |
Collapse
|
14
|
Lu F, Chen W, Jiang T, Cheng C, Wang B, Lu Z, Huang G, Qiu J, Wei W, Yang M, Huang X. Expression profile, clinical significance and biological functions of IGF2BP2 in esophageal squamous cell carcinoma. Exp Ther Med 2022; 23:252. [PMID: 35261624 DOI: 10.3892/etm.2022.11177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 09/17/2021] [Indexed: 11/05/2022] Open
Affiliation(s)
- Fenying Lu
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Weichang Chen
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| | - Tingwang Jiang
- Department of Science and Technology Division, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| | - Cuie Cheng
- Department of Gastroenterology, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| | - Bin Wang
- Department of Gastroenterology, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| | - Zhiping Lu
- Department of Gastroenterology, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| | - Guojin Huang
- Department of Gastroenterology, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| | - Jiaming Qiu
- Department of Pathology, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| | - Wei Wei
- Department of Pathology, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| | - Ming Yang
- Department of Thoracic Surgery, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| | - Xia Huang
- Department of Gastroenterology, The Second People's Hospital of Changshu, Suzhou, Jiangsu 215500, P.R. China
| |
Collapse
|