501
|
Ratovomanana T, Cohen R, Svrcek M, Renaud F, Cervera P, Siret A, Letourneur Q, Buhard O, Bourgoin P, Guillerm E, Dorard C, Nicolle R, Ayadi M, Touat M, Bielle F, Sanson M, Le Rouzic P, Buisine MP, Piessen G, Collura A, Fléjou JF, de Reyniès A, Coulet F, Ghiringhelli F, André T, Jonchère V, Duval A. Performance of Next-Generation Sequencing for the Detection of Microsatellite Instability in Colorectal Cancer With Deficient DNA Mismatch Repair. Gastroenterology 2021; 161:814-826.e7. [PMID: 33992635 DOI: 10.1053/j.gastro.2021.05.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/02/2022]
Abstract
BACKGROUND & AIMS Next-generation sequencing (NGS) was recently approved by the United States Food and Drug Administration to detect microsatellite instability (MSI) arising from defective mismatch repair (dMMR) in patients with metastatic colorectal cancer (mCRC) before treatment with immune checkpoint inhibitors (ICI). In this study, we aimed to evaluate and improve the performance of NGS to identify MSI in CRC, especially dMMR mCRC treated with ICI. METHODS CRC samples used in this post hoc study were reassessed centrally for MSI and dMMR status using the reference methods of pentaplex polymerase chain reaction and immunohistochemistry. Whole-exome sequencing (WES) was used to evaluate MSISensor, the Food and Drug Administration-approved and NGS-based method for assessment of MSI. This was performed in (1) a prospective, multicenter cohort of 102 patients with mCRC (C1; 25 dMMR/MSI, 24 treated with ICI) from clinical trials NCT02840604 and NCT033501260, (2) an independent retrospective, multicenter cohort of 113 patients (C2; 25 mCRC, 88 non-mCRC, all dMMR/MSI untreated with ICI), and (3) a publicly available series of 118 patients with CRC from The Cancer Genome Atlas (C3; 51 dMMR/MSI). A new NGS-based algorithm, namely MSICare, was developed. Its performance for assessment of MSI was compared with MSISensor in C1, C2, and C3 at the exome level or after downsampling sequencing data to the MSK-IMPACT gene panel. MSICare was validated in an additional retrospective, multicenter cohort (C4) of 152 patients with new CRC (137 dMMR/MSI) enriched in tumors deficient in MSH6 (n = 35) and PMS2 (n = 9) after targeted sequencing of samples with an optimized set of microsatellite markers (MSIDIAG). RESULTS At the exome level, MSISensor was highly specific but failed to diagnose MSI in 16% of MSI/dMMR mCRC from C1 (4 of 25; sensitivity, 84%; 95% confidence interval [CI], 63.9%-95.5%), 32% of mCRC (8 of 25; sensitivity, 68%; 95% CI, 46.5%-85.1%), and 9.1% of non-mCRC from C2 (8 of 88; sensitivity, 90.9%; 95% CI, 82.9%-96%), and 9.8% of CRC from C3 (5 of 51; sensitivity, 90.2%; 95% CI, 78.6%-96.7%). Misdiagnosis included 4 mCRCs treated with ICI, of which 3 showed an overall response rate without progression at this date. At the exome level, reevaluation of the MSI genomic signal using MSICare detected 100% of cases with true MSI status among C1 and C2. Further validation of MSICare was obtained in CRC tumors from C3, with 96.1% concordance for MSI status. Whereas misdiagnosis with MSISensor even increased when analyzing downsampled WES data from C1 and C2 with microsatellite markers restricted to the MSK-IMPACT gene panel (sensitivity, 72.5%; 95% CI, 64.2%-79.7%), particularly in the MSH6-deficient setting, MSICare sensitivity and specificity remained optimal (100%). Similar results were obtained with MSICare after targeted NGS of tumors from C4 with the optimized microsatellite panel MSIDIAG (sensitivity, 99.3%; 95% CI, 96%-100%; specificity, 100%). CONCLUSIONS In contrast to MSISensor, the new MSICare test we propose performs at least as efficiently as the reference method, MSI polymerase chain reaction, to detect MSI in CRC regardless of the defective MMR protein under both WES and targeted NGS conditions. We suggest MSICare may rapidly become a reference method for NGS-based testing of MSI in CRC, especially in mCRC, where accurate MSI status is required before the prescription of ICI.
Collapse
Affiliation(s)
- Toky Ratovomanana
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Romain Cohen
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Department of Medical Oncology, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Magali Svrcek
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Department of Pathology, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Florence Renaud
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277 CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France
| | - Pascale Cervera
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Genetics Department, AP-HP, Hospital Pitié-Salpêtrière, Paris, France
| | - Aurélie Siret
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Quentin Letourneur
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Olivier Buhard
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Pierre Bourgoin
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Erell Guillerm
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Genetics Department, AP-HP, Hospital Pitié-Salpêtrière, Paris, France
| | - Coralie Dorard
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Remy Nicolle
- Programme "Cartes d'Identité des Tumeurs," Ligue Nationale Contre le Cancer, Paris, France
| | - Mira Ayadi
- Programme "Cartes d'Identité des Tumeurs," Ligue Nationale Contre le Cancer, Paris, France
| | - Mehdi Touat
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Service de Neurologie 2-Mazarin, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Paris, France
| | - Franck Bielle
- Service de Neurologie 2-Mazarin, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Paris, France; AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Département de Neuropathologie, Paris, France
| | - Marc Sanson
- Service de Neurologie 2-Mazarin, Sorbonne Université, Inserm, CNRS, UMR S 1127, Institut du Cerveau, ICM, AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Paris, France; AP-HP, Hôpitaux Universitaires La Pitié Salpêtrière-Charles Foix, Département de Neuropathologie, Paris, France
| | - Philippe Le Rouzic
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Genetics Department, AP-HP, Hospital Pitié-Salpêtrière, Paris, France
| | - Marie-Pierre Buisine
- Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277 CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France; Molecular Oncogenetics, Department of Biochemistry and Molecular Biology, Lille University Hospital, Lille, France
| | - Guillaume Piessen
- Université de Lille, CNRS, INSERM, CHU Lille, UMR9020-U1277 CANTHER-Cancer Heterogeneity Plasticity and Resistance to Therapies, Lille, France; Department of Digestive and Oncological Surgery, Claude Huriez University Hospital, University Lille, Lille, France
| | - Ada Collura
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - Jean-François Fléjou
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Department of Pathology, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Aurélien de Reyniès
- Programme "Cartes d'Identité des Tumeurs," Ligue Nationale Contre le Cancer, Paris, France
| | - Florence Coulet
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Genetics Department, AP-HP, Hospital Pitié-Salpêtrière, Paris, France
| | - François Ghiringhelli
- Department of Medical Oncology, Centre Georges-François Leclerc, Dijon, France; INSERM, UMR866, Burgundy University, Dijon, France
| | - Thierry André
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Department of Medical Oncology, AP-HP, Hôpital Saint-Antoine, Paris, France
| | - Vincent Jonchère
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Genetics Department, AP-HP, Hospital Pitié-Salpêtrière, Paris, France
| | - Alex Duval
- Sorbonne Université, INSERM, Unité Mixte de Recherche Scientifique 938 and Site de Recherche Intégrée sur le Cancer (SIRIC) Cancer United Research Associating Medicine, University & Society (CURAMUS), Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne Université, Genetics Department, AP-HP, Hospital Pitié-Salpêtrière, Paris, France.
| |
Collapse
|
502
|
Ukkola I, Nummela P, Pasanen A, Kero M, Lepistö A, Kytölä S, Bützow R, Ristimäki A. Detection of microsatellite instability with Idylla MSI assay in colorectal and endometrial cancer. Virchows Arch 2021; 479:471-479. [PMID: 33755781 PMCID: PMC8448708 DOI: 10.1007/s00428-021-03082-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2021] [Revised: 02/23/2021] [Accepted: 03/11/2021] [Indexed: 02/06/2023]
Abstract
Universal testing of microsatellite instability (MSI) is recommended for colorectal cancer (CRC) and endometrial cancer (EC) to screen for Lynch syndrome and to aid in assessing prognosis and optimal treatment. We compared the performance of Idylla MSI test to immunohistochemistry (IHC) of mismatch repair (MMR) proteins in consecutive series of 100 CRC and 108 EC samples, as well as in retrospective series of 28 CRC and 33 EC specimens with known deficient MMR protein expression. The concordance between the Idylla test and IHC was 100% in all CRC samples (n=128) but lower in EC samples (87.2%; n=141). In the EC samples, sensitivity of Idylla test was 72.7% and specificity 100%. EC MSI/dMMR agreement was 85.4% for MLH1, 87.5% for MSH2, and only 35.3% for MSH6. When we analyzed 14 EC samples that were discrepant, i.e., dMMR using IHC and microsatellite stable using Idylla, with microsatellite markers BAT25 and BAT26, we found four cases to be replication error (RER) positive. All RER positive cases were deficient for MSH6 protein expression. We also re-analyzed EC samples with variable tumor cellularity to determine the limit of detection of the Idylla test and found that a 30% or higher tumor cellularity is required. We conclude that Idylla MSI test offers a sensitive and specific method for CRC diagnostics but is less sensitive in EC samples especially in the case of MSH6 deficiency.
Collapse
Affiliation(s)
- Iiris Ukkola
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, P.O. Box 400, HUS, FI-00029, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Pirjo Nummela
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Annukka Pasanen
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, P.O. Box 400, HUS, FI-00029, Helsinki, Finland
| | - Mia Kero
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, P.O. Box 400, HUS, FI-00029, Helsinki, Finland
| | - Anna Lepistö
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
- Department of Gastrointestinal Surgery, Helsinki University Hospital, Helsinki, Finland
| | - Soili Kytölä
- Department of Genetics, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital, Helsinki, Finland
| | - Ralf Bützow
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, P.O. Box 400, HUS, FI-00029, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland
| | - Ari Ristimäki
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, P.O. Box 400, HUS, FI-00029, Helsinki, Finland.
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
503
|
De Silva P, Bano S, Pogue BW, Wang KK, Maytin EV, Hasan T. Photodynamic priming with triple-receptor targeted nanoconjugates that trigger T cell-mediated immune responses in a 3D in vitro heterocellular model of pancreatic cancer. NANOPHOTONICS 2021; 10:3199-3214. [PMID: 37485044 PMCID: PMC10361703 DOI: 10.1515/nanoph-2021-0304] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/25/2023]
Abstract
Photodynamic priming (PDP), a collateral effect of photodynamic therapy, can transiently alter the tumor microenvironment (TME) beyond the cytotoxic zone. Studies have demonstrated that PDP increases tumor permeability and modulates immune-stimulatory effects by inducing immunogenic cell death, via the release of damage-associated molecular patterns and tumor-associated antigens. Pancreatic ductal adenocarcinoma (PDAC) is one of the deadliest of cancers with a stubborn immunosuppressive TME and a dense stroma, representing a challenge for current molecular targeted therapies often involving macromolecules. We, therefore, tested the hypothesis that PDP's TME modulation will enable targeted therapy and result in immune stimulation. Using triple-receptor-targeted photoimmuno-nanoconjugate (TR-PINs)-mediated PDP, targeting epidermal growth factor receptor, transferrin receptor, and human epidermal growth factor receptor 2 we show light dose-dependent TR-PINs mediated cytotoxicity inhuman PDA Ccells (MIAPaCa-2),co-cultured with human pancreatic cancer-associated fibroblasts (PCAFs) in spheroids. Furthermore, TR-PINs induced the expression of heat shock proteins (Hsp60, Hsp70), Calreticulin, and high mobility group box 1 in a light dose and time-dependent manner.TR-PINs-mediated T cell activation was observed in co-cultures of immune cells with the MIA PaCa-2-PCAF spheroids. Both CD4+ T and CD8+ T cells showed light dose and time-dependant antitumor reactivity by upregulating degranulation marker CD107a and interferon-gamma post-PDP. Substantial tumor cell death in immune cell-spheroid co-cultures by day 3 shows the augmentation by antitumor T cell activation and their ability to recognize tumors for a light dose-dependent kill. These data confirm enhanced destruction of heterogeneous pancreatic spheroids mediated by PDP-induced phototoxicity, TME modulation and increased immunogenicity with targeted nanoconstructs.
Collapse
Affiliation(s)
- Pushpamali De Silva
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Shazia Bano
- Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Kenneth K. Wang
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Edward V. Maytin
- Departments of Dermatology and Biomedical Engineering, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Tayyaba Hasan
- Corresponding author: Tayyaba Hasan, Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, 40 Blossom Street, BAR 314A, Boston, MA, 02114, USA; and Division of Health Sciences and Technology, Massachusetts Institute of Technology, Harvard University, Cambridge, MA, 02139, USA,
| |
Collapse
|
504
|
Efficacy of Immune Checkpoint Inhibitors in Upper Tract Urothelial Carcinomas: Current Knowledge and Future Directions. Cancers (Basel) 2021; 13:cancers13174341. [PMID: 34503152 PMCID: PMC8431384 DOI: 10.3390/cancers13174341] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 08/21/2021] [Accepted: 08/24/2021] [Indexed: 12/13/2022] Open
Abstract
Upper tract urothelial carcinoma (UTUC) represents a rare and aggressive malignancy arising from the renal pelvis or ureter. It can develop sporadically or have a hereditary origin, such as Lynch syndrome, caused by DNA mismatch repair deficiency, leading to microsatellite instability phenotype. According to molecular characterization studies, UTUC presents different mutational profiles as compared to urinary bladder urothelial carcinomas. In particular, it has been reported that UTUC harbored a higher level of FGFR3 alterations associated with a T-cell depleted immune microenvironment. The therapeutic landscape in urothelial carcinoma is rapidly evolving, with immune checkpoint inhibitors forming part of the standard of care. A greater understanding of the molecular alterations and immune microenvironment leads to the development of new treatment combinations and targeted therapy. This review summarizes the available evidence concerning the use of immune checkpoint inhibitors and the biological rationale underlying their use in high-grade UTUC.
Collapse
|
505
|
Trojan J, Stintzing S, Haase O, Koch C, Ziegler P, Demes M, Jelas I. Complete Pathological Response After Neoadjuvant Short-Course Immunotherapy with Ipilimumab and Nivolumab in Locally Advanced MSI-H/dMMR Rectal Cancer. Oncologist 2021; 26:e2110-e2114. [PMID: 34431576 PMCID: PMC8649008 DOI: 10.1002/onco.13955] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 08/13/2021] [Indexed: 12/21/2022] Open
Abstract
Background Patients with colorectal carcinoma and high‐grade microsatellite instability (MSI‐H) or deficiency in mismatch repair (dMMR) exceptionally respond to immune checkpoint inhibitors (ICIs). ICIs are more active in treatment‐naïve patients than in patients with refractory MSI‐H/dMMR metastatic colorectal cancer and even more active in patients with locally advanced tumors. Material and Methods A 33‐year‐old male patient with Lynch syndrome was diagnosed with a locally advanced rectal cancer and refused standard neoadjuvant chemoradiation because of the potential harm of sexual dysfunction. MMR and microsatellite instability status were analyzed by immunohistochemistry and fragment length polymerase chain reaction followed by capillary electrophoresis. Results After MSI‐H/dMMR was confirmed, the patient was treated with ICIs (1 mg/kg ipilimumab at day 1 and 3 mg/kg nivolumab at day 1 and 15). A complete clinical response was documented at day 21 after start of treatment. The patient underwent a total mesorectal excision at day 30. In the extirpated tissue, a complete pathological response was confirmed. Conclusion In MSI‐H/dMMR locally advanced rectal cancer short‐course ICI treatment is highly effective and may be discussed in patients with dMMR locally advanced rectal cancer. Key Points Immune checkpoint inhibitors are more active in treatment‐naïve patients than in patients with refractory high‐grade microsatellite instability (MSI‐H)/deficiency in mismatch repair (dMMR) colorectal cancer. Standard neoadjuvant chemoradiation is less effective in MSI‐H/dMMR rectal cancer patients than in patients with proficient mismatch repair. A young patient with Lynch syndrome and MSI‐H/dMMR locally advanced rectal cancer refused chemoradiation in order to preserve his fertility. After neoadjuvant treatment with one dose of ipilimumab and two doses of nivolumab a complete clinical and pathological response was documented. Clinical trials are needed to first establish neoadjuvant treatment with immune checkpoint inhibitors in patients with locally advanced MSI‐H/dMMR rectal cancer and thereafter to evaluate organ‐preservation strategies.
This report describes the case of a 33‐year‐old male patient with Lynch syndrome who was diagnosed with locally advanced rectal cancer. Instead of chemoradiation, neoadjuvant treatment with immune checkpoint inhibitors was given.
Collapse
Affiliation(s)
- Jörg Trojan
- Department of Medicine I, Goethe-University, Frankfurt, Germany.,University Cancer Center, Goethe-University, Frankfurt, Germany
| | - Sebastian Stintzing
- Department of Hematology, Oncology, and Cancer Immunology, Campus Charité Mitte, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| | - Oliver Haase
- Department of Surgery, Charité - Universitaetsmedizin, Berlin, Germany
| | - Christine Koch
- Department of Medicine I, Goethe-University, Frankfurt, Germany.,University Cancer Center, Goethe-University, Frankfurt, Germany
| | - Paul Ziegler
- University Cancer Center, Goethe-University, Frankfurt, Germany.,Dr. Senckenberg Institute of Pathology, Goethe-University, Frankfurt, Germany
| | - Melanie Demes
- University Cancer Center, Goethe-University, Frankfurt, Germany.,Dr. Senckenberg Institute of Pathology, Goethe-University, Frankfurt, Germany
| | - Ivan Jelas
- Department of Hematology, Oncology, and Cancer Immunology, Campus Charité Mitte, Charité - Universitaetsmedizin Berlin, Berlin, Germany
| |
Collapse
|
506
|
Błach J, Wojas-Krawczyk K, Nicoś M, Krawczyk P. Failure of Immunotherapy-The Molecular and Immunological Origin of Immunotherapy Resistance in Lung Cancer. Int J Mol Sci 2021; 22:9030. [PMID: 34445735 PMCID: PMC8396490 DOI: 10.3390/ijms22169030] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 08/05/2021] [Accepted: 08/18/2021] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have a huge impact on clinical treatment results in non-small cell lung cancer (NSCLC). Blocking antibodies targeting programmed cell death protein 1 (PD-1), programmed cell death protein ligand 1 (PD-L1) or CTLA-4 (cytotoxic T cell antigen 4) have been developed and approved for the treatment of NSCLC patients. However, a large number of patients develop resistance to this type of treatment. Primary and secondary immunotherapy resistance are distinguished. No solid biomarkers are available that are appropriate to predict the unique sensitivity to immunotherapy. Knowledge of predictive markers involved in treatment resistance is fundamental for planning of new treatment combinations. Scientists focused research on the use of immunotherapy as an essential treatment in combination with other therapy strategies, which could increase cancer immunogenicity by generating tumor cells death and new antigen release as well as by targeting other immune checkpoints and tumor microenvironment. In the present review, we summarize the current knowledge of molecular bases underlying immunotherapy resistance and discuss the capabilities and the reason of different therapeutic combinations.
Collapse
Affiliation(s)
- Justyna Błach
- Department of Clinical Immunology, Medical University of Lublin, W. Chodźki 4A, 20-093 Lublin, Poland
- Department of Clinical Immunology, University Children Hospital of Cracow, 30-663 Cracow, Poland
| | - Kamila Wojas-Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczeswskiego 8, 20-954 Lublin, Poland; (K.W.-K.); (M.N.); (P.K.)
| | - Marcin Nicoś
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczeswskiego 8, 20-954 Lublin, Poland; (K.W.-K.); (M.N.); (P.K.)
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, Jaczeswskiego 8, 20-954 Lublin, Poland; (K.W.-K.); (M.N.); (P.K.)
| |
Collapse
|
507
|
MEM: An Algorithm for the Reliable Detection of Microsatellite Instability (MSI) on a Small NGS Panel in Colorectal Cancer. Cancers (Basel) 2021; 13:cancers13164203. [PMID: 34439357 PMCID: PMC8394433 DOI: 10.3390/cancers13164203] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Microsatellite instability (MSI) assessment has become a major issue in the management of colorectal cancer, with the recent approval of anti-PD1 immunotherapies in MSI-metastatic colorectal cancer. The reference PCR method (MSI-PCR) can be costly, time and tissue-consuming. However, NGS could facilitate the assessment of MSI status while simultaneously screening for targetable oncogenic mutations (KRAS, NRAS, BRAF) for any colorectal cancer, but the algorithms developed to date use a large number of microsatellites that have not been approved by international guidelines and which are generally incompatible with small NGS panels. We present the MEM algorithm, which mimics the interpretation of MSI-PCR data by a human operator to reliably assess MSI status using only five validated microsatellites (BAT-25, BAT-26, NR-21, NR-24 and NR-27). We demonstrated that the MEM algorithm was in perfect agreement with MSI-PCR results, in terms of both MSI status and individual microsatellite status, in a cohort of 146 patients. Abstract Purpose: MEM is an NGS algorithm that uses Expectation-Maximisation to detect the presence of unstable alleles from the NGS sequences of five microsatellites (BAT-25, BAT-26, NR-21, NR-24 and NR-27). The purpose of this study was to compare the MEM algorithm with a reference PCR method (MSI-PCR) and MisMatch Repair protein immunohistochemistry (MMR-IHC). Methods: FFPE colorectal cancer samples from 146 patients were analysed in parallel by MSI-PCR and NGS using the MEM algorithm. MMR-IHC results were available for 133 samples. Serial dilutions of an MSI positive control were performed to estimate the limit of detection. Results: the MEM algorithm was able to detect unstable alleles of each microsatellite with up to a 5% allelic fraction. Of the 146 samples, 28 (19.2%) were MSI in MSI-PCR. MEM algorithm results were in perfect agreement with those of MSI-PCR, at both MSI status and individual microsatellite level (Cohen’s kappa = 1). A high level of agreement was noted between MSI-PCR/MEM algorithm results and MMR-IHC results (Cohen’s kappa = 0.931). Conclusion: the MEM algorithm can determine the MSI status of colorectal cancer samples on a small NGS panel, using only five microsatellites approved by international guidelines, and can be combined with screening for targetable mutations.
Collapse
|
508
|
Pyo J, Park HJ. Treatment Efficacy of Immune Checkpoint Inhibitors for Patients with Advanced or Metastatic Colorectal Cancer: A Systematic Review and Meta-Analysis. J Clin Med 2021; 10:3599. [PMID: 34441895 PMCID: PMC8397178 DOI: 10.3390/jcm10163599] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/13/2022] Open
Abstract
The treatment efficacy of immune checkpoint inhibitors (ICIs) in colorectal cancer (CRC) has been reported heterogeneously across clinical trials. We conducted a systematic review and meta-analysis to evaluate the efficacy of ICIs in patients with advanced/metastatic CRC. Ovid-Medline was searched to identify clinical trials providing the efficacy outcomes of overall response rate (ORR) or disease control rate (DCR). The pooled ORR and DCR were estimated across all studies and subgroups. Meta-regression was performed to find the influencing factors for treatment efficacy. A total of thirty studies (1870 patients) were eligible. The overall ORR and DCR were 20.1% and 58.5%, respectively, but these results were heterogeneous across studies. Multivariate meta-regression revealed that microsatellite phenotype (odds ratio of MSI-H/dMMR versus MSS/pMMR: 1.67, p < 0.001) and drug regimen (odds ratio of monotherapy versus combination therapy: 1.07, p = 0.019) were the source of heterogeneity and also significantly influenced factors for the efficacy of the treatment. Although the efficacy of ICIs as a first-line therapy was higher than that of ICIs as the second- or more-line therapy (ORR: 51.5% vs. 13.4%, DCR: 85% vs. 49.5%), multivariate regression showed that the line of therapy was not a significant factor for the treatment efficacy. Our study suggests that the microsatellite phenotype and drug regimen, rather than the line of treatment, are the primary factors influencing the treatment response among advanced/metastatic CRC patients treated with an ICI-based regimen.
Collapse
Affiliation(s)
- Junhee Pyo
- Asan Medical Center, Department of Biomedical Engineering, College of Medicine, University of Ulsan, Seoul 05505, Korea;
| | - Hyo-Jung Park
- Asan Medical Center, Department of Radiology and Research Institute of Radiology, College of Medicine, University of Ulsan, Seoul 05505, Korea
| |
Collapse
|
509
|
Zhao S, Chen L, Zang Y, Liu W, Liu S, Teng F, Xue F, Wang Y. Endometrial cancer in Lynch syndrome. Int J Cancer 2021; 150:7-17. [PMID: 34398969 DOI: 10.1002/ijc.33763] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/05/2021] [Accepted: 08/09/2021] [Indexed: 12/11/2022]
Abstract
Lynch syndrome (LS) is an autosomal dominant inherited disease caused by germline pathogenic variants (PVs) in mismatch repair (MMR) genes. LS-associated endometrial cancer (LS-EC) is the most common extraintestinal sentinel cancer caused by germline PVs in MMR genes, including MLH1, MSH2, MSH6 and PMS2. The clinicopathologic features of LS-EC include early age of onset, lower body mass index (BMI), endometrioid carcinoma and lower uterine segment involvement. There has been significant progress in screening, diagnosis, surveillance, prevention and treatment of LS-EC. Many studies support universal screening for LS among patients with EC. Screening mainly involves a combination of traditional clinical criteria and molecular techniques, including MMR-immunohistochemistry (MMR-IHC), microsatellite instability (MSI) testing, MLH1 promoter methylation testing and gene sequencing. The effectiveness of endometrial biopsy and transvaginal ultrasound (TVS) for clinical monitoring of asymptomatic women with LS are uncertain yet. Preventive strategies include hysterectomy and bilateral salpingo-oophorectomy (BSO) as well as chemoprophylaxis using exogenous progestin or aspirin. Recent research has revealed the benefits of immunotherapy for LS-EC. The NCCN guidelines recommend pembrolizumab and nivolumab for treating patients with advanced or recurrent microsatellite instability-high (MSI-H)/mismatch repair-deficient (dMMR) EC.
Collapse
Affiliation(s)
- Shuangshuang Zhao
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Lingli Chen
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuqin Zang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenlu Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Shiqi Liu
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fei Teng
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.,Tianjin Key Laboratory of Female Reproductive Health and Eugenics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
510
|
Choucair K, Radford M, Bansal A, Park R, Saeed A. Advances in immune therapies for the treatment of microsatellite instability‑high/deficient mismatch repair metastatic colorectal cancer (Review). Int J Oncol 2021; 59:74. [PMID: 34396449 PMCID: PMC8360619 DOI: 10.3892/ijo.2021.5254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 07/23/2021] [Indexed: 11/29/2022] Open
Abstract
Microsatellite instability-high/deficient mismatch repair colorectal cancer (MSI-H/dMMR CRC) is a molecular subtype characterized by high-frequency mutations within DNA mismatch repair genes. Defects in the DNA mismatch repair machinery lead to subsequent frame-shift mutations, resulting in the generation of frame-shift peptides that serve as neoantigens. This has translated into exquisite sensitivity to immune checkpoint inhibitors (ICIs) and a significant clinical benefit from immune therapies in this patient population. The present article provides a comprehensive review of the advances in the field of immune therapies for MSI-H/dMMR metastatic CRC, with a focus on the major randomized clinical trials that led to Food and Drug Administration approval of specific ICIs for this population, a detailed review of the molecular background responsible for tumor response, as well as the mechanisms of resistance to ICI therapy. Finally, ongoing investigations of other immunotherapeutic strategies to address and overcome the challenges that currently limit response and long-term response to ICIs were presented.
Collapse
Affiliation(s)
- Khalil Choucair
- Department of Medicine, Kansas University School of Medicine, Wichita, KS 67214, USA
| | - Maluki Radford
- Department of Medicine, Kansas University Medical Center, Kansas City, KS 66205, USA
| | - Ajay Bansal
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Kansas Medical Center and Kansas Cancer Institute, Kansas City, KS 66205, USA
| | - Robin Park
- Department of Medicine, MetroWest Medical Center/Tufts University School of Medicine, Framingham, Massachusetts, MA 01702, USA
| | - Anwaar Saeed
- Department of Medicine, Division of Medical Oncology, Kansas University Cancer Center, Kansas City, KS 66205, USA
| |
Collapse
|
511
|
Russo A, Incorvaia L, Malapelle U, Del Re M, Capoluongo E, Vincenzi B, Chiari R, Cortesi L, Danesi R, Florena AM, Fontanini G, Gori S, Marchetti A, Normanno N, Pinto C, Sangiolo D, Silvestris N, Tagliaferri P, Tallini G, Cinieri S, Beretta GD. The Tumor-Agnostic Treatment For Patients with Solid Tumors: a Position Paper on behalf of the AIOM- SIAPEC/IAP-SIBIOC-SIF Italian Scientific Societies. Crit Rev Oncol Hematol 2021; 165:103436. [PMID: 34371157 DOI: 10.1016/j.critrevonc.2021.103436] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/04/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
The personalized medicine is in a rapidly evolving scenario. The identification of actionable mutations is revolutionizing the therapeutic landscape of tumors. The morphological and histological tumor features are enriched by the extensive genomic profiling, and the first tumor-agnostic drugs have been approved regardless of tumor histology, guided by predictive and druggable genetic alterations. This new paradigm of "mutational oncology", presents a great potential to change the oncologic therapeutic scenario, but also some critical aspects need to be underlined. A process governance is mandatory to ensure the genomic testing accuracy and homogeneity, the economic sustainability, and the regulatory issues, ultimately granting the possibility of translating this model in the "real world". In this position paper, based on experts' opinion, the AIOM-SIAPEC-IAP-SIBIOC-SIF Italian Scientific Societies revised the new agnostic biomarkers, the diagnostic technologies available, the current availability of agnostic drugs and their present indication.
Collapse
Affiliation(s)
- Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy.
| | - Lorena Incorvaia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Medical Oncology, University of Palermo, 90127, Palermo, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, 80138, Naples, Italy
| | - Marzia Del Re
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Ettore Capoluongo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università di Napoli Federico II, Via Pansini 5, 80131, Naples, Italy; CEINGE, Biotecnologie Avanzate, Via Gaetano Salvatore, 486, 80131, Naples, Italy
| | - Bruno Vincenzi
- Department of Medical Oncology, Campus Bio-Medico University, 00128, Rome, Italy
| | - Rita Chiari
- Medical Oncology, AULSS 6 Euganea, South Padova Hospital, Monselice, PD, Italy
| | - Laura Cortesi
- Department of Oncology and Hematology, Azienda Ospedaliero-Universitaria di Modena, Modena, Italy
| | - Romano Danesi
- Unit of Clinical Pharmacology and Pharmacogenetics, Department of Clinical and Experimental Medicine, University Hospital of Pisa, Pisa, Italy
| | - Ada Maria Florena
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Palermo, Italy
| | - Gabriella Fontanini
- Department of Surgical, Medical, Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Stefania Gori
- Department of Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | - Antonio Marchetti
- Center of Predictive Molecular Medicine, University-Foundation, CeSI Biotech Chieti, Italy
| | - Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131, Naples, Italy
| | - Carmine Pinto
- Medical Oncology Unit, Clinical Cancer Centre, IRCCS-AUSL di Reggio Emilia, Reggio Emilia, Italy
| | - Dario Sangiolo
- Department of Oncology, University of Torino, Turin, Italy; Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy; Department of Biomedical Sciences and Human Oncology, Department of Internal Medicine and Oncology (DIMO), University of Bari, Bari, Italy
| | - Pierosandro Tagliaferri
- Medical and Translational Oncology Unit, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100, Catanzaro, Italy
| | - Giovanni Tallini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna School of Medicine, Bologna, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| | | |
Collapse
|
512
|
Tumour mutational burden, microsatellite instability, and actionable alterations in metastatic colorectal cancer: Next-generation sequencing results of TRIBE2 study. Eur J Cancer 2021; 155:73-84. [PMID: 34365081 DOI: 10.1016/j.ejca.2021.06.037] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 11/23/2022]
Abstract
BACKGROUND We performed a comprehensive genomic profiling of tumour samples from metastatic colorectal cancer (mCRC) patients enrolled in the TRIBE2 study to assess the concordance among different techniques to evaluate mismatch repair (MMR) and microsatellite instability (MSI) status, to characterize tumours according to the tumour mutational burden (TMB) and explore the clinical relevance of different TMB cutpoints, and to investigate the prevalence of alterations actionable with targeted approaches or immune checkpoint inhibitors. MATERIAL AND METHODS Tumour samples of 296 (44%) of 679 enrolled patients underwent 592-gene DNA next-generation sequencing (NGS). MMR status was assessed by immunohistochemistry (MMR-IHC), and MSI status was assessed by NGS (MSI-NGS). TMB was defined as low, intermediate, or high if <7, 7-16, or ≥17 mutations/megabase (mut/Mb) were found. The performance of TMB to predict MSI status was tested by receiver operating characteristic (ROC) curve. Actionable alterations included BRAF V600E, KRAS G12C, POLE mutations, HER2 amplification and mutations, and MSI-H. RESULTS Of 216 paired cases, concordance between MMR-IHC and MSI-NGS was 98.6%. Among 11 TMB-high tumours, eight (73%) were MSI-H and three (27%) were microsatellite stable and harboured POLE or MSH6 mutations. High TMB had a trend for a better outcome than low/intermediate TMB (hazard ratio for overall survival 0.45, 95% confidence interval 0.28-1.33; P = 0.106). No interaction effect between TMB and treatment arm was observed. Seventeen mut/Mb was identified as the optimal threshold of TMB for predicting MSI status. Actionable alterations were found in 62 (21%) of 296 patients. CONCLUSIONS Genomic profiling provides an overview of the genomic landscape of mCRC in a single analysis, including actionable targets and markers of immune sensitivity.
Collapse
|
513
|
The role of molecular tests for adjuvant and post-surgical treatment in gynaecological cancers. Best Pract Res Clin Obstet Gynaecol 2021; 78:14-35. [PMID: 34456153 DOI: 10.1016/j.bpobgyn.2021.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 06/09/2021] [Indexed: 11/23/2022]
Abstract
The adjuvant and post-surgical treatment of gynaecological cancers has historically been guided by the estimation of relapse risk based on clinicopathological factors determined at the time of cancer diagnosis. The recent advancement of genomic and molecular characterisation of gynaecological cancers has begun to shift paradigms in the selection of adjuvant treatment strategy. Recent data regarding the predictive and/or prognostic value of molecular tests in the treatment of advanced ovarian cancer as well as early stage endometrial cancer have been the first such examples to enter adjuvant treatment guidelines for these diseases. In this article, we discuss the current state and future development of molecular assays for gynaecological cancers and how they impact upon treatment selection for ovarian, endometrial and cervical cancers in the post-surgical setting.
Collapse
|
514
|
Arrichiello G, Poliero L, Borrelli C, Paragliola F, Nacca V, Napolitano S, Corte CMD, Martini G, Martinelli E. Immunotherapy in colorectal cancer: is the long-awaited revolution finally happening? Cancer Treat Res Commun 2021; 28:100442. [PMID: 34391139 DOI: 10.1016/j.ctarc.2021.100442] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/21/2021] [Accepted: 07/25/2021] [Indexed: 12/30/2022]
Abstract
Immunotherapy has recently become a major treatment modality for several types of solid tumours, achieving remarkable and long-lasting remissions. In metastatic colorectal cancer patients (mCRC), immune checkpoint inhibitors (ICIs) were found to be effective as treatment for deficient mismatch repair (dMMR)/ microsatellite instability high (MSI-H) tumours and received regulatory approval for this indication. However, mCRC is a complex disease and dMMR/MSI-H tumours represent a minority of the cases; therefore, new strategies are needed to extend the benefits of immunotherapy to a larger population of patients. This review explores the immunological differences between dMMR/MSI-H and proficient mismatch repair (pMMR)/ microsatellite instability low (MSI-L) tumours, focuses on new proposed biomarkers to predict response to immunotherapy and illustrates results reported from the main clinical trials with immunotherapeutic agents in CRC, addressing the most promising approaches being currently developed.
Collapse
Affiliation(s)
- Gianluca Arrichiello
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Luca Poliero
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Carola Borrelli
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Fernando Paragliola
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Valeria Nacca
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Stefania Napolitano
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Carminia Maria Della Corte
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Giulia Martini
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy
| | - Erika Martinelli
- Oncologia Medica, Dipartimento di Medicina di Precisione, Università degli Studi della Campania "L. Vanvitelli", Naples, Italy.
| |
Collapse
|
515
|
Pietrantonio F, Lonardi S, Corti F, Infante G, Elez ME, Fakih M, Jayachandran P, Shah AT, Salati M, Fenocchio E, Salvatore L, Curigliano G, Cremolini C, Ambrosini M, Ros J, Intini R, Nappo F, Damian S, Morano F, Fucà G, Overman M, Miceli R. Nomogram to predict the outcomes of patients with microsatellite instability-high metastatic colorectal cancer receiving immune checkpoint inhibitors. J Immunother Cancer 2021; 9:e003370. [PMID: 34429334 PMCID: PMC8386222 DOI: 10.1136/jitc-2021-003370] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/01/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND The efficacy of immune checkpoint inhibitors (ICIs) in patients with microsatellite instability (MSI)-high metastatic colorectal cancer (mCRC) is unprecedented. A relevant proportion of subjects achieving durable disease control may be considered potentially 'cured', as opposed to patients experiencing primary ICI refractoriness or short-term clinical benefit. We developed and externally validated a nomogram to estimate the progression-free survival (PFS) and the time-independent event-free probability (EFP) in patients with MSI-high mCRC receiving ICIs. METHODS The PFS and EFP were estimated using a cure model fitted on a developing set of 163 patients and validated on a set of 146 patients with MSI-high mCRC receiving anti-programmed death (ligand)1 (PD-(L)1) ± anticytotoxic T-lymphocyte antigen 4 (CTLA-4) agents. A total of 23 putative prognostic factors were chosen and then selected using a random survival forest (RSF). The model performance in estimating PFS probability was evaluated by assessing calibration (internally-developing set and externally-validating set) and quantifying the discriminative ability (Harrell C index). RESULTS RFS selected five variables: ICI type (anti-PD-(L)1 monotherapy vs anti-CTLA-4 combo), ECOG PS (0 vs >0), neutrophil-to-lymphocyte ratio (≤3 vs >3), platelet count, and prior treatment lines. As both in the developing and validation series most PFS events occurred within 12 months, this was chosen as cut-point for PFS prediction. The combination of the selected variables allowed estimation of the 12-month PFS (focused on patients with low chance of being cured) and the EFP (focused on patients likely to be event-free at a certain point of their follow-up). ICI type was significantly associated with disease control, as patients receiving the anti-CTLA-4-combination experienced the best outcomes. The calibration of PFS predictions was good both in the developing and validating sets. The median value of the EFP (46%) allowed segregation of two prognostic groups in both the developing (PFS HR=3.73, 95% CI 2.25 to 6.18; p<0.0001) and validating (PFS HR=1.86, 95% CI 1.07 to 3.23; p=0.0269) sets. CONCLUSIONS A nomogram based on five easily assessable variables including ICI treatment was built to estimate the outcomes of patients with MSI-high mCRC, with the potential to assist clinicians in their clinical practice. The web-based system 'MSI mCRC Cure' was released.
Collapse
Affiliation(s)
- Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Lonardi
- Medical Oncology 3, Istituto Oncologico Veneto IOV-IRCSS, Padua, Italy
| | - Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Gabriele Infante
- Unit of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Maria Elena Elez
- Department of Medical Oncology, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Marwan Fakih
- Department of Medical Oncology, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Priya Jayachandran
- Division of Medical Oncology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | | | - Massimiliano Salati
- Division of Oncology, Department of Oncology and Hematology, University Hospital of Modena, PhD Clinical and Experimental Medicine (CEM), University of Modena and Reggio Emilia, Modena, Italy
| | - Elisabetta Fenocchio
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute FPO-IRCCS, Candiolo, Italy
| | - Lisa Salvatore
- Department of Medical Oncology, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Giuseppe Curigliano
- European Institute of Oncology (IEO), IRCCS, Milan, Italy; Department of Oncology and Hemato-oncology, University of Milan, Milan, Italy
| | - Chiara Cremolini
- Unit of Medical Oncology 2, Azienda Ospedaliero-Universitaria Pisana; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Margherita Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Javier Ros
- Department of Medical Oncology, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology (VHIO), Universitat Autonoma de Barcelona, Barcelona, Spain
| | - Rossana Intini
- Medical Oncology 1, Istituto Oncologico Veneto IOV-IRCCS, Padua, Italy
| | - Floriana Nappo
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Silvia Damian
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giovanni Fucà
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Michael Overman
- Department of Gastrointestinal Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Rosalba Miceli
- Unit of Clinical Epidemiology and Trial Organization, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| |
Collapse
|
516
|
Marchetti A, Barbareschi M, Barberis M, Buglioni S, Buttitta F, Fassan M, Fontanini G, Marchiò C, Papotti M, Pruneri G, Scarpa A, Stanta G, Tallini G, Troncone G, Veronese SM, Truini M, Sapino A. Real-World Data on NGS Diagnostics: a survey from the Italian Society of Pathology (SIAPeC) NGS Network. Pathologica 2021; 113:262-271. [PMID: 34463674 PMCID: PMC8488986 DOI: 10.32074/1591-951x-324] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2021] [Accepted: 07/26/2021] [Indexed: 12/31/2022] Open
Abstract
Next Generation Sequencing (NGS) is increasingly used in diagnostic centers for the assessment of genomic alterations to select patients for precision oncology. The Italian Society of Anatomic Pathology and Diagnostic Cytopathology (SIAPEC) through the Molecular Pathology and Predictive Medicine Study Group (PMMP) has been following the progressive development of centers that have adopted NGS technology in diagnostics over time. In July 2017, a study network on massive parallel sequencing was activated in Italy and recognized as the NGS SIAPeC National Network by the SIAPeC Scientific Society Board. Since then, activities have been implemented within the network that provide for alignment of laboratories through diagnostic concordance analysis and monitoring of centers adhering to the Network. Recently, considering the growing need for extended genomic analyses, the PMMP distributed a national survey to assess activities related to the use of genomic diagnostics in oncology within the NGS SIAPEC National Network. Thirty centers participated in the survey. Eighty percent of the centers are laboratories within Pathology Departments. The distribution of laboratories in the country, the diagnostic laboratory/population ratio, the staff dedicated, the type and number of sequencing and mechatronics platforms available, the genomic panels utilized, and the type and number of diagnostic tests carried out in the last year in each center, are reported. The centers were also asked whether they participated in a multidisciplinary Molecular Tumor Board (MTB) for management of patients. Thirty percent of the centers had a MTB that was ratified by regional decree. The professionals most frequently involved in the core team of the MTB are the pathologist, oncologist, molecular biologist, geneticist, pharmacologist, and bioinformatician. The data from this survey indicate that NGS diagnostics in Italy is still heterogeneous in terms of geographical distribution and the characteristics of laboratories and diagnostic test performed. The implementation of activities that favors harmonization, the logistics and the convergence of biological material in reference centers for molecular analyses is a priority for the development of a functional laboratory network.
Collapse
Affiliation(s)
- Antonio Marchetti
- Laboratory of Molecular Diagnostics, Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, Italy
| | | | - Massimo Barberis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Lombardy, Italy
| | - Simonetta Buglioni
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Fiamma Buttitta
- Laboratory of Molecular Diagnostics, Center for Advanced Studies and Technology (CAST), University of Chieti-Pescara, Italy
| | - Matteo Fassan
- Unit of Surgical Pathology, Department of Medicine (DIMED), University of Padua
- Veneto Institute of Oncology IRCCS, Padua, Italy
| | - Gabriella Fontanini
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| | - Caterina Marchiò
- Pathology Unit, Candiolo Cancer Institute, FPO-IRCCS, Candiolo (TO), Italy
| | - Mauro Papotti
- Pathology Unit, Department of Oncology, Città Della Salute e della Scienza Hospital, University of Turin, Turin, Italy
| | - Giancarlo Pruneri
- Department of Pathology and Laboratory Medicine, IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, and ARC-Net Research Centre, University and Hospital Trust of Verona, Verona, Italy
| | - Giorgio Stanta
- DSM - Department of Medical Sciences, University of Trieste, Trieste, Italy
| | - Giovanni Tallini
- Department of Medicine (Dipartimento di Medicina Specialistica, Diagnostica e Sperimentale, DIMES), University of Bologna, Italy
- Molecular Pathology Laboratory, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Giancarlo Troncone
- Division of Pathology, Department of Public Health, University of Naples Federico II, Naples, Italy
| | | | - Mauro Truini
- Pathological Anatomy Unit, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Anna Sapino
- Department of Surgical, Medical and Molecular Pathology and Critical Area, University of Pisa, Pisa, Italy
| |
Collapse
|
517
|
Alphones S, Chatterjee U, Singh A, Das A, Zameer L, Achari R, Bhattacharya A, Roy P. Immunohistochemical screening for mismatch repair protein deficiency in paediatric high-grade gliomas - institutional experience and review of literature. Childs Nerv Syst 2021; 37:2521-2530. [PMID: 34097097 DOI: 10.1007/s00381-021-05229-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 05/24/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Immunohistochemical (IHC) testing for mismatch repair (MMR) deficiency (MMRD) is used as a screening tool to identify microsatellite instability in various cancers (especially colon). This not only identifies hereditary cancer syndromes like Lynch and constitutional mismatch repair deficiency (CMMRD) but also aids in prognostication and prediction of sensitivity to checkpoint inhibitor drugs. There are very few reported studies on MMRD status of pediatric high-grade gliomas (pHGG) and none from the Indian subcontinent. The aim of this study is to evaluate the frequency of MMRD in pHGG and to assess if there is a need for universal screening with immunohistochemistry. METHODS Paraffin blocks of consecutive cases of pHGG (< 18 years) were retrieved from 2 centres, and IHC with four MMR antibodies - MLH1, PMS2, MSH2 and MSH6 - was performed using tissue microarray-based technique. RESULTS Three out of nine cases (33%) studied showed loss of staining. One case had loss of MSH2 and MSH6 confirmed by gene sequencing. Eight of the cases were glioblastoma. One case of IDH1-mutated anaplastic astrocytoma showed loss of MLH1 and PMS2 staining. Isolated PMS2 loss was noted in 1 case, where the non-tumour cells also showed loss of staining, indicative CMMRD syndrome. This patient had prior colon cancer with isolated PMS2 loss and responded to check-point inhibitor therapy with nivolumab. CONCLUSION Our study shows that the frequency of MMRD to be about one-third of pHGG. Universal IHC screening for MMRD in all pHGGs may benefit early diagnosis and play a role in therapeutic decisions. A larger multi-institutional study will help better assess the prevalence and treatment implications in MMRD tumours.
Collapse
Affiliation(s)
- Sheena Alphones
- Department of Pathology, Tata Medical Center, Kolkata, India
| | | | - Angad Singh
- Department of Pathology, Tata Medical Center, Kolkata, India
| | - Anirban Das
- Department of Pediatric Oncology, Tata Medical Center, Kolkata, India
| | - Lateef Zameer
- Department of Pathology, Tata Medical Center, Kolkata, India
| | - Rimpa Achari
- Department of Radiation Oncology, Tata Medical Center, Kolkata, India
| | | | - Paromita Roy
- Department of Pathology, Tata Medical Center, Kolkata, India.
| |
Collapse
|
518
|
Wang Y, Cuggia A, Pacis A, Boileau JC, Marcus VA, Gao ZH, Chong G, Foulkes WD, Zogopoulos G. Pancreatic Cancer Progression in a Patient With Lynch Syndrome Receiving Immunotherapy: A Cautionary Tale. J Natl Compr Canc Netw 2021; 19:883-887. [PMID: 34416708 DOI: 10.6004/jnccn.2021.7049] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 04/21/2021] [Indexed: 11/17/2022]
Abstract
Pancreatic ductal adenocarcinomas (PDACs) with DNA mismatch repair deficiency (MMRd) respond preferentially to immune checkpoint inhibitors (ICIs). However, a subset of MMRd PDACs does not respond to these agents. This report describes a patient with PDAC who experienced rapid disease progression suggestive of hyperprogressive disease. The case involved a 63-year-old man carrying a pathogenic germline PMS2 mutation who developed metastatic PDAC. His tumor showed isolated loss of PMS2 expression by immunohistochemistry (IHC). He was treated with pembrolizumab, but his disease rapidly progressed. Whole-genome and transcriptome sequencing of a liver metastasis biopsy, acquired at disease progression, showed a retained wild-type PMS2 allele and hallmarks of microsatellite stability, including low tumor mutational burden and low MSIsensor score. PCR-based microsatellite instability (MSI) testing of the treatment-naïve tumor showed microsatellite stability. The ICI-treated tumor had a lower density of CD8+ T-cell infiltration than the treatment-naïve tumor, which is contrary to the expected evolution with ICI responsiveness. Through this case and a review of the literature, we highlight the low penetrance of PMS2 germline mutations in PDAC and discuss pitfalls in ascertaining MMRd and MSI based on IHC testing alone. An orthogonal confirmatory assay is warranted in the presence of uncommon immunophenotypes, such as isolated PMS2 loss, to optimize selection of patients with PDAC for immunotherapy.
Collapse
Affiliation(s)
- Yifan Wang
- Department of Surgery, McGill University.,Research Institute of the McGill University Health Centre.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University
| | - Adeline Cuggia
- Research Institute of the McGill University Health Centre
| | - Alain Pacis
- The Rosalind and Morris Goodman Cancer Research Centre, McGill University.,Canadian Centre for Computational Genomics, McGill University and Genome Quebec Innovation Center
| | | | | | - Zu-Hua Gao
- Research Institute of the McGill University Health Centre.,Department of Pathology, McGill University
| | - George Chong
- Molecular Diagnostics Laboratory, Sir Mortimer B. Davis-Jewish General Hospital; and
| | - William D Foulkes
- Research Institute of the McGill University Health Centre.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - George Zogopoulos
- Department of Surgery, McGill University.,Research Institute of the McGill University Health Centre.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University.,Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
519
|
Piredda ML, Ammendola S, Sciammarella C, Turri G, Bagante F, Fassan M, Mafficini A, Mombello A, Cataldi S, Paolino G, Mattiolo P, Florena AM, Genna M, Fior F, Cheng L, Lawlor RT, Scarpa A, Pedrazzani C, Luchini C. Colorectal cancer with microsatellite instability: Right-sided location and signet ring cell histology are associated with nodal metastases, and extranodal extension influences disease-free survival. Pathol Res Pract 2021; 224:153519. [PMID: 34119815 DOI: 10.1016/j.prp.2021.153519] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/06/2021] [Accepted: 06/07/2021] [Indexed: 02/08/2023]
Abstract
Colorectal cancer (CRC) with microsatellite instability (MSI) accounts for 15-18 % of all CRCs and represents the category with the best prognosis. This study aimed at determining any possible clinical/pathological features associated with a higher risk of nodal metastasization in MSI-CRC, and at defining any possible prognostic moderators in this setting. All surgically resected CRCs of the last 20 years (mono-institutional series) with a PCR-based diagnosis of MSI, with and without nodal metastasis, have been retrieved for histological review, which was performed following WHO guidelines. Furthermore, the most important prognostic moderators have been investigated with a survival analysis. The study of 33 cases of MSI-CRCs with nodal metastasis highlighted a high fidelity of histology maintenance between primary tumors and matched nodal metastases. At survival analysis, the strongest prognostic variable in MSI-CRCs with nodal metastasis was the extranodal extension (multivariate analysis, HR: 14.4, 95 %CI: 1.46-140.9, p = 0.022). Furthermore, through a comparison between nodal positive (33 cases) and nodal negative (71 cases) MSI-CRCs, right-sided location (p < 0.0001), pT4 stage (p = 0.0004) and signet-ring histology (p = 0.0089) emerged as parameters more commonly associated with nodal metastasization. These findings shed new light on the biology of MSI-CRC and can be of help for the prognostic stratification of MSI-CRC patients.
Collapse
Affiliation(s)
- Maria L Piredda
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Serena Ammendola
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | | | - Giulia Turri
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Unit of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Fabio Bagante
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Unit of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Matteo Fassan
- Department of Medicine (DIMED), Surgical Pathology and Cytopathology Unit, University of Padua, Padua, Italy
| | - Andrea Mafficini
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Mombello
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Simone Cataldi
- Department of Sciences for Promotion of Health and Mother and Child Care, Anatomic Pathology, University of Palermo, Palermo, Italy
| | - Gaetano Paolino
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Ada M Florena
- Department of Sciences for Promotion of Health and Mother and Child Care, Anatomic Pathology, University of Palermo, Palermo, Italy
| | - Michele Genna
- Department of General Surgery, Unit of General and Obesity Surgery, University and Hospital Trust of Verona, Verona, Italy
| | - Francesca Fior
- Department of General Surgery, Unit of General and Obesity Surgery, University and Hospital Trust of Verona, Verona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rita T Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy; Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Corrado Pedrazzani
- Department of Surgical Sciences, Dentistry, Gynecology and Pediatrics, Unit of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy.
| |
Collapse
|
520
|
Chang YC, Chan MH, Li CH, Fang CY, Hsiao M, Chen CL. Exosomal Components and Modulators in Colorectal Cancer: Novel Diagnosis and Prognosis Biomarkers. Biomedicines 2021; 9:biomedicines9080931. [PMID: 34440135 PMCID: PMC8391321 DOI: 10.3390/biomedicines9080931] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/23/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
The relatively high incidence and mortality rates for colorectal carcinoma (CRC) make it a formidable malignant tumor. Comprehensive strategies have been applied to predict patient survival and diagnosis. Various clinical regimens have also been developed to improve the therapeutic outcome. Extracellular vesicles (EVs) are recently proposed cellular structures that can be produced by natural or artificial methods and have been extensively studied. In addition to their innate functions, EVs can be manipulated to be drug carriers and exert many biological functions. The composition of EVs, their intravesicular components, and the surrounding tumor microenvironment are closely related to the development of colorectal cancer. Determining the expression profiles of exocytosis samples and using them as indicators for selecting effective combination therapy is an indispensable direction for EV study and should be regarded as a novel prediction platform in addition to cancer stage, prognosis, and other clinical assessments. In this review, we summarize the function, regulation, and application of EVs in the colon cancer research field. We provide an update on and discuss potential values for clinical applications of EVs. Moreover, we illustrate the specific markers, mediators, and genetic alterations of EVs in colorectal carcinogenesis. Furthermore, we outline the vital markers present in the EVs and discuss their plausible uses in colon cancer patient therapy in combination with the currently used clinical strategies. The development and application of these EVs will significantly improve the accuracy of diagnosis, lead to more precise prognoses, and may lead to the improved treatment of colorectal cancer.
Collapse
Affiliation(s)
- Yu-Chan Chang
- Department of Biomedical Imaging and Radiological Science, National Yang-Ming University, Taipei 112, Taiwan;
- Department of Biomedical Imaging and Radiological Science, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ming-Hsien Chan
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
| | - Chien-Hsiu Li
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
| | - Chih-Yeu Fang
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli 350, Taiwan;
| | - Michael Hsiao
- Genomics Research Center, Academia Sinica, Taipei 115, Taiwan; (M.-H.C.); (C.-H.L.)
- Department of Biochemistry, Kaohsiung Medical University, Kaohsiung 807, Taiwan
- Correspondence: (M.H.); (C.-L.C.); Tel.: +886-2-2787-1243 (M.H.); +886-2-2736-1661 (ext. 3139) (C.-L.C.); Fax: +886-2-2789-9931 (M.H.)
| | - Chi-Long Chen
- Department of Pathology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
- Department of Pathology, Taipei Medical University Hospital, Taipei 110, Taiwan
- Correspondence: (M.H.); (C.-L.C.); Tel.: +886-2-2787-1243 (M.H.); +886-2-2736-1661 (ext. 3139) (C.-L.C.); Fax: +886-2-2789-9931 (M.H.)
| |
Collapse
|
521
|
Auclin E, Vuagnat P, Smolenschi C, Taieb J, Adeva J, Nebot-Bral L, Garcia de Herreros M, Vidal Tocino R, Longo-Muñoz F, El Dakdouki Y, Martín-Romano P, Gaba L, Saurí T, Oliveres H, Castañón E, Garcia-Carbonero R, Besse B, Massard C, Mezquita L, Hollebecque A. Association of the Lung Immune Prognostic Index with Immunotherapy Outcomes in Mismatch Repair Deficient Tumors. Cancers (Basel) 2021; 13:3776. [PMID: 34359675 PMCID: PMC8345164 DOI: 10.3390/cancers13153776] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
Background: MSI-H/dMMR is considered the first predictive marker of efficacy for immune checkpoint inhibitors (ICIs). However, around 39% of cases are refractory and additional biomarkers are needed. We explored the prognostic value of pretreatment LIPI in MSI-H/dMMR patients treated with ICIs, including identification of fast-progressors. Methods: A multicenter retrospective study of patients with metastatic MSI-H/dMMR tumors treated with ICIs between April 2014 and May 2019 was performed. LIPI was calculated based on dNLR > 3 and LDH > upper limit of normal. LIPI groups were good (zero factors), intermediate (one factor) and poor (two factors). The primary endpoint was overall survival (OS), including the fast-progressor rate (OS < 3 months). Results: A total of 151 patients were analyzed, mainly female (59%), with median age 64 years, performance status (PS) 0 (42%), and sporadic dMMR status (68%). ICIs were administered as first or second-line for 59%. The most frequent tumor types were gastrointestinal (66%) and gynecologic (22%). LIPI groups were good (47%), intermediate (43%), and poor (10%). The median follow-up was 32 months. One-year OS rates were 81.0%, 67.1%, and 21.4% for good, intermediate, and poor-risk groups (p < 0.0001). After adjustment for tumor site, metastatic sites and PS, LIPI remained independently associated with OS (HR, poor-LIPI: 3.50, 95%CI: 1.46-8.40, p = 0.02. Overall, the fast-progressor rate was 16.0%, and 35.7% with poor-LIPI vs. 7.5% in the good-LIPI group (p = 0.02). Conclusions: LIPI identifies dMMR patients who do not benefit from ICI treatment, particularly fast-progressors. LIPI should be included as a stratification factor for future trials.
Collapse
Affiliation(s)
- Edouard Auclin
- Gastrointestinal and Medical Oncology Department, Hôpital Européen Georges Pompidou, Université de Paris, 75015 Paris, France; (E.A.); (J.T.)
| | - Perrine Vuagnat
- Early Drug Development Department, Institut Gustave Roussy, 94805 Villejuif, France; (P.V.); (C.S.); (Y.E.D.); (P.M.-R.); (C.M.); (A.H.)
| | - Cristina Smolenschi
- Early Drug Development Department, Institut Gustave Roussy, 94805 Villejuif, France; (P.V.); (C.S.); (Y.E.D.); (P.M.-R.); (C.M.); (A.H.)
| | - Julien Taieb
- Gastrointestinal and Medical Oncology Department, Hôpital Européen Georges Pompidou, Université de Paris, 75015 Paris, France; (E.A.); (J.T.)
| | - Jorge Adeva
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Imas 12, UCM, 28041 Madrid, Spain; (J.A.); (R.G.-C.)
| | - Laetitia Nebot-Bral
- UMR9019 Genome Integrity and Cancers, Gustave Roussy Cancer Campus, 94805 Villejuif, France;
- Paris Saclay, Paris Sud University Orsay, 91400 Orsay, France
| | - Marta Garcia de Herreros
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (M.G.d.H.); (L.G.); (T.S.); (H.O.)
| | - Rosario Vidal Tocino
- Medical Oncology Department, Hospital Universitario de Salamanca, IBSAL, 37007 Salamanca, Spain;
| | - Federico Longo-Muñoz
- Medical Oncology Department, Hospital Universitario Ramon y Cajal, IRYCIS, CIBERONC, 28034 Madrid, Spain;
| | - Yola El Dakdouki
- Early Drug Development Department, Institut Gustave Roussy, 94805 Villejuif, France; (P.V.); (C.S.); (Y.E.D.); (P.M.-R.); (C.M.); (A.H.)
| | - Patricia Martín-Romano
- Early Drug Development Department, Institut Gustave Roussy, 94805 Villejuif, France; (P.V.); (C.S.); (Y.E.D.); (P.M.-R.); (C.M.); (A.H.)
| | - Lydia Gaba
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (M.G.d.H.); (L.G.); (T.S.); (H.O.)
- Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, 08036 Barcelona, Spain
| | - Tamara Saurí
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (M.G.d.H.); (L.G.); (T.S.); (H.O.)
- Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, 08036 Barcelona, Spain
| | - Helena Oliveres
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (M.G.d.H.); (L.G.); (T.S.); (H.O.)
- Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, 08036 Barcelona, Spain
| | | | - Rocio Garcia-Carbonero
- Medical Oncology Department, Hospital Universitario 12 de Octubre, Imas 12, UCM, 28041 Madrid, Spain; (J.A.); (R.G.-C.)
| | - Benjamin Besse
- Medical Oncology, Institut Gustave Roussy, 94805 Villejuif, France;
| | - Christophe Massard
- Early Drug Development Department, Institut Gustave Roussy, 94805 Villejuif, France; (P.V.); (C.S.); (Y.E.D.); (P.M.-R.); (C.M.); (A.H.)
| | - Laura Mezquita
- Medical Oncology Department, Hospital Clinic of Barcelona, 08036 Barcelona, Spain; (M.G.d.H.); (L.G.); (T.S.); (H.O.)
- Translational Genomics and Targeted Therapies in Solid Tumors, IDIBAPS, 08036 Barcelona, Spain
- Medical Oncology, Institut Gustave Roussy, 94805 Villejuif, France;
| | - Antoine Hollebecque
- Early Drug Development Department, Institut Gustave Roussy, 94805 Villejuif, France; (P.V.); (C.S.); (Y.E.D.); (P.M.-R.); (C.M.); (A.H.)
| |
Collapse
|
522
|
Evaluation of Micro Satellite Instability and Mismatch Repair Status in Different Solid Tumors: A Multicenter Analysis in a Real World Setting. Cells 2021. [PMID: 34440647 DOI: 10.3390/cells1008187828] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs) play a key role in the treatment of advanced stage colorectal cancer (CRC) patients featuring a deficient DNA mismatch repair (dMMR) system or a high microsatellite instability (MSI-H) profile. However, beyond the established role in CRC patients, ICIs have highly proven efficacy in other solid tumors featuring MSI-H/dMMR status represented by endometrial, gastric, ovarian, prostatic, and pancreatic carcinomas (EC, GC, OC, PrC, and PaC). Our aim was to compare the concordance rates among the Idylla™ MSI test, TapeStation 4200, and immunohistochemical (IHC) analysis in assessing MSI-H/dMMR status in EC, GC, OC, PrC, and PaC patients. The Sanger sequencing-based Titano MSI test was used in discordant cases. One hundred and eighty-five cases (n = 40 PrC, n = 39 GC, n = 38 OC, n = 35 PaC, and n = 33 EC) were retrospectively selected. MMR protein expression was evaluated by IHC. After DNA quality and quantity evaluations, the IdyllaTM and TapeStation 4200 platforms were adopted for the evaluation of MSI status. Remarkably, compared to IHC, the Idylla™ platform achieved a global concordance rate of 94.5% (154/163) for the microsatellite stable (MSS)/proficient MMR (pMMR) cases and 77.3% (17/22) for the MSI-H/dMMR cases. Similarly, a global concordance rate of 91.4% (149/163) and 68.2% (15/22) for MSS/pMMR and MSI-H/dMMR cases was also identified between IHC and the TapeStation 4200 microfluidic system. In addition, a global concordance of 93.1% (148/159) and 69.2% (18/26) for MSS/pMMR and MSI-H/dMMR cases was observed between the Idylla™ and TapeStation 4200 platforms. Discordant cases were analyzed using the Titano MSI kit. Overall, our data pinpointed a central role for molecular techniques in the diagnostic evaluation of dMMR/MSI-H status not only in CRC patients but also in other types of solid tumors.
Collapse
|
523
|
Malapelle U, Parente P, Pepe F, De Luca C, Pisapia P, Sgariglia R, Nacchio M, Gragnano G, Russo G, Conticelli F, Bellevicine C, Vigliar E, Iaccarino A, Covelli C, Balistreri M, Clemente C, Perrone G, Danza A, Scaramuzzi F, Fassan M, Troncone G, Graziano P. Evaluation of Micro Satellite Instability and Mismatch Repair Status in Different Solid Tumors: A Multicenter Analysis in a Real World Setting. Cells 2021; 10:1878. [PMID: 34440647 PMCID: PMC8391221 DOI: 10.3390/cells10081878] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/21/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022] Open
Abstract
Immune-checkpoint inhibitors (ICIs) play a key role in the treatment of advanced stage colorectal cancer (CRC) patients featuring a deficient DNA mismatch repair (dMMR) system or a high microsatellite instability (MSI-H) profile. However, beyond the established role in CRC patients, ICIs have highly proven efficacy in other solid tumors featuring MSI-H/dMMR status represented by endometrial, gastric, ovarian, prostatic, and pancreatic carcinomas (EC, GC, OC, PrC, and PaC). Our aim was to compare the concordance rates among the Idylla™ MSI test, TapeStation 4200, and immunohistochemical (IHC) analysis in assessing MSI-H/dMMR status in EC, GC, OC, PrC, and PaC patients. The Sanger sequencing-based Titano MSI test was used in discordant cases. One hundred and eighty-five cases (n = 40 PrC, n = 39 GC, n = 38 OC, n = 35 PaC, and n = 33 EC) were retrospectively selected. MMR protein expression was evaluated by IHC. After DNA quality and quantity evaluations, the IdyllaTM and TapeStation 4200 platforms were adopted for the evaluation of MSI status. Remarkably, compared to IHC, the Idylla™ platform achieved a global concordance rate of 94.5% (154/163) for the microsatellite stable (MSS)/proficient MMR (pMMR) cases and 77.3% (17/22) for the MSI-H/dMMR cases. Similarly, a global concordance rate of 91.4% (149/163) and 68.2% (15/22) for MSS/pMMR and MSI-H/dMMR cases was also identified between IHC and the TapeStation 4200 microfluidic system. In addition, a global concordance of 93.1% (148/159) and 69.2% (18/26) for MSS/pMMR and MSI-H/dMMR cases was observed between the Idylla™ and TapeStation 4200 platforms. Discordant cases were analyzed using the Titano MSI kit. Overall, our data pinpointed a central role for molecular techniques in the diagnostic evaluation of dMMR/MSI-H status not only in CRC patients but also in other types of solid tumors.
Collapse
Affiliation(s)
- Umberto Malapelle
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Paola Parente
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (C.C.); (G.P.); (A.D.); (F.S.); (P.G.)
| | - Francesco Pepe
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Caterina De Luca
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Pasquale Pisapia
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Roberta Sgariglia
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Mariantonia Nacchio
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Gianluca Gragnano
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Gianluca Russo
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Floriana Conticelli
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Claudio Bellevicine
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Elena Vigliar
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Antonino Iaccarino
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Claudia Covelli
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (C.C.); (G.P.); (A.D.); (F.S.); (P.G.)
| | - Mariangela Balistreri
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.B.); (M.F.)
| | - Celeste Clemente
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (C.C.); (G.P.); (A.D.); (F.S.); (P.G.)
| | - Giovanni Perrone
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (C.C.); (G.P.); (A.D.); (F.S.); (P.G.)
| | - Angela Danza
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (C.C.); (G.P.); (A.D.); (F.S.); (P.G.)
| | - Fabio Scaramuzzi
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (C.C.); (G.P.); (A.D.); (F.S.); (P.G.)
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, 35128 Padua, Italy; (M.B.); (M.F.)
- Veneto Institute of Oncology, IOV-IRCCS, 35128 Padua, Italy
| | - Giancarlo Troncone
- Department of Public Health, University of Naples Federico II, 80131 Naples, Italy; (U.M.); (F.P.); (C.D.L.); (P.P.); (R.S.); (M.N.); (G.G.); (G.R.); (F.C.); (C.B.); (E.V.); (A.I.)
| | - Paolo Graziano
- Unit of Pathology, Fondazione IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo, 71013 Foggia, Italy; (P.P.); (C.C.); (C.C.); (G.P.); (A.D.); (F.S.); (P.G.)
| |
Collapse
|
524
|
Wu C, Duan Y, Gong S, Kallendrusch S, Schopow N, Osterhoff G. Integrative and Comprehensive Pancancer Analysis of Regulator of Chromatin Condensation 1 (RCC1). Int J Mol Sci 2021; 22:ijms22147374. [PMID: 34298996 PMCID: PMC8305170 DOI: 10.3390/ijms22147374] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/23/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Regulator of Chromatin Condensation 1 (RCC1) is the only known guanine nucleotide exchange factor that acts on the Ras-like G protein Ran and plays a key role in cell cycle regulation. Although there is growing evidence to support the relationship between RCC1 and cancer, detailed pancancer analyses have not yet been performed. In this genome database study, based on The Cancer Genome Atlas, Genotype-Tissue Expression and Gene Expression Omnibus databases, the potential role of RCC1 in 33 tumors' entities was explored. The results show that RCC1 is highly expressed in most human malignant neoplasms in contrast to healthy tissues. RCC1 expression is closely related to the prognosis of a broad variety of tumor patients. Enrichment analysis showed that some tumor-related pathways such as "cell cycle" and "RNA transport" were involved in the functional mechanism of RCC1. In particular, the conducted analysis reveals the relation of RCC1 to multiple immune checkpoint genes and suggests that the regulation of RCC1 is closely related to tumor infiltration of cancer-associated fibroblasts and CD8+ T cells. Coherent data demonstrate the association of RCC1 with the tumor mutation burden and microsatellite instability in various tumors. These findings provide new insights into the role of RCC1 in oncogenesis and tumor immunology in various tumors and indicate its potential as marker for therapy prognosis and targeted treatment strategies.
Collapse
Affiliation(s)
- Changwu Wu
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (C.W.); (S.K.); (N.S.)
| | - Yingjuan Duan
- Faculty of Chemistry and Mineralogy, University of Leipzig, 04103 Leipzig, Germany;
| | - Siming Gong
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (C.W.); (S.K.); (N.S.)
- Correspondence:
| | - Sonja Kallendrusch
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (C.W.); (S.K.); (N.S.)
| | - Nikolas Schopow
- Institute of Anatomy, University of Leipzig, Liebigstraße 13, 04103 Leipzig, Germany; (C.W.); (S.K.); (N.S.)
- Sarcoma Center, Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, 04103 Leipzig, Germany;
| | - Georg Osterhoff
- Sarcoma Center, Department of Orthopedics, Trauma and Plastic Surgery, University Hospital Leipzig, 04103 Leipzig, Germany;
| |
Collapse
|
525
|
Criscitiello C, Guerini-Rocco E, Viale G, Fumagalli C, Sajjadi E, Venetis K, Piciotti R, Invernizzi M, Malapelle U, Fusco N. Immunotherapy in Breast Cancer Patients: A Focus on the Use of the Currently Available Biomarkers in Oncology. Anticancer Agents Med Chem 2021; 22:787-800. [PMID: 34229592 DOI: 10.2174/1871520621666210706144112] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 01/23/2021] [Accepted: 02/01/2021] [Indexed: 11/22/2022]
Abstract
Immune checkpoint inhibitors (ICIs) have remarkably modified the way solid tumors are managed, including breast cancer. Unfortunately, only a relatively small number of breast cancer patients significantly respond to these treatments. To maximize the immunotherapy benefit in breast cancer, several efforts are currently being put forward for the identification of i) the best therapeutic strategy (i.e. ICI monotherapy or in association with chemotherapy, radiotherapy, or other drugs); ii) the optimal timing for administration (e.g. early/advanced stage of disease; adjuvant/neoadjuvant setting); iii) the most effective and reliable predictive biomarkers of response (e.g. tumor-infiltrating lymphocytes, programmed death-ligand 1, microsatellite instability associated with mismatch repair deficiency, and tumor mutational burden). This article reviews the impacts and gaps in the characterization of immune-related biomarkers raised by clinical and translational research studies with immunotherapy treatments. Particular emphasis has been put on the documented evidence of significant clinical benefits of ICI in different randomized clinical trials, along with preanalytical and analytical issues in predictive biomarkers pathological assessment.
Collapse
Affiliation(s)
| | | | - Giulia Viale
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Caterina Fumagalli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Elham Sajjadi
- Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | | | - Roberto Piciotti
- Department of Oncology and Hemato-Oncology, University of Milan, Italy
| | - Marco Invernizzi
- Physical and Rehabilitative Medicine, Department of Health Sciences, University of Eastern Piedmont, Viale Piazza D'Armi 1, Novara, Italy
| | - Umberto Malapelle
- Department of Public Health, University of Naples Federico II, Naples, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
| |
Collapse
|
526
|
Pursuit of Gene Fusions in Daily Practice: Evidence from Real-World Data in Wild-Type and Microsatellite Instable Patients. Cancers (Basel) 2021; 13:cancers13133376. [PMID: 34282766 PMCID: PMC8269381 DOI: 10.3390/cancers13133376] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/26/2022] Open
Abstract
Agnostic biomarkers such as gene fusions allow to address cancer patients to targeted therapies; however, the low prevalence of these alterations across common malignancies poses challenges and needs a feasible and sensitive diagnostic process. RNA-based targeted next generation sequencing was performed on 125 samples of patients affected either by colorectal carcinoma, melanoma, or lung adenocarcinoma lacking genetic alterations in canonical driver genes, or by a colorectal carcinoma with microsatellite instability. Gene fusion rates were compared with in silico data from MSKCC datasets. For NTRK gene fusion detection we also employed a multitarget qRT-PCR and pan-TRK immunohistochemistry. Gene fusions were detected in 7/55 microsatellite instable colorectal carcinomas (12.73%), and in 4/70 of the "gene driver free" population (5.71%: 3/28 melanomas, 10.7%, and 1/12 lung adenocarcinomas, 8.3%). Fusion rates were significantly higher compared with the microsatellite stable and "gene driver positive" MSKCC cohorts. Pan-TRK immunohistochemistry showed 100% sensitivity, 91.7% specificity, and the occurrence of heterogeneous and/or subtle staining patterns. The enrichment of gene fusions in this "real-world" cohort highlights the feasibility of a workflow applicable in clinical practice. The heterogeneous expression in NTRK fusion positive tumours unveils challenging patterns to recognize and raises questions on the effective translation of the chimeric protein.
Collapse
|
527
|
D'Alessio A, Cammarota A, Prete MG, Pressiani T, Rimassa L. The evolving treatment paradigm of advanced hepatocellular carcinoma: putting all the pieces back together. Curr Opin Oncol 2021; 33:386-394. [PMID: 33867478 DOI: 10.1097/cco.0000000000000744] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW The therapeutic landscape of advanced hepatocellular carcinoma (HCC) has become notably complex in recent years. With this review, we aimed to put the most recent findings in perspective and tried to delineate the rapidly changing treatment algorithm. RECENT FINDINGS The combination of atezolizumab and bevacizumab has become the new first-line standard of care treatment for unresectable HCC after the positive results of the phase 3 IMbrave150 study. Nivolumab monotherapy failed to demonstrate advantage versus sorafenib in the CheckMate 459 trial, while two different therapeutic strategies (sintilimab and bevacizumab biosimilar and donafenib) outperformed sorafenib in two phase 2/3 studies conducted in the Chinese population. Several immunotherapy combinations are currently under study in large phase 3 trials after promising results in earlier phase studies. About further lines of treatment, the combination of ipilimumab and nivolumab was approved for sorafenib-pretreated patients after the positive results of the phase 1/2 CheckMate 040 study and apatinib was proven effective in the Chinese population in a phase 2/3 study, while pembrolizumab as monotherapy did not show statistically significant superiority when compared with placebo in the KEYNOTE-240 study. SUMMARY Because of the growing complexity of advanced HCC treatment, the implementation of predictive biomarkers of response is eagerly needed.
Collapse
Affiliation(s)
- Antonio D'Alessio
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Antonella Cammarota
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Maria Giuseppina Prete
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Tiziana Pressiani
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Lorenza Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele
- Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
528
|
Qi R, Zhao Y, Guo Q, Mi X, Cheng M, Hou W, Zheng H, Hua B. Exosomes in the lung cancer microenvironment: biological functions and potential use as clinical biomarkers. Cancer Cell Int 2021; 21:333. [PMID: 34193120 PMCID: PMC8247080 DOI: 10.1186/s12935-021-01990-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 05/22/2021] [Indexed: 12/14/2022] Open
Abstract
Lung cancer is one of the most common malignant tumours worldwide. however, emerging immunotherapy and targeted therapies continue to show limited efficacy. In the search for new targets for lung cancer treatment, exosomes have become a major focus of research. Exosomes play an important role in the tumour microenvironment (TME) of lung cancer and affect invasion, metastasis, and treatment responses. This review describes our current understanding of the release of exosomes derived from different cells in the TME, the effects of exosomes on T/Tregs, myeloid-derived suppressor cells, tumour-associated macrophages, dendritic cells, and natural killer cells, and the role of exosomes in the endothelial–mesenchymal transition, angiogenesis, and cancer-associated fibroblasts. In particular, this review focuses on the potential clinical applications of exosomes in the lung cancer microenvironment and their prognostic and diagnostic value.
Collapse
Affiliation(s)
- Runzhi Qi
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China
| | - Yuwei Zhao
- Beijing University of Chinese Medicine, Chaoyang District, Beijing, China
| | - Qiujun Guo
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China
| | - Xue Mi
- Shaanxi University of Chinese Medicine, Qindu District, Xianyang, Shaanxi, China
| | - Mengqi Cheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China
| | - Wei Hou
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China
| | - Honggang Zheng
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China.
| | - Baojin Hua
- Department of Oncology, Guang'anmen Hospital, China Academy of Chinese Medicine Sciences, Xicheng District, Beijing, China.
| |
Collapse
|
529
|
Rizzo A, Ricci AD, Gadaleta-Caldarola G. MSI-H/dMMR and cancer immunotherapy: current state and future implications. EXPERT REVIEW OF PRECISION MEDICINE AND DRUG DEVELOPMENT 2021. [DOI: 10.1080/23808993.2021.1946391] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Alessandro Rizzo
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italia
- Medical Oncology Unit, “Mons. R. Dimiccoli” Hospital, Barletta, Italy
| | - Angela Dalia Ricci
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italia
- Medical Oncology Unit, “Mons. R. Dimiccoli” Hospital, Barletta, Italy
| | | |
Collapse
|
530
|
Grassi E, Corbelli J, Papiani G, Barbera MA, Gazzaneo F, Tamberi S. Current Therapeutic Strategies in BRAF-Mutant Metastatic Colorectal Cancer. Front Oncol 2021; 11:601722. [PMID: 34249672 PMCID: PMC8262685 DOI: 10.3389/fonc.2021.601722] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 05/18/2021] [Indexed: 01/02/2023] Open
Abstract
Around 8–12% of patients with advanced colon rectal cancer (CRC) present with BRAF alterations, in particular V600E mutation, which is associated with right-side, poorly differentiated and mucinous type tumors. The presence of BRAF mutation (BRAF-mt) has been identified as a hallmark of poor prognosis and treatment optimization in this patient subgroup is an important goal. Currently, the standard of care is an aggressive strategy involving triplet chemotherapy and anti-VEGF agents, but new therapeutic approaches are emerging. Very promising results have been obtained with targeted therapy combinations, such as anti-BRAF agents plus anti-EGFR agents. Furthermore, around 60% of BRAF-mt patients show a strong association with high microsatellite instability (MSI-H) and immune checkpoint inhibitors could represent the new standard of care for this subgroup. The focus of this review is to summarize current strategies for BRAF-mt CRC treatment and highlight new therapeutic options.
Collapse
Affiliation(s)
- Elisa Grassi
- Department of Oncology, Infermi's Hospital, Azienda Unità Sanitaria Locale (AUSL) della Romagna, Faenza, Italy
| | - Jody Corbelli
- Department of Oncology, Infermi's Hospital, Azienda Unità Sanitaria Locale (AUSL) della Romagna, Faenza, Italy
| | - Giorgio Papiani
- Department of Oncology, Infermi's Hospital, Azienda Unità Sanitaria Locale (AUSL) della Romagna, Faenza, Italy
| | - Maria Aurelia Barbera
- Department of Oncology, Infermi's Hospital, Azienda Unità Sanitaria Locale (AUSL) della Romagna, Faenza, Italy
| | - Federica Gazzaneo
- Department of Oncology, Infermi's Hospital, Azienda Unità Sanitaria Locale (AUSL) della Romagna, Faenza, Italy
| | - Stefano Tamberi
- Department of Oncology, Infermi's Hospital, Azienda Unità Sanitaria Locale (AUSL) della Romagna, Faenza, Italy
| |
Collapse
|
531
|
Lawlor RT, Mattiolo P, Mafficini A, Hong SM, Piredda ML, Taormina SV, Malleo G, Marchegiani G, Pea A, Salvia R, Kryklyva V, Shin JI, Brosens LA, Milella M, Scarpa A, Luchini C. Tumor Mutational Burden as a Potential Biomarker for Immunotherapy in Pancreatic Cancer: Systematic Review and Still-Open Questions. Cancers (Basel) 2021; 13:3119. [PMID: 34206554 PMCID: PMC8269341 DOI: 10.3390/cancers13133119] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/13/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023] Open
Abstract
Tumor mutational burden (TMB) is a numeric index that expresses the number of mutations per megabase (muts/Mb) harbored by tumor cells in a neoplasm. TMB can be determined using different approaches based on next-generation sequencing. In the case of high values, it indicates a potential response to immunotherapy. In this systematic review, we assessed the potential predictive role of high-TMB in pancreatic ductal adenocarcinoma (PDAC), as well as the histo-molecular features of high-TMB PDAC. High-TMB appeared as a rare but not-negligible molecular feature in PDAC, being present in about 1.1% of cases. This genetic condition was closely associated with mucinous/colloid and medullary histology (p < 0.01). PDAC with high-TMB frequently harbored other actionable alterations, with microsatellite instability/defective mismatch repair as the most common. Immunotherapy has shown promising results in high-TMB PDAC, but the sample size of high-TMB PDAC treated so far is quite small. This study highlights interesting peculiarities of PDAC harboring high-TMB and may represent a reliable starting point for the assessment of TMB in the clinical management of patients affected by pancreatic cancer.
Collapse
Affiliation(s)
- Rita T. Lawlor
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134 Verona, Italy; (R.T.L.); (A.M.); (M.L.P.); (S.V.T.)
| | - Paola Mattiolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Andrea Mafficini
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134 Verona, Italy; (R.T.L.); (A.M.); (M.L.P.); (S.V.T.)
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Seung-Mo Hong
- Asan Medical Center, Department of Pathology, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Maria L. Piredda
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134 Verona, Italy; (R.T.L.); (A.M.); (M.L.P.); (S.V.T.)
| | - Sergio V. Taormina
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134 Verona, Italy; (R.T.L.); (A.M.); (M.L.P.); (S.V.T.)
| | - Giuseppe Malleo
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134 Verona, Italy; (G.M.); (G.M.); (A.P.); (R.S.)
| | - Giovanni Marchegiani
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134 Verona, Italy; (G.M.); (G.M.); (A.P.); (R.S.)
| | - Antonio Pea
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134 Verona, Italy; (G.M.); (G.M.); (A.P.); (R.S.)
| | - Roberto Salvia
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, 37134 Verona, Italy; (G.M.); (G.M.); (A.P.); (R.S.)
| | - Valentyna Kryklyva
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (V.K.); (L.A.B.)
| | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul 120-752, Korea;
| | - Lodewijk A. Brosens
- Department of Pathology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (V.K.); (L.A.B.)
- Department of Pathology, University Medical Center Utrecht, Utrecht University, 3584 CX Utrecht, The Netherlands
| | - Michele Milella
- Department of Medicine, Section of Oncology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Aldo Scarpa
- ARC-Net Research Center, University and Hospital Trust of Verona, 37134 Verona, Italy; (R.T.L.); (A.M.); (M.L.P.); (S.V.T.)
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, 37134 Verona, Italy;
| |
Collapse
|
532
|
Lengyel CG. Microsatellite Instability as a Predictor of Outcomes in Colorectal Cancer in the Era of Immune-Checkpoint Inhibitors. Curr Drug Targets 2021; 22:968-976. [PMID: 33970843 DOI: 10.2174/1389450122666210325121322] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/15/2020] [Accepted: 01/08/2021] [Indexed: 11/22/2022]
Abstract
The microsatellite instable phenotype resulting from errors in DNA mismatch repair proteins accounts for as far as 15 to 20% of non-hereditary colon cancers but is scarce in rectal cancer. It has been shown that the increased existence of tumor-specific neoantigens in hypermutated tumors is correlated with higher tumor-infiltrating lymphocytes (TILs) and overexpression of immune checkpoint receptors and ligands, mainly PD-1 and PD-L1. In particular, the data gained up to now gives evidence that neoantigen recognition constitutes a dominant component in the course of immunotherapies. This review's primary objective is to describe current approvals and summarize present knowledge about the outcomes of immuno-oncology treatment of microsatellite instable colorectal cancer (CRC). The secondary objective is to give a narrative report about testing methodologies, prognostics, and the predictive value of microsatellite instability. For this purpose, a literature review was performed, focusing on published clinical trial results, ongoing clinical trials and timelines, testing methods, and prognostic and predictive value of MSI. Following four recent FDA approvals of immunotherapy of MSI-high CRC, further work should be warranted by pathology societies towards standardization and rising concordance and reproducibility across the IHC/MSI testing landscape in order to facilitate professionals to offer better survival options for patients with CRC.
Collapse
|
533
|
Dedeurwaerdere F, Claes KB, Van Dorpe J, Rottiers I, Van der Meulen J, Breyne J, Swaerts K, Martens G. Comparison of microsatellite instability detection by immunohistochemistry and molecular techniques in colorectal and endometrial cancer. Sci Rep 2021; 11:12880. [PMID: 34145315 PMCID: PMC8213758 DOI: 10.1038/s41598-021-91974-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 05/20/2021] [Indexed: 02/08/2023] Open
Abstract
DNA mismatch repair deficiency (dMMR) testing is crucial for diagnosing Lynch syndrome and detection of microsatellite unstable (MSI) tumors eligible for immunotherapy. The aim of this study was to compare the relative diagnostic performance of three molecular MSI assays: polymerase chain reaction (PCR), MSI testing by Idylla and next-generation-sequencing (NGS) on 49 tumor samples (28 colorectal and 21 endometrial adenocarcinomas) versus immunohistochemistry (IHC). Discrepancies were investigated by MLH1 methylation analysis and integrated with germline results if available. Overall, the molecular assays achieved equivalent diagnostic performance for MSI detection with area under the ROC curves (AUC) of respectively 0.91 for Idylla and PCR, and 0.93 for NGS. In colorectal cancers with tumor cell percentages ≥ 30% all three molecular assays achieved 100% sensitivity and specificity (AUC = 1) versus IHC. Also, in endometrial cancers, all three molecular assays showed equivalent diagnostic performance, albeit at a clearly lower sensitivity ranging from 58% for Idylla to 75% for NGS, corresponding to negative predictive values from 78 to 86%. PCR, Idylla and NGS show similar diagnostic performance for dMMR detection in colorectal and endometrial cancers. Molecular MSI analysis has lower sensitivity for dMMR detection in endometrial cancer indicating that combined use of both IHC and molecular methods is recommended.Clinical Trial Number/IRB: B1172020000040, Ethical Committee, AZ Delta General Hospital.
Collapse
Affiliation(s)
| | - Kathleen Bm Claes
- Center for Medical Genetics, Ghent University Hospital, Gent, Belgium
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Gent, Belgium
| | - Jo Van Dorpe
- Department of Pathology, Ghent University Hospital, Gent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Gent, Belgium
| | | | - Joni Van der Meulen
- Center for Medical Genetics, Ghent University Hospital, Gent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent University, Gent, Belgium
- Molecular Diagnostics, Ghent University Hospital, Gent, Belgium
| | - Joke Breyne
- Department of Laboratory Medicine, Department of Laboratory Medicine, AZ Delta General Hospital, AZ Delta General Hospital, Deltalaan 1, 8800, Roeselare, Belgium
| | - Koen Swaerts
- Department of Laboratory Medicine, Department of Laboratory Medicine, AZ Delta General Hospital, AZ Delta General Hospital, Deltalaan 1, 8800, Roeselare, Belgium
| | - Geert Martens
- Department of Laboratory Medicine, Department of Laboratory Medicine, AZ Delta General Hospital, AZ Delta General Hospital, Deltalaan 1, 8800, Roeselare, Belgium.
- Department of Biomolecular Medicine, Ghent University, Gent, Belgium.
| |
Collapse
|
534
|
Cao W, Ma X, Fischer JV, Sun C, Kong B, Zhang Q. Immunotherapy in endometrial cancer: rationale, practice and perspectives. Biomark Res 2021; 9:49. [PMID: 34134781 PMCID: PMC8207707 DOI: 10.1186/s40364-021-00301-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022] Open
Abstract
Tumor immunotherapy has attracted more and more attention nowadays, and multiple clinical trials have confirmed its effect in a variety of solid tumors. Immune checkpoint inhibitors (ICIs), cancer vaccines, adoptive cell transfer (ACT), and lymphocyte-promoting cytokines are the main immunotherapy methods. Endometrial cancer (EC) is one of the most frequent tumors in women and the prognosis of recurrent or metastatic EC is poor. Since molecular classification has been applied to EC, immunotherapy for different EC subtypes (especially POLE and MSI-H) has gradually attracted attention. In this review, we focus on the expression and molecular basis of the main biomarkers in the immunotherapy of EC firstly, as well as their clinical application significance and limitations. Blocking tumor immune checkpoints is one of the most effective strategies for cancer treatment in recent years, and has now become the focus in the field of tumor research and treatment. We summarized clinical date of planned and ongoing clinical trials and introduced other common immunotherapy methods in EC, such as cancer vaccine and ACT. Hormone aberrations, metabolic syndrome (MetS) and p53 mutant and that affect the immunotherapy of endometrial cancer will also be discussed in this review.
Collapse
Affiliation(s)
- Wenyu Cao
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Xinyue Ma
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Jean Victoria Fischer
- Department of Pathology, Northwestern Medicine, Gynecologic Pathology Fellow, Chicago, Illinois, USA
| | - Chenggong Sun
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Beihua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China.,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, 107 West Wenhua Road, Ji'nan, Shandong, 250012, P.R. China. .,Gynecology Oncology Key Laboratory, Qilu Hospital, Shandong University, Ji'nan, Shandong, 250012, P.R. China.
| |
Collapse
|
535
|
Effects of a Novel Thiadiazole Derivative with High Anticancer Activity on Cancer Cell Immunogenic Markers: Mismatch Repair System, PD-L1 Expression, and Tumor Mutation Burden. Pharmaceutics 2021; 13:pharmaceutics13060885. [PMID: 34203761 PMCID: PMC8232699 DOI: 10.3390/pharmaceutics13060885] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 06/08/2021] [Accepted: 06/09/2021] [Indexed: 12/31/2022] Open
Abstract
Microsatellite instability (MSI), tumor mutation burden (TMB), and programmed cell death ligand-1 (PD-L1) are particularly known as immunotherapy predictive biomarkers. MSI and TMB are closely related to DNA mismatch repair (MMR) pathway functionality, while the PD-L1 checkpoint mediates cancer cell evasion from immune surveillance via the PD-L1/PD-1 axis. Among all the novel triazolo[3,4-b]thiadiazole derivatives, the compound KA39 emerged as the most potent anticancer agent. In the present study, potential alterations in MSI, TMB, and/or PD-L1 expression upon cell treatment with KA39 are explored. We tested three MMR-deficient (DLD-1, LS174T, and DU-145) and two MMR-proficient (HT-29 and PC-3) human cancer cell lines. Our findings support KA39-induced PD-L1 overexpression in all cancer cell lines, although the most outstanding increase was observed in MMR-proficient HT-29 cells. MSI analysis showed that KA39 affects the MMR system, impairing its recognition or repair activity, particularly in MMR-deficient DLD-1 and DU-145 cells, enhancing oligonucleotide production. There were no remarkable alterations in the TMB between untreated and treated cells, indicating that KA39 does not belong to mutagenic agents. Taking together the significant in vitro anticancer activity with PD-L1 upregulation and MSI increase, KA39 should be investigated further for its implication in chemo-immunotherapy of cancer.
Collapse
|
536
|
Beta2-microglobulin(B2M) in cancer immunotherapies: Biological function, resistance and remedy. Cancer Lett 2021; 517:96-104. [PMID: 34129878 DOI: 10.1016/j.canlet.2021.06.008] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 05/31/2021] [Accepted: 06/09/2021] [Indexed: 12/30/2022]
Abstract
Cancer immunotherapies have made much headway during the past decades. Techniques including the immune checkpoint inhibition (ICI) and adoptive cell therapy (ACT) have harvested impressive efficacy and provided far-reaching tools for treating cancer patients. However, due to inadequate priming of the immune system, a certain subgroup of patients remains resistant to cancer immunotherapies during or after the treatment. β2-microglobulin (B2M) is an important subunit of major histocompatibility complex (MHC) class I which exerts substantive biological functions in tumorigenesis and immune control. Accumulating evidence has shown that alterations of B2M gene and B2M proteins contribute to poor reaction to cancer immunotherapies by dampening antigen presentation. Here, we discuss the basic biological functions of B2M, its distribution in a spectrum of cancers, and current understanding of its role in ICI, cancer vaccines and chimeric antigen receptor T cell (CAR-T) therapies. Furthermore, we summarize some promising therapeutic strategies to improve the efficacy inhibited by B2M defects.
Collapse
|
537
|
Wang Y, Tong Z, Zhang W, Zhang W, Buzdin A, Mu X, Yan Q, Zhao X, Chang HH, Duhon M, Zhou X, Zhao G, Chen H, Li X. FDA-Approved and Emerging Next Generation Predictive Biomarkers for Immune Checkpoint Inhibitors in Cancer Patients. Front Oncol 2021; 11:683419. [PMID: 34164344 PMCID: PMC8216110 DOI: 10.3389/fonc.2021.683419] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022] Open
Abstract
A patient's response to immune checkpoint inhibitors (ICIs) is a complex quantitative trait, and determined by multiple intrinsic and extrinsic factors. Three currently FDA-approved predictive biomarkers (progra1mmed cell death ligand-1 (PD-L1); microsatellite instability (MSI); tumor mutational burden (TMB)) are routinely used for patient selection for ICI response in clinical practice. Although clinical utility of these biomarkers has been demonstrated in ample clinical trials, many variables involved in using these biomarkers have poised serious challenges in daily practice. Furthermore, the predicted responders by these three biomarkers only have a small percentage of overlap, suggesting that each biomarker captures different contributing factors to ICI response. Optimized use of currently FDA-approved biomarkers and development of a new generation of predictive biomarkers are urgently needed. In this review, we will first discuss three widely used FDA-approved predictive biomarkers and their optimal use. Secondly, we will review four novel gene signature biomarkers: T-cell inflamed gene expression profile (GEP), T-cell dysfunction and exclusion gene signature (TIDE), melanocytic plasticity signature (MPS) and B-cell focused gene signature. The GEP and TIDE have shown better predictive performance than PD-L1, and PD-L1 or TMB, respectively. The MPS is superior to PD-L1, TMB, and TIDE. The B-cell focused gene signature represents a previously unexplored predictive biomarker to ICI response. Thirdly, we will highlight two combined predictive biomarkers: TMB+GEP and MPS+TIDE. These integrated biomarkers showed improved predictive outcomes compared to a single predictor. Finally, we will present a potential nucleic acid biomarker signature, allowing DNA and RNA biomarkers to be analyzed in one assay. This comprehensive signature could represent a future direction of developing robust predictive biomarkers, particularly for the cold tumors, for ICI response.
Collapse
Affiliation(s)
- Ye Wang
- Clinical Laboratory, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
| | - Zhuang Tong
- Liaoning Cancer Hospital and Institute, Cancer Hospital of China Medical University, Shenyang, China
| | - Wenhua Zhang
- Clinical Laboratory, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
| | - Weizhen Zhang
- Department of Biology, University of California – Santa Cruz, Santa Cruz, CA, United States
| | - Anton Buzdin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Department of Biological and Medical Physics, Moscow Institute of Physics and Technology, Moscow, Russia
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Xiaofeng Mu
- Clinical Laboratory, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Qing Yan
- Clinical Laboratory, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
| | - Xiaowen Zhao
- Clinical Laboratory, Qingdao Central Hospital, The Second Affiliated Hospital of Medical College of Qingdao University, Qingdao, China
| | - Hui-Hua Chang
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA) Technology Center for Genomics & Bioinformatics, Los Angeles, CA, United States
| | - Mark Duhon
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA) Technology Center for Genomics & Bioinformatics, Los Angeles, CA, United States
| | - Xin Zhou
- Department of Medicine, Qiqihaer First Hospital, Qiqihar, China
| | - Gexin Zhao
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA) Technology Center for Genomics & Bioinformatics, Los Angeles, CA, United States
| | - Hong Chen
- Department of Medicine, Qiqihaer First Hospital, Qiqihar, China
| | - Xinmin Li
- Department of Pathology & Laboratory Medicine, University of California, Los Angeles (UCLA) Technology Center for Genomics & Bioinformatics, Los Angeles, CA, United States
| |
Collapse
|
538
|
Zhang H, Dai Z, Wu W, Wang Z, Zhang N, Zhang L, Zeng WJ, Liu Z, Cheng Q. Regulatory mechanisms of immune checkpoints PD-L1 and CTLA-4 in cancer. J Exp Clin Cancer Res 2021; 40:184. [PMID: 34088360 PMCID: PMC8178863 DOI: 10.1186/s13046-021-01987-7] [Citation(s) in RCA: 309] [Impact Index Per Article: 77.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/17/2021] [Indexed: 02/01/2023] Open
Abstract
The cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4)/B7 and programmed death 1 (PD-1)/ programmed cell death-ligand 1 (PD-L1) are two most representative immune checkpoint pathways, which negatively regulate T cell immune function during different phases of T-cell activation. Inhibitors targeting CTLA-4/B7 and PD1/PD-L1 pathways have revolutionized immunotherapies for numerous cancer types. Although the combined anti-CTLA-4/B7 and anti-PD1/PD-L1 therapy has demonstrated promising clinical efficacy, only a small percentage of patients receiving anti-CTLA-4/B7 or anti-PD1/PD-L1 therapy experienced prolonged survival. Regulation of the expression of PD-L1 and CTLA-4 significantly impacts the treatment effect. Understanding the in-depth mechanisms and interplays of PD-L1 and CTLA-4 could help identify patients with better immunotherapy responses and promote their clinical care. In this review, regulation of PD-L1 and CTLA-4 is discussed at the levels of DNA, RNA, and proteins, as well as indirect regulation of biomarkers, localization within the cell, and drugs. Specifically, some potential drugs have been developed to regulate PD-L1 and CTLA-4 expressions with high efficiency.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Ziyu Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wantao Wu
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
| | - Zeyu Wang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Nan Zhang
- One-third Lab, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-Jing Zeng
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
| | - Zhixiong Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China.
| |
Collapse
|
539
|
Early memory differentiation and cell death resistance in T cells predicts melanoma response to sequential anti-CTLA4 and anti-PD1 immunotherapy. Genes Immun 2021; 22:108-119. [PMID: 34079092 DOI: 10.1038/s41435-021-00138-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 05/11/2021] [Accepted: 05/21/2021] [Indexed: 11/09/2022]
Abstract
Immune checkpoint blockers (ICBs)-based immunotherapy has revolutionised oncology. However, the benefits of ICBs are limited to only a subset of patients. Herein, the biomarkers-driven application of ICBs promises to increase their efficacy. Such biomarkers include lymphocytic IFNγ-signalling and/or cytolytic activity (granzymes and perforin-1) footprints, whose levels in pre-treatment tumours can predict favourable patient survival following ICB-treatment. However, it is not clear whether such biomarkers have the same value in predicting survival of patients receiving first-line anti-CTLA4 ICB-therapy, and subsequently anti-PD1 ICB-therapy (i.e., sequential ICB-immunotherapy regimen). To address this, we applied highly integrated systems/computational immunology approaches to existing melanoma bulk-tumour transcriptomic and single-cell (sc)RNAseq data originating from immuno-oncology clinical studies applying ICB-treatment. Interestingly, we observed that CD8+/CD4+T cell-associated IFNγ-signalling or cytolytic activity signatures fail to predict tumour response in patients treated with anti-CTLA4 ICB-therapy as a first-line and anti-PD1 ICB-therapy in the second-line setting. On the contrary, signatures associated with early memory CD8+/CD4+T cells (integrating TCF1-driven stem-like transcriptional programme), capable of resisting cell death/apoptosis, better predicted objective response rates to ICB-immunotherapy, and favourable survival in the setting of sequential ICB-immunotherapy. These observations suggest that sequencing of ICB-therapy might have a specific impact on the T cell-repertoire and may influence the predictive value of tumoural immune biomarkers.
Collapse
|
540
|
Corti F, Lonardi S, Intini R, Salati M, Fenocchio E, Belli C, Borelli B, Brambilla M, Prete AA, Quarà V, Antista M, Fassan M, Morano F, Spallanzani A, Ambrosini M, Curigliano G, de Braud F, Zagonel V, Fucà G, Pietrantonio F. The Pan-Immune-Inflammation Value in microsatellite instability-high metastatic colorectal cancer patients treated with immune checkpoint inhibitors. Eur J Cancer 2021; 150:155-167. [PMID: 33901794 DOI: 10.1016/j.ejca.2021.03.043] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 01/26/2023]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) yielded unprecedented efficacy in patients with microsatellite instability (MSI)-high metastatic colorectal cancer (mCRC). Since the Pan-Immune-Inflammation Value (PIV) is a blood-based biomarker with prognostic usefulness in mCRC, it might predict clinical outcomes and primary resistance to ICIs. METHODS We retrospectively analysed the association of PIV and its early modulation at 3/4 weeks after treatment initiation with the outcomes of MSI-high mCRC patients receiving anti-programmed death-(ligand)1 (PD-[L]1) +/- anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) agents. PIV was calculated as follows: (neutrophil count × platelet count × monocyte count)/lymphocyte count. PIV cut-offs were determined using the maximally selected rank statistics. RESULTS A total of 163 patients were included. In the multivariable models for overall survival (OS) and progression-free survival (PFS), both high (>492) baseline PIV (OS: adjusted [a] HR, 3.00; 95% CI, 1.49-6.04, p = 0.002; PFS: aHR, 1.91; 95% CI, 1.06-3.44, p = 0.031) and early PIV increase ≥+30% (OS: aHR, 3.21; 95% CI, 1.65-6.23, p < 0.001; PFS: aHR, 2.25; 95% CI, 1.30-3.89, p = 0.003) confirmed an independent prognostic impact. After stratifying patients according to baseline PIV and ICI regimen, OS and PFS were significantly worse in subjects with high PIV receiving anti-PD-1/PD-L1 monotherapy. Early PIV increase was an independent predictor of clinical benefit (aOR, 0.23; 95% CI, 0.08-0.66; p = 0.007), whereas a trend was observed for baseline PIV (aOR, 0.33; 95% CI, 0.10-1.07; p = 0.065). CONCLUSION PIV is a strong predictor of outcomes in MSI-high mCRC patients receiving ICIs. Prospective validation of these results is required to establish its role as a stratification factor for personalised combination strategies.
Collapse
Affiliation(s)
- Francesca Corti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Sara Lonardi
- Department of Oncology, Istituto Oncologico Veneto, IRCCS, Via Gattamelata, 64, 35128, Padua, Italy.
| | - Rossana Intini
- Department of Oncology, Istituto Oncologico Veneto, IRCCS, Via Gattamelata, 64, 35128, Padua, Italy.
| | - Massimiliano Salati
- Department of Oncology and Hematology, University Hospital of Modena, Via del Pozzo, 71, 41125, Modena, Italy; PhD Program in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, Via Università, 4, 41121, Modena Italy.
| | - Elisabetta Fenocchio
- Multidisciplinary Outpatient Oncology Clinic, Candiolo Cancer Institute, FPO-IRCCS, Strada Provinciale 142, km 3.95, 10060 Candiolo (TO), Italy.
| | - Carmen Belli
- Division of Early Drugs Development for Innovative Therapies, Istituto Europeo di Oncologia, IRCCS, Via Ripamonti 435, 20141, Milan, Italy.
| | - Beatrice Borelli
- Unit of Medical Oncology, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy.
| | - Marta Brambilla
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Alessandra A Prete
- Department of Oncology, Istituto Oncologico Veneto, IRCCS, Via Gattamelata, 64, 35128, Padua, Italy.
| | - Virginia Quarà
- Department of Oncology, University of Turin, Via Giuseppe Verdi, 8, 10124, Torino, Italy.
| | - Maria Antista
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, Via Giustiniani 2, 35128, Padua, Italy.
| | - Federica Morano
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Andrea Spallanzani
- Department of Oncology and Hematology, University Hospital of Modena, Via del Pozzo, 71, 41125, Modena, Italy.
| | - Margherita Ambrosini
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Giuseppe Curigliano
- Division of Early Drugs Development for Innovative Therapies, Istituto Europeo di Oncologia, IRCCS, Via Ripamonti 435, 20141, Milan, Italy; Oncology and Hemato-oncology Department, University of Milan, via Festa del Perdono 7, 20122, Milan, Italy.
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy; Oncology and Hemato-oncology Department, University of Milan, via Festa del Perdono 7, 20122, Milan, Italy.
| | - Vittorina Zagonel
- Department of Oncology, Istituto Oncologico Veneto, IRCCS, Via Gattamelata, 64, 35128, Padua, Italy.
| | - Giovanni Fucà
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy.
| | - Filippo Pietrantonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Giacomo Venezian 1, 20133, Milan, Italy; Oncology and Hemato-oncology Department, University of Milan, via Festa del Perdono 7, 20122, Milan, Italy.
| |
Collapse
|
541
|
Zhang W, Yin H, Huang Z, Zhao J, Zheng H, He D, Li M, Tan W, Tian S, Song B. Development and validation of MRI-based deep learning models for prediction of microsatellite instability in rectal cancer. Cancer Med 2021; 10:4164-4173. [PMID: 33963688 PMCID: PMC8209621 DOI: 10.1002/cam4.3957] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Microsatellite instability (MSI) predetermines responses to adjuvant 5-fluorouracil and immunotherapy in rectal cancer and serves as a prognostic biomarker for clinical outcomes. Our objective was to develop and validate a deep learning model that could preoperatively predict the MSI status of rectal cancer based on magnetic resonance images. METHODS This single-center retrospective study included 491 rectal cancer patients with pathologically proven microsatellite status. Patients were randomly divided into the training/validation cohort (n = 395) and the testing cohort (n = 96). A clinical model using logistic regression was constructed to discriminate MSI status using only clinical factors. Based on a modified MobileNetV2 architecture, deep learning models were tested for the predictive ability of MSI status from magnetic resonance images, with or without integrating clinical factors. RESULTS The clinical model correctly classified 37.5% of MSI status in the testing cohort, with an AUC value of 0.573 (95% confidence interval [CI], 0.468 ~ 0.674). The pure imaging-based model and the combined model correctly classified 75.0% and 85.4% of MSI status in the testing cohort, with AUC values of 0.820 (95% CI, 0.718 ~ 0.884) and 0.868 (95% CI, 0.784 ~ 0.929), respectively. Both deep learning models performed better than the clinical model (p < 0.05). There was no statistically significant difference between the deep learning models with or without integrating clinical factors. CONCLUSIONS Deep learning based on high-resolution T2-weighted magnetic resonance images showed a good predictive performance for MSI status in rectal cancer patients. The proposed model may help to identify patients who would benefit from chemotherapy or immunotherapy and determine individualized therapeutic strategies for these patients.
Collapse
Affiliation(s)
- Wei Zhang
- Department of RadiologyWest China HospitalSichuan UniversityChengduChina
- Department of RadiologySichuan Provincial Corps HospitalChinese People's Armed Police ForcesLeshanChina
| | - Hongkun Yin
- Institute of Advanced ResearchInferVisionBeijingChina
| | - Zixing Huang
- Department of RadiologyWest China HospitalSichuan UniversityChengduChina
| | - Jian Zhao
- Department of RadiologyWest China HospitalSichuan UniversityChengduChina
- Department of RadiologySichuan Provincial Corps HospitalChinese People's Armed Police ForcesLeshanChina
| | - Haoyu Zheng
- Department of RadiologySichuan Provincial Corps HospitalChinese People's Armed Police ForcesLeshanChina
| | - Du He
- Department of PathologyWest China HospitalSichuan UniversityChengduChina
| | - Mou Li
- Department of RadiologyWest China HospitalSichuan UniversityChengduChina
| | - Weixiong Tan
- Institute of Advanced ResearchInferVisionBeijingChina
| | - Song Tian
- Institute of Advanced ResearchInferVisionBeijingChina
| | - Bin Song
- Department of RadiologyWest China HospitalSichuan UniversityChengduChina
| |
Collapse
|
542
|
Shimada Y, Okuda S, Watanabe Y, Tajima Y, Nagahashi M, Ichikawa H, Nakano M, Sakata J, Takii Y, Kawasaki T, Homma KI, Kamori T, Oki E, Ling Y, Takeuchi S, Wakai T. Histopathological characteristics and artificial intelligence for predicting tumor mutational burden-high colorectal cancer. J Gastroenterol 2021; 56:547-559. [PMID: 33909150 DOI: 10.1007/s00535-021-01789-w] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 04/15/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Tumor mutational burden-high (TMB-H), which is detected with gene panel testing, is a promising biomarker for immune checkpoint inhibitors (ICIs) in colorectal cancer (CRC). However, in clinical practice, not every patient is tested for TMB-H using gene panel testing. We aimed to identify the histopathological characteristics of TMB-H CRC for efficient selection of patients who should undergo gene panel testing. Moreover, we attempted to develop a convolutional neural network (CNN)-based algorithm to predict TMB-H CRC directly from hematoxylin and eosin (H&E) slides. METHODS We used two CRC cohorts tested for TMB-H, and whole-slide H&E digital images were obtained from the cohorts. The Japanese CRC (JP-CRC) cohort (N = 201) was evaluated to detect the histopathological characteristics of TMB-H using H&E slides. The JP-CRC cohort and The Cancer Genome Atlas (TCGA) CRC cohort (N = 77) were used to develop a CNN-based TMB-H prediction model from the H&E digital images. RESULTS Tumor-infiltrating lymphocytes (TILs) were significantly associated with TMB-H CRC (P < 0.001). The area under the curve (AUC) for predicting TMB-H CRC was 0.910. We developed a CNN-based TMB-H prediction model. Validation tests were conducted 10 times using randomly selected slides, and the average AUC for predicting TMB-H slides was 0.934. CONCLUSIONS TILs, a histopathological characteristic detected with H&E slides, are associated with TMB-H CRC. Our CNN-based model has the potential to predict TMB-H CRC directly from H&E slides, thereby reducing the burden on pathologists. These approaches will provide clinicians with important information about the applications of ICIs at low cost.
Collapse
Affiliation(s)
- Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan.,Medical Genome Center, Niigata University Medical and Dental Hospital, Niigata, Japan
| | - Shujiro Okuda
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan. .,Medical Genome Center, Niigata University Medical and Dental Hospital, Niigata, Japan.
| | - Yu Watanabe
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan.,Division of Cancer Genome Informatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Yosuke Tajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Masato Nakano
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Jun Sakata
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Yasumasa Takii
- Department of Surgery, Niigata Cancer Center Hospital, Niigata, Japan
| | - Takashi Kawasaki
- Department of Pathology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Kei-Ichi Homma
- Department of Pathology, Niigata Cancer Center Hospital, Niigata, Japan
| | - Tomohiro Kamori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yiwei Ling
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan
| | - Shiho Takeuchi
- Division of Bioinformatics, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan.,Division of Cancer Genome Informatics, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata, Niigata, 951-8510, Japan. .,Medical Genome Center, Niigata University Medical and Dental Hospital, Niigata, Japan.
| |
Collapse
|
543
|
Xu J, Yang X, Mao Y, Mei J, Wang H, Ding J, Hua D. Removal of N-Linked Glycosylation Enhances PD-L1 Detection in Colon Cancer: Validation Research Based on Immunohistochemistry Analysis. Technol Cancer Res Treat 2021; 20:15330338211019442. [PMID: 34060360 PMCID: PMC8173990 DOI: 10.1177/15330338211019442] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In recent years, immunotherapies have emerged as effective therapeutic strategies for treating human cancers. However, accumulating evidence has revealed an inconsistency between the response to immune checkpoint inhibitors and programmed death ligand 1 (PD-L1) expression status detected by immunohistochemistry staining. Recent research has revealed that the removal of N-Linked glycosylation significantly enhanced PD-L1 detection, resulting in both more accurate PD-L1 quantification and clinical outcome prediction. In the present study, we evaluated natural and deglycosylated PD-L1 expression in colon cancer using the PD-L1 28–8 antibody. The results of the present study validated the hypothesis that PD-L1 had a higher expression in colon cancer tissues compared with normal tissues. Additionally, colon tumors with defective mismatch repair tended to express higher PD-L1 than those without. Most importantly, the results of the present study indicated that the removal of N-linked glycosylation remarkably enhanced PD-L1 detection. Moreover, the PD-L1 signal intensity of samples with a low natural PD-L1 signal was enhanced more remarkably than that of samples with high signal intensity. Overall, our research provides an improved strategy for patient stratification for anti-PD-1/PD-L1 therapy, which deepens the clinical significance of this established strategy for treatment of colon cancer.
Collapse
Affiliation(s)
- Junying Xu
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Xuejing Yang
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Yong Mao
- Department of Oncology, Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu Province, China
| | - Jie Mei
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Huiyu Wang
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Junli Ding
- Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, China
| | - Dong Hua
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China.,Department of Oncology, Wuxi People's Hospital Affiliated to Nanjing Medical University, Wuxi, Jiangsu Province, China
| |
Collapse
|
544
|
Guyot D'Asnières De Salins A, Tachon G, Cohen R, Karayan-Tapon L, Junca A, Frouin E, Godet J, Evrard C, Randrian V, Duval A, Svrcek M, Lascols O, Vignot S, Coulet F, André T, Fléjou JF, Cervera P, Tougeron D. Discordance between immunochemistry of mismatch repair proteins and molecular testing of microsatellite instability in colorectal cancer. ESMO Open 2021; 6:100120. [PMID: 33930657 PMCID: PMC8102173 DOI: 10.1016/j.esmoop.2021.100120] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND DNA mismatch repair system deficiency (dMMR) is found in 15% of colorectal cancers (CRCs). Two methods are used to determine dMMR, immunohistochemistry (IHC) of MMR proteins and molecular testing of microsatellite instability (MSI). Only studies with a low number of patients have reported rates of discordance between these two methods, ranging from 1% to 10%. MATERIALS AND METHODS Overall, 3228 consecutive patients with CRCs from two centers were included. Molecular testing was carried out using the Pentaplex panel and IHC evaluated four (MLH1, MSH2, MSH6, and PMS2; cohort 1; n = 1085) or two MMR proteins (MLH1 and MSH2; cohort 2; n = 2143). The primary endpoint was the rate of discordance between MSI and MMR IHC tests. RESULTS Fifty-one discordant cases (1.6%) were initially observed. Twenty-nine out of 51 discordant cases were related to IHC misclassifications. In cohort 1, after re-reading IHC and/or carrying out new IHC, 16 discordant cases were reclassified as nondiscordant. In cohort 2, after the addition of MSH6/PMS2 IHC and re-examination, 13 were reclassified as nondiscordant. In addition, 10 misclassifications of molecular tests were identified. Finally, only 12 discordant cases (0.4%) remained: 5 were proficient MMR/MSI and 7 were dMMR/microsatellite stable. CONCLUSIONS Our study confirmed the high degree of concordance between MSI and MMR IHC tests. Discordant cases must be reviewed, and if needed, tests must be repeated and analyzed by an expert team.
Collapse
Affiliation(s)
| | - G Tachon
- Faculty of Medicine, University of Poitiers, Poitiers, France; INSERM 1084, Experimental and Clinical Neurosciences Laboratory, University of Poitiers, Poitiers, France; Cancer Biology Department, Poitiers University Hospital, Poitiers, France
| | - R Cohen
- Sorbonne University, Department of Medical Oncology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - L Karayan-Tapon
- Faculty of Medicine, University of Poitiers, Poitiers, France; INSERM 1084, Experimental and Clinical Neurosciences Laboratory, University of Poitiers, Poitiers, France; Cancer Biology Department, Poitiers University Hospital, Poitiers, France
| | - A Junca
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - E Frouin
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - J Godet
- Pathology Department, Poitiers University Hospital, Poitiers, France
| | - C Evrard
- Medical Oncology Department, Poitiers University Hospital, Poitiers, France
| | - V Randrian
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France; Faculty of Medicine, University of Poitiers, Poitiers, France
| | - A Duval
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - M Svrcek
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne University, Department of Pathology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - O Lascols
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - S Vignot
- Sorbonne University, Department of Medical Oncology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - F Coulet
- Department of Genetics, Pitié Salpétrière Hospital, AP-HP and Sorbonne University, Paris, France
| | - T André
- Sorbonne University, Department of Medical Oncology, Saint-Antoine Hospital, AP-HP, Paris, France; Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France
| | - J-F Fléjou
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne University, Department of Pathology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - P Cervera
- Sorbonne University, INSERM, Unité Mixte de Recherche Scientifique 938 and SIRIC CURAMUS, Centre de Recherche Saint-Antoine, Equipe Instabilité des Microsatellites et Cancer, Equipe labellisée par la Ligue Nationale contre le Cancer, Paris, France; Sorbonne University, Department of Pathology, Saint-Antoine Hospital, AP-HP, Paris, France
| | - D Tougeron
- Gastroenterology Department, Poitiers University Hospital, Poitiers, France; Faculty of Medicine, University of Poitiers, Poitiers, France; Medical Oncology Department, Poitiers University Hospital, Poitiers, France.
| |
Collapse
|
545
|
Rüschoff J, Baretton G, Bläker H, Dietmaier W, Dietel M, Hartmann A, Horn LC, Jöhrens K, Kirchner T, Knüchel R, Mayr D, Merkelbach-Bruse S, Schildhaus HU, Schirmacher P, Tiemann M, Tiemann K, Weichert W, Büttner R. [MSI testing : What is new? What should be considered? German version]. DER PATHOLOGE 2021; 42:414-423. [PMID: 34043067 DOI: 10.1007/s00292-021-00944-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/25/2021] [Indexed: 12/21/2022]
Abstract
Based on new trial data regarding immune checkpoint inhibitors (ICIs), the detection of high-grade microsatellite instability (MSI-H) or underlying deficient mismatch repair protein (dMMR) is now becoming increasingly important for predicting treatment response. For the first time, a PD‑1 ICI (pembrolizumab) has been approved by the European Medicines Agency (EMA) for first-line treatment of advanced (stage IV) dMMR/MSI‑H colorectal cancer (CRC). Further indications, such as dMMR/MSI‑H endometrial carcinoma (EC), have already succeeded (Dostarlimab, 2nd line treatment) and others are expected to follow before the end of 2021. The question of optimal testing in routine diagnostics should therefore be re-evaluated. Based on a consideration of the strengths and weaknesses of the widely available methods (immunohistochemistry and PCR), a test algorithm is proposed that allows quality assured, reliable, and cost-effective dMMR/MSI‑H testing. For CRC and EC, testing is therefore already possible at the primary diagnosis stage, in line with international recommendations (NICE, NCCN). The clinician is therefore enabled from the outset to consider not only the predictive but also the prognostic and predispositional implications of such a test when counseling patients and formulating treatment recommendations. As a basis for quality assurance, participation in interlaboratory comparisons and continuous documentation of results (e.g., QuIP Monitor) are strongly recommended.
Collapse
Affiliation(s)
- Josef Rüschoff
- Institut für Pathologie Nordhessen, TARGOS Molecular Pathology GmbH, Germaniastr. 7, 34119, Kassel, Deutschland.
| | - Gustavo Baretton
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - Hendrik Bläker
- Institut für Pathologie, Universitätsklinikum Leipzig, Liebigstr. 26, Gebäude G, 04103, Leipzig, Deutschland
| | - Wolfgang Dietmaier
- Institut für Pathologie/Zentrum für molekularpathologische Diagnostik, Universität Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Deutschland
| | - Manfred Dietel
- Institut für Pathologie, Campus Mitte, Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Deutschland
| | - Arndt Hartmann
- Pathologisches Institut, Universität Erlangen-Nürnberg, Krankenhausstr. 8-10, 91054, Erlangen, Deutschland
| | - Lars-Christian Horn
- Institut für Pathologie, Universitätsklinikum Leipzig, Liebigstr. 26, Gebäude G, 04103, Leipzig, Deutschland
| | - Korinna Jöhrens
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus, Fetscherstr. 74, 01307, Dresden, Deutschland
| | - Thomas Kirchner
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland
| | - Ruth Knüchel
- Institut für Pathologie, Universitätsklinikum RWTH Aachen, Pauwelsstr. 30, 52074, Aachen, Deutschland
| | - Doris Mayr
- Pathologisches Institut, Ludwig-Maximilians-Universität München, Thalkirchner Str. 36, 80337, München, Deutschland
| | - Sabine Merkelbach-Bruse
- Institut für Pathologie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| | - Hans-Ulrich Schildhaus
- Institut für Pathologie, Universitätsklinikum Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| | - Peter Schirmacher
- Pathologisches Institut, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 224, 69120, Heidelberg, Deutschland
| | - Markus Tiemann
- Institut für Hämatopathologie Hamburg, Fangdieckstr. 75a, 22547, Hamburg, Deutschland
| | - Katharina Tiemann
- Institut für Hämatopathologie Hamburg, Fangdieckstr. 75a, 22547, Hamburg, Deutschland
| | - Wilko Weichert
- Institut für Pathologie, Technische Universität München, Trogerstr. 18, 81675, München, Deutschland
| | - Reinhard Büttner
- Institut für Pathologie, Universitätsklinikum Köln, Kerpener Str. 62, 50937, Köln, Deutschland
| |
Collapse
|
546
|
Mechanisms of Immune Escape and Resistance to Checkpoint Inhibitor Therapies in Mismatch Repair Deficient Metastatic Colorectal Cancers. Cancers (Basel) 2021; 13:cancers13112638. [PMID: 34072037 PMCID: PMC8199207 DOI: 10.3390/cancers13112638] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/20/2021] [Accepted: 05/21/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary A subset of colorectal cancers (CRCs) is characterized by a mismatch repair deficiency that is frequently associated with microsatellite instability (MSI). The compromised DNA repair machinery leads to the accumulation of tumor neoantigens affecting the sensitivity of MSI metastatic CRC to immune checkpoint inhibitors (CPIs), both upfront and in later lines of treatment. However, up to 30% of MSI CRCs exhibit primary resistance to frontline immune based therapy, and an additional subset develops acquired resistance. Here, we first discuss the clinical and molecular features of MSI CRCs and then we review how the loss of antigenicity, immunogenicity, and a hostile tumor microenvironment could influence primary and acquired resistance to CPIs. Finally, we describe strategies to improve the outcome of MSI CRC patients upon CPI treatment. Abstract Immune checkpoint inhibitors (CPIs) represent an effective therapeutic strategy for several different types of solid tumors and are remarkably effective in mismatch repair deficient (MMRd) tumors, including colorectal cancer (CRC). The prevalent view is that the elevated and dynamic neoantigen burden associated with the mutator phenotype of MMRd fosters enhanced immune surveillance of these cancers. In addition, recent findings suggest that MMRd tumors have increased cytosolic DNA, which triggers the cGAS STING pathway, leading to interferon-mediated immune response. Unfortunately, approximately 30% of MMRd CRC exhibit primary resistance to CPIs, while a substantial fraction of tumors acquires resistance after an initial benefit. Profiling of clinical samples and preclinical studies suggests that alterations in the Wnt and the JAK-STAT signaling pathways are associated with refractoriness to CPIs. Intriguingly, mutations in the antigen presentation machinery, such as loss of MHC or Beta-2 microglobulin (B2M), are implicated in initial immune evasion but do not impair response to CPIs. In this review, we outline how understanding the mechanistic basis of immune evasion and CPI resistance in MMRd CRC provides the rationale for innovative strategies to increase the subset of patients benefiting from CPIs.
Collapse
|
547
|
T Danley K, Schmitz K, Ghai R, Sclamberg JS, Buckingham LE, Burgess K, Kuzel TM, Usha L. A Durable Response to Pembrolizumab in a Patient with Uterine Serous Carcinoma and Lynch Syndrome due to the MSH6 Germline Mutation. Oncologist 2021; 26:811-817. [PMID: 34018286 DOI: 10.1002/onco.13832] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 05/07/2021] [Indexed: 11/06/2022] Open
Abstract
Pembrolizumab, a programmed death 1 ligand (PD-1) checkpoint inhibitor, has elicited responses in mismatch repair (MMR)-deficient advanced solid tumors, leading to its agnostic approval by the US Food and Drug Administration in 2017 when no other therapeutic options are available. However, there are still insufficient data on the response to checkpoint inhibitors in advanced endometrial cancer related to Lynch syndrome (LS) and, specifically, in uterine serous carcinoma, which is uncommon in LS. Here we report a case of metastatic uterine serous carcinoma due to a germline MSH6 mutation (Lynch syndrome) that was discovered because of a patient's tumor MMR deficiency. The patient was started on first-line pembrolizumab in 2018 and sustained a partial response. She remains asymptomatic and progression free for more than 2 years. Tumor sequencing showed a high mutational burden and an upstream somatic mutation in the same gene, p.F1088fs. Immunohistochemical staining was negative for PD-L1 expression. We discuss clinical characteristics of the patient, molecular features of her tumor, and the mechanism of her tumor response. We also discuss the duration of immunotherapy in her case. Our case demonstrated a partial response and a long-term remission from the frontline single-agent pembrolizumab in a woman with metastatic uterine serous carcinoma and Lynch syndrome due to a germline MSH6 gene mutation. Our experience suggests a potential significant clinical benefit of checkpoint inhibitors used as single agents early on in the treatment of MMR-deficient/high microsatellite instability/hypermutated uterine cancers in women with Lynch syndrome. KEY POINTS: Even though checkpoint inhibitors are effective in mismatch repair-deficient endometrial cancer, it is unknown whether the response to them differs between women with endometrial cancer due to germline mutations in a mismatch repair gene (Lynch syndrome) and women with sporadic endometrial cancer. In our case, a patient with Lynch syndrome and recurrent mismatch repair-deficient serous endometrial cancer achieved a durable remission on the first-line therapy with the checkpoint inhibitor pembrolizumab and remains progression free after more than 2 years. Based on our observation and the data, suggesting the stronger immune activation in women with Lynch syndrome-associated endometrial cancer, we propose to use checkpoint inhibitor monotherapy early in the course of their treatment and stratify patients for the presence of Lynch syndrome in clinical trials.
Collapse
Affiliation(s)
- Kelsey T Danley
- Department of Medicine, Division of Hematology, Oncology, and Stem Cell Transplant Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Karen Schmitz
- Department of Medicine, Division of Hematology, Oncology, and Stem Cell Transplant Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Ritu Ghai
- Department of Pathology, Advocate Christ Medical Center, Chicago, Illinois, USA.,Department of Pathology, Rush University Medical Center, Chicago, Illinois, USA
| | - Joy S Sclamberg
- Department of Diagnostic Radiology & Nuclear Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Lela E Buckingham
- Department of Pathology, Advocate Christ Medical Center, Chicago, Illinois, USA
| | - Kelly Burgess
- Department of Medicine, Division of Hematology, Oncology, and Stem Cell Transplant Medicine, Rush University Medical Center, Chicago, Illinois, USA.,Ambry Genetics, Enterprise, Aliso Viejo, California, USA
| | - Timothy M Kuzel
- Department of Medicine, Division of Hematology, Oncology, and Stem Cell Transplant Medicine, Rush University Medical Center, Chicago, Illinois, USA
| | - Lydia Usha
- Department of Medicine, Division of Hematology, Oncology, and Stem Cell Transplant Medicine, Rush University Medical Center, Chicago, Illinois, USA
| |
Collapse
|
548
|
Sun J, Xu J, Sun C, Zheng M, Li Y, Zhu S, Zhang S. Screening and Prognostic Value of Methylated Septin9 and its Association With Clinicopathological and Molecular Characteristics in Colorectal Cancer. Front Mol Biosci 2021; 8:568818. [PMID: 34095214 PMCID: PMC8173126 DOI: 10.3389/fmolb.2021.568818] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 04/22/2021] [Indexed: 12/24/2022] Open
Abstract
Screening of CRC continues to show poor compliance of endoscopy examination. The detection of mSEPT9 in peripheral blood is among the safe and simple early screening methods for CRC. The issue of how to elucidate whether detection of mSEPT9 in peripheral blood can effectively improve compliance of endoscopy and increase the early diagnosis rate of CRC and the relationship between levels of mSEPT9 in the peripheral blood and clinical stage, pathological classification, and expression of characteristic molecules in CRC remains unsolved. A total of 7759 individuals participated in the study that was performed using a questionnaire for screening of high-risk CRC. The endoscopic detection compliance of individuals with high-risk CRC who underwent the fecal occult blood test (FOBT) or mSEPT9 test was compared based on the results of the questionnaire. Additionally, correlation of mSEPT9 levels in the peripheral blood with clinicopathological features, mutation status of TP53, mismatch repair deficiency (dMMR), and KRAS/NRAS/BRAF/PIK3CA genotype was analyzed, and association of biomarkers with cancer-specific survival (CSS) and time to recurrence (TTR) was compared. We also detected levels of mSEPT9 in the peripheral blood of patients with CRC 7 days after surgery and compared the prognostic value of mSEPT9 with CEA. Results of our study showed that the mSEPT9 test could improve compliance of endoscopy and indicated a higher percentage of patients with positive mSEPT9 willing to undergo endoscopy detection than in those with positive FOBT. The specificity and sensitivity of mSEPT9 were better than that of FOBT for the detection of CRC. mSEPT9 was associated with the TNM stage, dMMR, and mutations in TP53, BRAF, and PIK3CA. A Ct value of mSEPT9 ≤ 37.5 was significantly related to poor CSS. mSEPT9 could affect association of dMMR and BRAF and PIK3CA mutations with CSS in a specific stage of CRC. The positive rate of mSEPT9 after surgery was found to correlate with poor TTR, and sensitivity was higher than CEA. The combination of mSEPT9 with CEA had a better prognostic value than that of mSEPT9 alone. The level of mSEPT9 was related to dMMR, mutations in TP53, BRAF, and PIK3CA, and was an effective biomarker for the prognosis of patients with CRC.
Collapse
Affiliation(s)
- Jie Sun
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Jinling Xu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Chao Sun
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Minying Zheng
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Yuwei Li
- Department of Colorectal Surgery, Tianjin Union Medical Center, Tianjin, China
| | - Siwei Zhu
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| | - Shiwu Zhang
- Department of Pathology, Tianjin Union Medical Center, Tianjin, China
| |
Collapse
|
549
|
Bateman AC. DNA mismatch repair protein immunohistochemistry - an illustrated guide. Histopathology 2021; 79:128-138. [PMID: 33829541 DOI: 10.1111/his.14367] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/09/2021] [Accepted: 03/19/2021] [Indexed: 01/28/2023]
Abstract
DNA mismatch repair (MMR) is an essential physiological process for correcting mutations occurring during DNA replication. Deficient MMR (dMMR) leads to increased tumour mutational burden, with a heightened risk of neoplasia. Demonstration of dMMR via the immunohistochemical assessment of MMR proteins is useful when screening for inherited cancer syndromes (especially Lynch syndrome) and for the prediction of clinical cancer response to conventional chemotherapy and novel immunotherapies. Identification of dMMR may also be helpful in other situations e.g. when considering a diagnosis of conventional dysplasia in sessile serrated lesions of the large intestine. This article provides a practical illustrated guide to DNA MMR interpretation, using a series of clinical examples.
Collapse
Affiliation(s)
- Adrian C Bateman
- Department of Cellular Pathology, University Hospitals Southampton NHS Foundation Trust, Southampton, UK
| |
Collapse
|
550
|
Sajjadi E, Venetis K, Piciotti R, Invernizzi M, Guerini-Rocco E, Haricharan S, Fusco N. Mismatch repair-deficient hormone receptor-positive breast cancers: Biology and pathological characterization. Cancer Cell Int 2021; 21:266. [PMID: 34001143 PMCID: PMC8130151 DOI: 10.1186/s12935-021-01976-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/07/2021] [Indexed: 12/15/2022] Open
Abstract
The clinical outcome of patients with a diagnosis of hormone receptor (HR)+ breast cancer has improved remarkably since the arrival of endocrine therapy. Yet, resistance to standard treatments is a major clinical challenge for breast cancer specialists and a life-threatening condition for the patients. In breast cancer, mismatch repair (MMR) status assessment has been demonstrated to be clinically relevant not only in terms of screening for inherited conditions such as Lynch syndrome, but also for prognostication, selection for immunotherapy, and early identification of therapy resistance. Peculiar traits characterize the MMR biology in HR+ breast cancers compared to other cancer types. In these tumors, MMR genetic alterations are relatively rare, occurring in ~3 % of cases. On the other hand, modifications at the protein level can be observed also in the absence of gene alterations and vice versa. In HR+ breast cancers, the prognostic role of MMR deficiency has been confirmed by several studies, but its predictive value remains a matter of controversy. The characterization of MMR status in these patients is troubled by the lack of tumor-specific guidelines and/or companion diagnostic tests. For this reason, precise identification of MMR-deficient breast cancers can be problematic. A deeper understanding of the MMR biology and clinical actionability in HR+ breast cancer may light the path to effective tumor-specific diagnostic tools. For a precise MMR status profiling, the specific strengths and limitations of the available technologies should be taken into consideration. This article aims at providing a comprehensive overview of the current state of knowledge of MMR alterations in HR+ breast cancer. The available armamentarium for MMR testing in these tumors is also examined along with possible strategies for a tailored pathological characterization.
Collapse
Affiliation(s)
- Elham Sajjadi
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Konstantinos Venetis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Roberto Piciotti
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Marco Invernizzi
- Physical and Rehabilitative Medicine, Department of Health Sciences, University of Eastern Piedmont, Viale Piazza D'Armi, 1, 28100, Novara, Italy
| | - Elena Guerini-Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy
| | - Svasti Haricharan
- Department of Tumor Microenvironment and Cancer Immunology, Sanford Burnham Prebys Medical Discovery Institute, 10901 N Torrey Pines Rd, 92037, La Jolla, CA, USA
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, University of Milan, Via Giuseppe Ripamonti 435, 20141, Milan, Italy.
- Department of Oncology and Hemato-Oncology, University of Milan, Via Festa del Perdono 7, 20122, Milan, Italy.
| |
Collapse
|