1
|
Matsubara J, Mukai K, Kondo T, Yoshioka M, Kage H, Oda K, Ikeda S, Ebi H, Muro K, Kajiura S, Hayashi R, Ashida R, Kitano M, Muto M. First-Line Genomic Profiling in Previously Untreated Advanced Solid Tumors: 1-Year Follow-Up of the FIRST-Dx Study. Cancer Sci 2025. [PMID: 40217582 DOI: 10.1111/cas.70077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/11/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025] Open
Abstract
The FIRST-Dx study prospectively evaluated the clinical utility of the comprehensive genomic profiling (CGP) test (FoundationOne CDx) in the first-line setting for patients with chemotherapy-naïve advanced solid tumors (gastrointestinal, biliary, pancreatic, lung, breast, gynecologic, melanoma) in six hospitals in Japan. Here, we report the results of the 1-year interim analysis of the follow-up study about the clinical benefits provided by the upfront CGP test. The primary endpoint was overall survival (OS), and secondary endpoints were the proportion of patients who actually received molecular-based recommended therapy (MBRT) determined by the molecular tumor board, best overall response rate (ORR) in each line of therapy, and progression-free survival (PFS) ratio (PFS on MBRT/PFS on the first-line therapy). Data from 172 patients with a median follow-up of 15.1 months (range: 0.1-21.5 months) were available. The median OS was not reached. Thirty-nine patients (22.7%) received MBRT during this follow-up period. ORR in first-line therapy was 56.3% in the MBRT group (n = 16) vs. 42.3% in the non-MBRT group (n = 137), and in the second-line was 26.3% in the MBRT group (n = 19) vs. 17.1% in the non-MBRT group (n = 82). Regarding the PFS ratio of second-line MBRT (n = 12), the median PFS ratio was 1.1, and four patients (33.3%) had a ratio ≥ 1.3, indicating that MBRT might be effective in changing the clinical outcome. The findings of this study imply that CGP testing before the standard of care for patients with advanced solid tumors could prove to be a clinically beneficial strategy for guiding subsequent precision anticancer treatments. Trial Registration: Japan Registry of Clinical Trials (jRCT) ID: jRCT1050220041.
Collapse
Affiliation(s)
- Junichi Matsubara
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Kumi Mukai
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Masahiro Yoshioka
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Hidenori Kage
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Katsutoshi Oda
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Shinya Kajiura
- Department of Clinical Oncology, Toyama University Hospital, Toyama, Japan
| | - Ryuji Hayashi
- Department of Clinical Oncology, Toyama University Hospital, Toyama, Japan
| | - Reiko Ashida
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Masayuki Kitano
- Second Department of Internal Medicine, Wakayama Medical University, Wakayama, Japan
| | - Manabu Muto
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
2
|
Hiken J, Earls J, Flanagan KC, Wellinghoff RL, Ponder M, Messina DN, Glasscock JI, Duncavage EJ. Analytical performance of OncoPrism-HNSCC, an RNA-based assay to inform immune checkpoint inhibitor treatment decisions for recurrent/metastatic head and neck squamous cell carcinoma. BMC Cancer 2025; 25:21. [PMID: 39773366 PMCID: PMC11705923 DOI: 10.1186/s12885-024-13362-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND While immune checkpoint inhibitor (ICI) therapies can significantly improve outcomes for patients with recurrent/metastatic head and neck squamous cell carcinoma (RM-HNSCC), only about 15-20% benefit from such treatments. Clinical tests that guide the use of ICIs are therefore critically needed. OncoPrism-HNSCC was developed to address this need. The assay combines next generation RNA sequencing-based immunomodulatory gene expression signatures with machine learning algorithms to generate an OncoPrism score that classifies patients as having low, medium, or high likelihood of disease control in response to ICI treatment. Also, OncoPrism-HNSCC leverages the same FFPE patient tumor RNA used for ICI response prediction to identify rare cases where oncogenic rearrangements in NTRK1/2/3 or ALK genes may occur, and which may indicate the use of potentially highly effective targeted therapies. The clinical performance of OncoPrism-HNSCC has been validated. Here, we report its analytical performance in the presence of potentially confounding sources of variation. METHODS The assay's analytical sensitivity was assessed by varying RNA input quantity and quality, observing the effect on ICI response prediction scores. Analytical specificity was tested by spiking increasing percentages of genomic DNA into input RNA. Intra-assay and inter-assay precision were evaluated, and the analytical sensitivity, specificity, and precision of gene fusion detection were assessed. Concordance with orthogonal methods of gene fusion detection was tested on 67 FFPE clinical samples. RESULTS Varying RNA inputs as low as four-fold below the nominal input amount had little effect on ICI response prediction scores. RNA quality levels below the test threshold had no significant effect. Genomic DNA spike-ins up to 30% had only a small effect on scores. The pooled standard deviation for multiple operators, reagent lots, batches, and sequencers yielded an overall variance represented by just 0.87% of the score range of the test (0-100). NTRK and ALK gene fusion detection was 100% concordant with orthogonal methods. CONCLUSIONS Robust and reliable analytical performance of the OncoPrism-HNSCC assay supports its clinical use, even in the presence of variation typically encountered in the laboratory setting.
Collapse
Affiliation(s)
- Jeffrey Hiken
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | - Jon Earls
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | - Kevin C Flanagan
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | | | - Michelle Ponder
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | - David N Messina
- Cofactor Genomics, Inc, 4044 Clayton Ave, St. Louis, MO, 63110, USA
| | | | - Eric J Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
3
|
Mete O, Boucher A, Schrader KA, Abdel-Rahman O, Bahig H, Ho C, Hasan OK, Lemieux B, Winquist E, Wong R, Wu J, Chau N, Ezzat S. Consensus Statement: Recommendations on Actionable Biomarker Testing for Thyroid Cancer Management. Endocr Pathol 2024; 35:293-308. [PMID: 39579327 DOI: 10.1007/s12022-024-09836-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2024] [Indexed: 11/25/2024]
Abstract
Thyroid cancer management is rapidly changing. The identification of actionable biomarkers through both germline and somatic testing are now an integral part of directing patient management. However, deficiencies and disparities within existing thyroid cancer biomarker test approaches are resulting in inconsistent application for patient care. An expert panel was convened to create consensus biomarker testing algorithms and recommendations on actionable biomarker testing for patients diagnosed with medullary thyroid cancer, non-anaplastic follicular cell-derived thyroid cancer, or anaplastic follicular cell-derived thyroid cancer who may benefit from targeted therapies. A review of international guidelines was performed to determine the current state, and a literature review was carried out to further evaluate the evidence supporting the use of actionable biomarkers in patients diagnosed with thyroid cancer. Thyroid biomarker-related gaps impacting patient care were also discussed, with an emphasis on the importance of a multidisciplinary team approach for optimal patient care. The recommendations are presented with the aim to help physicians navigate the current thyroid cancer biomarker testing landscape with its many challenges, balancing aspirational care with what is practical and feasible in terms of economic realities and jurisdictional constraints. By remaining therapy-agnostic, these algorithms and recommendations are broadly applicable.
Collapse
Affiliation(s)
- Ozgur Mete
- Department of Pathology, University Health Network, Toronto General Hospital, University of Toronto, Toronto, Ontario, M5G 2C4, Canada.
| | - Andrée Boucher
- Department of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | | | - Omar Abdel-Rahman
- Department of Oncology, University of Alberta, Edmonton, Alberta, Canada
| | - Houda Bahig
- Department of Radiation Oncology, Centre Hospitalier de L'Université de Montréal, Montreal, Quebec, Canada
| | - Cheryl Ho
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Olfat Kamel Hasan
- Department of Medicine and Department of Medical Imaging, McMaster University, Hamilton, Ontario, Canada
| | - Bernard Lemieux
- Department of Internal Medicine, Centre Hospitalier Universite de Montreal, Montreal, Quebec, Canada
| | - Eric Winquist
- Department of Oncology, Western University, London, Ontario, Canada
| | - Ralph Wong
- Department of Oncology and Hematology, Cancer Care Manitoba, University of Manitoba, Winnipeg, Canada
| | - Jonn Wu
- Department of Radiation Oncology, BC Cancer Agency, Vancouver, British Columbia, Canada
| | - Nicole Chau
- Department of Medical Oncology, BC Cancer, Vancouver, British Columbia, Canada
| | - Shereen Ezzat
- Endocrine Oncology Site Group, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| |
Collapse
|
4
|
La Salvia A, Meyer ML, Hirsch FR, Kerr KM, Landi L, Tsao MS, Cappuzzo F. Rediscovering immunohistochemistry in lung cancer. Crit Rev Oncol Hematol 2024; 200:104401. [PMID: 38815876 DOI: 10.1016/j.critrevonc.2024.104401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 05/23/2024] [Indexed: 06/01/2024] Open
Abstract
Several observations indicate that protein expression analysis by immunohistochemistry (IHC) remains relevant in individuals with non-small-cell lung cancer (NSCLC) when considering targeted therapy, as an early step in diagnosis and for therapy selection. Since the advent of next-generation sequencing (NGS), the role of IHC in testing for NSCLC biomarkers has been forgotten or ignored. We discuss how protein-level investigations maintain a critical role in defining sensitivity to lung cancer therapies in oncogene- and non-oncogene-addicted cases and in patients eligible for immunotherapy, suggesting that IHC testing should be reconsidered in clinical practice. We also argue how a panel of IHC tests should be considered complementary to NGS and other genomic assays. This is relevant to current clinical diagnostic practice but with potential future roles to optimize the selection of patients for innovative therapies. At the same time, strict validation of antibodies, assays, scoring systems, and intra- and interobserver reproducibility is needed.
Collapse
Affiliation(s)
- Anna La Salvia
- National Center for Drug Research and Evaluation, National Institute of Health (ISS), Rome 00161, Italy
| | - May-Lucie Meyer
- Center for Thoracic Oncology/Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Fred R Hirsch
- Center for Thoracic Oncology/Tisch Cancer Institute and Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Keith M Kerr
- Aberdeen University School of Medicine & Aberdeen Royal Infirmary, Aberdeen, UK
| | - Lorenza Landi
- Medical Oncology, Istituto Nazionale Tumori IRCCS "Regina Elena", Rome, Italy
| | - Ming-Sound Tsao
- University Health Network, Princess Margaret Cancer Centre, Toronto, Ontario, Canada
| | - Federico Cappuzzo
- Medical Oncology, Istituto Nazionale Tumori IRCCS "Regina Elena", Rome, Italy.
| |
Collapse
|
5
|
Kim JY, Park K, Park WY, Ahn JS, Im YH, Lee JE, Kim SW, Nam SJ, Yu J, Park YH. Prognostic value of structural variants in early breast cancer patients. NPJ Breast Cancer 2024; 10:64. [PMID: 39068172 PMCID: PMC11283467 DOI: 10.1038/s41523-024-00669-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Accepted: 07/08/2024] [Indexed: 07/30/2024] Open
Abstract
Genomic analysis of structural variants(SVs) in breast cancer (BC) patients has been conducted, but the relationship between genomic alterations and BC prognosis remains unclear. We performed RNA sequencing of 297 early BC fresh-frozen tissues. We identified SVs using three tools (STAR.Arriba, STAR.fusion, and STAR.SEQR) with the COSMIC and Mitelman databases as guide references. We found a median of five to eight fusions per sample. In BC intrinsic subtypes, normal subtype had the fewest fusions (median: 1, interquartile range [IQR]: 0, 3) followed by luminal A (median: 5.5, IQR: 2.75, 10.25), luminal B (median: 9, IQR: 6, 16.5), HER2-enriched (median: 9, IQR: 6, 16.5) and basal (median 10, IQR: 6, 15.5) subtypes (p < 0.05). Intrachromosomal fusion was more frequent observed rather than interchromosomal fusion. In location, chromosome 17 had the most fusions followed by chromosome 1 and 11. When samples were divided into high and low fusion groups based on a cut-off value of 11 fusions, five-year event-free survival (5Y-EFS) was 68.1% in the high fusion group (n = 72) and 80.1% in the low fusion group (n = 125) (p = 0.024) while 75.6% among all patients (95% confidence interval: 0.699, 0.819). Among BC subtype, TNBCs with more fusions had shorter EFS compared to those with fewer fusions (5Y-EFS rate: 65.1% vs. 85.7%; p = 0.013) but no EFS differences were observed in other BC subtypes. ESTIMATE ImmuneScore was also associated with the number of fusions in TNBC (p < 0.005) and TNBCs with high ImmuneScore had better 5Y-EFS compared to those with low ImmuneScore (p = 0.041). In conclusion, diverse fusions were observed by BC subtype, and the number of fusions was associated with BC survival outcome and immune status in TNBC.
Collapse
Affiliation(s)
- Ji-Yeon Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Kyunghee Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical Center, Seoul, Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Hyuck Im
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jeong Eon Lee
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Won Kim
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seok Jin Nam
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jonghan Yu
- Department of Surgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University School of Medicine, Seoul, Korea.
| |
Collapse
|
6
|
Zhang W, Schmitz AA, Kallionpää RE, Perälä M, Pitkänen N, Tukiainen M, Alanne E, Jöhrens K, Schulze-Rath R, Farahmand B, Zong J. Neurotrophic tyrosine receptor kinase gene fusions in adult and pediatric patients with solid tumors: a clinicogenomic biobank and record linkage study of expression frequency and patient characteristics from Finland. Acta Oncol 2024; 63:542-551. [PMID: 38967220 PMCID: PMC11332464 DOI: 10.2340/1651-226x.2024.26452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 05/14/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND Neurotrophic tyrosine receptor kinase (NTRK) gene fusions are oncogenic drivers. Using the Auria Biobank in Finland, we aimed to identify and characterize patients with these gene fusions, and describe their clinical and tumor characteristics, treatments received, and outcomes. MATERIAL AND METHODS We evaluated pediatrics with any solid tumor type and adults with colorectal cancer (CRC), non-small cell lung cancer (NSCLC), sarcoma, or salivary gland cancer. We determined tropomyosin receptor kinase (TRK) protein expression by pan-TRK immunohistochemistry (IHC) staining of tumor samples from the Auria Biobank, scored by a certified pathologist. NTRK gene fusion was confirmed by next generation sequencing (NGS). All 2,059 patients were followed-up starting 1 year before their cancer diagnosis. RESULTS Frequency of NTRK gene fusion tumors was 3.1% (4/127) in pediatrics, 0.7% (8/1,151) for CRC, 0.3% (1/288) for NSCLC, 0.9% (1/114) for salivary gland cancer, and 0% (0/379) for sarcoma. Among pediatrics there was one case each of fibrosarcoma (TPM3::NTRK1), Ewing's sarcoma (LPPR1::NTRK2), primitive neuroectodermal tumor (DAB2IP::NTRK2), and papillary thyroid carcinoma (RAD51B::NTRK3). Among CRC patients, six harbored tumors with NTRK1 fusions (three fused with TPM3), one harbored a NTRK3::GABRG1 fusion, and the other a NTRK2::FXN/LPPR1 fusion. Microsatellite instability was higher in CRC patients with NTRK gene fusion tumors versus wild-type tumors (50.0% vs. 4.4%). Other detected fusions were SGCZ::NTRK3 (NSCLC) and ETV6::NTRK3 (salivary gland cancer). Four patients (three CRC, one NSCLC) received chemotherapy; one patient (with CRC) received radiotherapy. CONCLUSION NTRK gene fusions are rare in adult CRC, NSCLC, salivary tumors, sarcoma, and pediatric solid tumors.
Collapse
Affiliation(s)
| | | | - Roosa E Kallionpää
- Auria Biobank, University of Turku and Turku University Hospital, Turku, Finland
| | - Merja Perälä
- Auria Biobank, University of Turku and Turku University Hospital, Turku, Finland
| | - Niina Pitkänen
- Auria Biobank, University of Turku and Turku University Hospital, Turku, Finland
| | - Mikko Tukiainen
- Auria Biobank, University of Turku and Turku University Hospital, Turku, Finland
| | - Erika Alanne
- Department of Oncology and Radiotherapy, Turku University Hospital, Turku, Finland; Western Finland Cancer Centre, Turku, Finland
| | - Korinna Jöhrens
- Dresden University Hospital, Technical University Dresden, Dresden, Germany
| | | | | | | |
Collapse
|
7
|
Repetto M, Chiara Garassino M, Loong HH, Lopez-Rios F, Mok T, Peters S, Planchard D, Popat S, Rudzinski ER, Drilon A, Zhou C. NTRK gene fusion testing and management in lung cancer. Cancer Treat Rev 2024; 127:102733. [PMID: 38733648 DOI: 10.1016/j.ctrv.2024.102733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 05/13/2024]
Abstract
Neurotrophic tyrosine receptor kinase (NTRK) gene fusions are recurrent oncogenic drivers found in a variety of solid tumours, including lung cancer. Several tropomyosin receptor kinase (TRK) inhibitors have been developed to treat tumours with NTRK gene fusions. Larotrectinib and entrectinib are first-generation TRK inhibitors that have demonstrated efficacy in patients with TRK fusion lung cancers. Genomic testing is recommended for all patients with metastatic non-small cell lung cancer for optimal drug therapy selection. Multiple testing methods can be employed to identify NTRK gene fusions in the clinic and each has its own advantages and limitations. Among these assays, RNA-based next-generation sequencing (NGS) can be considered a gold standard for detecting NTRK gene fusions; however, several alternatives with minimally acceptable sensitivity and specificity are also available in areas where widespread access to NGS is unfeasible. This review highlights the importance of testing for NTRK gene fusions in lung cancer, ideally using the gold-standard method of RNA-based NGS, the various assays that are available, and treatment algorithms for patients.
Collapse
Affiliation(s)
- Matteo Repetto
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA.
| | - Marina Chiara Garassino
- Department of Medicine, Thoracic Oncology Program, The University of Chicago, Chicago, IL, USA
| | | | | | - Tony Mok
- The Chinese University of Hong Kong, Hong Kong, China
| | - Solange Peters
- Centre Hospitalier Universitaire Vaudois, Lausanne University, Lausanne, Switzerland
| | | | - Sanjay Popat
- Royal Marsden NHS Foundation Trust, London, UK; Institute of Cancer Research, London, UK
| | - Erin R Rudzinski
- Seattle Children's Hospital and University of Washington Medical Center, Seattle, WA, USA
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, New York, NY, USA; Weill Cornell Medical College, New York, NY, USA
| | - Caicun Zhou
- Shanghai Pulmonary Hospital, Tongji University, Shanghai, China
| |
Collapse
|
8
|
Pathak PS, Chan G, Deming DA, Chee CE. State-of-the-Art Management of Colorectal Cancer: Treatment Advances and Innovation. Am Soc Clin Oncol Educ Book 2024; 44:e438466. [PMID: 38768405 DOI: 10.1200/edbk_438466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Colorectal cancer (CRC) remains a significant global health challenge, ranking among the leading causes of cancer-related morbidity and mortality worldwide. Recent advancements in molecular characterization have revolutionized our understanding of the heterogeneity within colorectal tumors, particularly in the context of tumor sidedness. Tumor sidedness, referring to the location of the primary tumor in either the right or left colon, has emerged as a critical factor influencing prognosis and treatment responses in metastatic CRC. Molecular underpinnings of CRC, the impact of tumor sidedness, and how this knowledge guides therapeutic decisions in the era of precision medicine have led to improved outcomes and better quality of life in patients. The emergence of circulating tumor DNA as a prognostic and predictive tool in CRC heralds promising advancements in the diagnosis and monitoring of the disease. This innovation facilitates better patient selection for exploration of additional treatment options. As the field progresses, with investigational agents demonstrating potential as future treatments for refractory metastatic CRC, new avenues for enhancing outcomes in this challenging disease are emerging.
Collapse
Affiliation(s)
- Priyadarshini S Pathak
- Division of Hematology/Oncology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Gloria Chan
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore
| | - Dustin A Deming
- Division of Hematology, Medical Oncology, and Palliative Care, Department of Medicine, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
- University of Wisconsin Carbone Cancer Center, Madison, WI
- McArdle Laboratory for Cancer Research, Department of Oncology, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI
| | - Cheng Ean Chee
- Department of Hematology-Oncology, National University Cancer Institute, Singapore, National University Health System, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| |
Collapse
|
9
|
Hernandez S, Conde E, Molero A, Suarez-Gauthier A, Martinez R, Alonso M, Plaza C, Camacho C, Chantada D, Juaneda-Magdalena L, Garcia-Toro E, Saiz-Lopez P, Rojo F, Abad M, Boni V, Del Carmen S, Regojo RM, Sanchez-Frias ME, Teixido C, Paz-Ares L, Lopez-Rios F. Efficient Identification of Patients With NTRK Fusions Using a Supervised Tumor-Agnostic Approach. Arch Pathol Lab Med 2024; 148:318-326. [PMID: 37270803 DOI: 10.5858/arpa.2022-0443-oa] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2023] [Indexed: 06/06/2023]
Abstract
CONTEXT.— The neurotrophic tropomyosin receptor kinase (NTRK) family gene rearrangements have been recently incorporated as predictive biomarkers in a "tumor-agnostic" manner. However, the identification of these patients is extremely challenging because the overall frequency of NTRK fusions is below 1%. Academic groups and professional organizations have released recommendations on the algorithms to detect NTRK fusions. The European Society for Medical Oncology proposal encourages the use of next-generation sequencing (NGS) if available, or alternatively immunohistochemistry (IHC) could be used for screening with NGS confirmation of all positive IHC results. Other academic groups have included histologic and genomic information in the testing algorithm. OBJECTIVE.— To apply some of these triaging strategies for a more efficient identification of NTRK fusions within a single institution, so pathologists can gain practical insight on how to start looking for NTRK fusions. DESIGN.— A multiparametric strategy combining histologic (secretory carcinomas of the breast and salivary gland; papillary thyroid carcinomas; infantile fibrosarcoma) and genomic (driver-negative non-small cell lung carcinomas, microsatellite instability-high colorectal adenocarcinomas, and wild-type gastrointestinal stromal tumors) triaging was put forward. RESULTS.— Samples from 323 tumors were stained with the VENTANA pan-TRK EPR17341 Assay as a screening method. All positive IHC cases were simultaneously studied by 2 NGS tests, Oncomine Comprehensive Assay v3 and FoundationOne CDx. With this approach, the detection rate of NTRK fusions was 20 times higher (5.57%) by only screening 323 patients than the largest cohort in the literature (0.30%) comprising several hundred thousand patients. CONCLUSIONS.— Based on our findings, we propose a multiparametric strategy (ie, "supervised tumor-agnostic approach") when pathologists start searching for NTRK fusions.
Collapse
Affiliation(s)
- Susana Hernandez
- From the Department of Pathology, 12 de Octubre University Hospital, Research Institute 12 de Octubre University Hospital (i+12), Madrid, Spain (Hernandez, Alonso)
| | - Esther Conde
- the Department of Pathology, 12 de Octubre University Hospital, Universidad Complutense de Madrid, Research Institute 12 de Octubre University Hospital (i+12), CIBERONC, Madrid, Spain (Conde, Lopez-Rios)
| | - Aida Molero
- the Department of Pathology, Segovia General Hospital, Segovia, Spain (Molero)
| | - Ana Suarez-Gauthier
- the Department of Pathology, Jimenez Diaz Foundation University Hospital, Madrid, Spain (Suarez-Gauthier)
| | - Rebeca Martinez
- the Department of Pathology, Health Diagnostic-Grupo Quiron Salud, Madrid, Spain (Martinez)
| | - Marta Alonso
- From the Department of Pathology, 12 de Octubre University Hospital, Research Institute 12 de Octubre University Hospital (i+12), Madrid, Spain (Hernandez, Alonso)
| | - Carlos Plaza
- the Department of Pathology, Clinico San Carlos University Hospital, Madrid, Spain (Plaza)
| | - Carmen Camacho
- the Department of Pathology, Insular Materno-Infantil University Hospital, Las Palmas de Gran Canaria, Spain (Camacho)
| | - Debora Chantada
- the Department of Pathology, Alvaro Cunqueiro Hospital, Vigo, Spain (Chantada, Juaneda-Magdalena)
| | - Laura Juaneda-Magdalena
- the Department of Pathology, Alvaro Cunqueiro Hospital, Vigo, Spain (Chantada, Juaneda-Magdalena)
| | - Enrique Garcia-Toro
- the Department of Pathology, Burgos University Hospital, Burgos, Spain (Garcia-Toro, Saiz-Lopez)
| | - Patricia Saiz-Lopez
- the Department of Pathology, Burgos University Hospital, Burgos, Spain (Garcia-Toro, Saiz-Lopez)
| | - Federico Rojo
- the Institute of Health Research-Jimenez Diaz Foundation, CIBERONC, Madrid, Spain (Rojo)
| | - Mar Abad
- the Department of Pathology, Salamanca University Hospital, Salamanca, Spain (Abad)
| | - Valentina Boni
- NEXT Oncology Madrid, Quiron Salud Madrid University Hospital, Madrid, Spain (Boni)
| | - Sofia Del Carmen
- the Department of Pathology, Marques de Valdecilla University Hospital, Santander, Spain (del Carmen)
| | - Rita Maria Regojo
- the Department of Pathology, La Paz University Hospital, Madrid, Spain (Regojo)
| | | | - Cristina Teixido
- the Department of Pathology, Thoracic Oncology Unit, Hospital Clínic of Barcelona, University of Barcelona, Barcelona, Spain (Teixido)
| | - Luis Paz-Ares
- the Department of Oncology, 12 de Octubre University Hospital, Department of Medicine, Universidad Complutense de Madrid, Research Institute 12 de Octubre University Hospital (i+12), CIBERONC, Madrid, Spain (Paz-Ares)
| | - Fernando Lopez-Rios
- the Department of Pathology, 12 de Octubre University Hospital, Universidad Complutense de Madrid, Research Institute 12 de Octubre University Hospital (i+12), CIBERONC, Madrid, Spain (Conde, Lopez-Rios)
| |
Collapse
|
10
|
Nádorvári ML, Kenessey I, Kiss A, Barbai T, Kulka J, Rásó E, Tímár J. Comparison of standard mismatch repair deficiency and microsatellite instability tests in a large cancer series. J Transl Med 2024; 22:150. [PMID: 38350968 PMCID: PMC10863158 DOI: 10.1186/s12967-024-04960-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/07/2024] [Indexed: 02/15/2024] Open
Abstract
BACKGROUND The tumor-agnostic indication of immune checkpoint inhibitors to treat cancers with mismatch repair deficiency (dMMR)/microsatellite instability (MSI) increased the demand for such tests beyond Lynch syndrome. International guideline recommendations accept immunohistochemistry (IHC) for dMMR or molecular techniques (PCR or NGS) for MSI status determinations considering the two tests are equal, although there are scattered reports contradicting to this presumption. MATERIALS AND METHODS Here we have directly compared four protein MMR immunohistochemistry (IHC) to MSI Pentaplex PCR test in a large cancer patient cohort (n = 1306) of our diagnostic center where the two tests have been run parallel in 703 cases. RESULTS In this study we have found a high discrepancy rate (19.3%) of the two tests which was independent of the tumor types. The MSI PCR sensitivity for MMR IHC status was found to be very low resulting in a relatively low positive and negative predicting values. As a consequence, the correlation of the two tests was low (kappa < 0.7). During analysis of the possible contributing factors of this poor performance, we have excluded low tumor percentage of the samples, but identified dMMR phenotypes (classic versus non-classic or unusual) as possible contributors. CONCLUSION Although our cohort did not include samples with identified technical errors, our data strongly support previous reports that unidentified preanalytical factors might have the major influence on the poor performance of the MSI PCR and MMR IHC. Furthermore, the case is open whether the two test types are equally powerful predictive markers of immunotherapies.
Collapse
Affiliation(s)
- Maja L Nádorvári
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - István Kenessey
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - András Kiss
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Tamás Barbai
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Janina Kulka
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - Erzsébet Rásó
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary
| | - József Tímár
- Department of Pathology, Forensic and Insurance Medicine, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
11
|
Hyrcza MD, Martins-Filho SN, Spatz A, Wang HJ, Purgina BM, Desmeules P, Park PC, Bigras G, Jung S, Cutz JC, Xu Z, Berman DM, Sheffield BS, Cheung CC, Leduc C, Hwang DM, Ionescu D, Klonowski P, Chevarie-Davis M, Chami R, Lo B, Stockley TL, Tsao MS, Torlakovic E. Canadian Multicentric Pan-TRK (CANTRK) Immunohistochemistry Harmonization Study. Mod Pathol 2024; 37:100384. [PMID: 37972928 DOI: 10.1016/j.modpat.2023.100384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/19/2023] [Accepted: 11/09/2023] [Indexed: 11/19/2023]
Abstract
Tumor-agnostic testing for NTRK1-3 gene rearrangements is required to identify patients who may benefit from TRK inhibitor therapies. The overarching objective of this study was to establish a high-quality pan-TRK immunohistochemistry (IHC) screening assay among 18 large regional pathology laboratories across Canada using pan-TRK monoclonal antibody clone EPR17341 in a ring study design. TRK-fusion positive and negative tumor samples were collected from participating sites, with fusion status confirmed by panel next-generation sequencing assays. Each laboratory received: (1) unstained sections from 30 cases of TRK-fusion-positive or -negative tumors, (2) 2 types of reference standards: TRK calibrator slides and IHC critical assay performance controls (iCAPCs), (3) EPR17341 antibody, and (4) suggestions for developing IHC protocols. Participants were asked to optimize the IHC protocol for their instruments and detection systems by using iCAPCs, to stain the 30 study cases, and to report the percentage scores for membranous, cytoplasmic, and nuclear staining. TRK calibrators were used to assess the analytical sensitivity of IHC protocols developed by using the 2 reference standards. Fifteen of 18 laboratories achieved diagnostic sensitivity of 100% against next-generation sequencing. The diagnostic specificity ranged from 40% to 90%. The results did not differ significantly between positive scores based on the presence of any type of staining vs the presence of overall staining in ≥1% of cells. The median limit of detection measured by TRK calibrators was 76,000 molecules/cell (range 38,000 to >200,000 molecules/cell). Three different patterns of staining were observed in 19 TRK-positive cases, cytoplasmic-only in 7 samples, nuclear and cytoplasmic in 9 samples, and cytoplasmic and membranous in 3 samples. The Canadian multicentric pan-TRK study illustrates a successful strategy to accelerate the multicenter harmonization and implementation of pan-TRK immunohistochemical screening that achieves high diagnostic sensitivity by using laboratory-developed tests where laboratories used centrally developed reference materials. The measurement of analytical sensitivity by using TRK calibrators provided additional insights into IHC protocol performance.
Collapse
Affiliation(s)
- Martin D Hyrcza
- Department of Pathology and Laboratory Medicine, University of Calgary, Arnie Charbonneau Cancer Institute, Calgary, Alberta, Canada
| | - Sebastiao N Martins-Filho
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada; University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Alan Spatz
- McGill University Health Center, Lady Davis Institute, McGill University, Montreal, Quebec, Canada
| | - Han-Jun Wang
- McGill University Health Center, Lady Davis Institute, McGill University, Montreal, Quebec, Canada
| | - Bibianna M Purgina
- Department of Pathology and Laboratory Medicine, Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Patrice Desmeules
- Institut Universitaire de Cardiologie et de Pneumologie de Québec, Université Laval, Quebec City, Quebec, Canada
| | - Paul C Park
- Shared Health, Department of Pathology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Gilbert Bigras
- Department of Laboratory Medicine and Pathology, University of Alberta, Edmonton, Alberta, Canada
| | - Sungmi Jung
- Department of Pathology, McGill University Health Centre, McGill University, Montreal, Quebec, Canada
| | - Jean-Claude Cutz
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Zhaolin Xu
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - David M Berman
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Brandon S Sheffield
- Department of Pathology, William Osler Health System, Brampton, Ontario, Canada
| | - Carol C Cheung
- University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Charles Leduc
- Centre Hospitalier de l'Université de Montréal, Montreal, Quebec, Canada
| | - David M Hwang
- Department of Laboratory Medicine and Pathobiology, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | - Diana Ionescu
- Department of Pathology and Laboratory Medicine, British Columbia Cancer Agency, University of British Columbia, Vancouver, British Columbia, Canada
| | - Paul Klonowski
- Department of Pathology and Laboratory Medicine, University of Calgary Cumming School of Medicine Diagnostic and Scientific Centre, Calgary, Alberta, Canada
| | - Myriam Chevarie-Davis
- Département de Pathologie et Biologie Cellulaire, Hôpital Maisonneuve-Rosemont, Université de Montréal, Montreal, Quebec, Canada
| | - Rose Chami
- Department of Laboratory Medicine and Pathobiology, The Hospital for Sick Children, University of Toronto, Toronto, Ontario, Canada
| | - Bryan Lo
- Department of Pathology and Laboratory Medicine, Ottawa Hospital, University of Ottawa, Ottawa, Ontario, Canada
| | - Tracy L Stockley
- University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Ming-Sound Tsao
- University Health Network, Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| | - Emina Torlakovic
- Department of Pathology and Laboratory Medicine, Royal University Hospital, Saskatchewan Health Authority, and College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.
| |
Collapse
|
12
|
Kikuchi Y, Shimada H, Hatanaka Y, Kinoshita I, Ikarashi D, Nakatsura T, Kitano S, Naito Y, Tanaka T, Yamashita K, Oshima Y, Nanami T. Clinical practice guidelines for molecular tumor markers, 2nd edition review part 1. Int J Clin Oncol 2024; 29:1-19. [PMID: 38019341 DOI: 10.1007/s10147-023-02430-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 10/14/2023] [Indexed: 11/30/2023]
Abstract
With advances in gene and protein analysis technologies, many target molecules that may be useful in cancer diagnosis have been reported. Therefore, the "Tumor Marker Study Group" was established in 1981 with the aim of "discovering clinically" useful molecules. Later, the name was changed to "Japanese Society for Molecular Tumor Marker Research" in 2000 in response to the remarkable progress in gene-related research. Currently, the world of cancer treatment is shifting from the era of representative tumor markers of each cancer type used for tumor diagnosis and treatment evaluation to the study of companion markers for molecular-targeted therapeutics that target cancer cells. Therefore, the first edition of the Molecular Tumor Marker Guidelines, which summarizes tumor markers and companion markers in each cancer type, was published in 2016. After publication of the first edition, the gene panel testing using next-generation sequencing became available in Japan in June 2019 for insured patients. In addition, immune checkpoint inhibitors have been indicated for a wide range of cancer types. Therefore, the 2nd edition of the Molecular Tumor Marker Guidelines was published in September 2021 to address the need to revise the guidelines. Here, we present an English version of the review (Part 1) of the Molecular Tumor Marker Guidelines, Second Edition.
Collapse
Affiliation(s)
| | - Hideaki Shimada
- Department of Clinical Oncology, Toho University, Tokyo, Japan.
- Division of General and Gastroenterological Surgery, Department of Surgery (Omori), Toho University, Tokyo, Japan.
| | - Yutaka Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Kinoshita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Hokkaido, Japan
| | - Daiki Ikarashi
- Department of Urology, Iwate Medical University, Iwate, Japan
| | - Tetsuya Nakatsura
- Division of Cancer Immunotherapy, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Shigehisa Kitano
- Department of Advanced Medical Development, The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yoichi Naito
- Department of General Internal Medicine, National Cancer Center Hospital East, Chiba, Japan
| | - Toshimichi Tanaka
- Department of Lower Gastrointestinal Surgery, Kitasato University School of Medicine, Tokyo, Japan
| | - Keishi Yamashita
- Division of Advanced Surgical Oncology, Department of Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Tokyo, Japan
| | - Yoko Oshima
- Division of General and Gastroenterological Surgery, Department of Surgery (Omori), Toho University, Tokyo, Japan
| | - Tatsuki Nanami
- Division of General and Gastroenterological Surgery, Department of Surgery (Omori), Toho University, Tokyo, Japan
| |
Collapse
|
13
|
Yamada CAF, Malheiros SMF, Do Amaral LLF, Lancellotti CLP. SOMATIC DEFICIENT MISMATCH REPAIR ASSESSED BY IMMUNOHISTOCHEMISTRY AND CLINICAL FEATURES IN BRAZILIAN GLIOBLASTOMA PATIENTS. Exp Oncol 2023; 45:297-311. [PMID: 38186025 DOI: 10.15407/exp-oncology.2023.03.297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Indexed: 01/09/2024]
Abstract
BACKGROUND Glioblastoma (GBM) is the most frequent primary malignant CNS tumor. Deficient mismatch repair (dMMR) is associated with better prognosis and is a biomarker for immunotherapy. Evaluation of MMR by immunohistochemistry (IHC) is accessible, cost effective, sensitive, and specific. AIM Our objective was to investigate MMR proteins in adult GBM patients. MATERIALS AND METHODS We retrospectively analyzed 68 GBM samples to evaluate the proficiency of MMR genes expression assessed by IHC. Clinicopathologic and molecular features were compared in proficient (pMMR) or dMMR. RESULTS 10 (14.7%) samples showed dMMR, and the most frequent was MSH6 (100%) followed by MSH2, PMS2, and MLH1. We observed heterogeneous expression of dMMR in 5 GBMs. The median overall survival did not differ between pMMR (19.8 months; 0.2-30) and dMMR (16.9 months; 6.4-27.5) (p = 0.31). We observed a significantly higher overall survival associated with gross total resection compared to subtotal resection or biopsy (30.7 vs. 13.6 months, p = 0.02) and MGMT methylated status (29.6 vs. 19.8 months, p = 0.049). At the analysis time, 10 patients were still alive, all in the pMMR group. CONCLUSIONS Our data demonstrated dMMR phenotype assessed by IHC in an expressive portion of GBM patients, however without significant impact on overall survival.
Collapse
Affiliation(s)
- C A F Yamada
- Hospital Beneficência Portuguesa de São Paulo, São Paulo, Brazil
- Latin American Cooperative Oncology Group (LACOG), Porto Alegre, Brazil
- Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | | | - L L F Do Amaral
- Hospital Beneficência Portuguesa de São Paulo, São Paulo, Brazil
- Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
| | - C L P Lancellotti
- Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, Brazil
- Carmen Lucia Penteado Lancellotti Neuropathology Laboratory, São Paulo, Brazil
| |
Collapse
|
14
|
Cuello M, García-Rivello H, Huamán-Garaicoa F, Irigoyen-Piñeiros P, Lara-Torres CO, Rizzo MM, Ticona-Castro M, Trejo R, Zoroquiain P. Detection of NTRK gene fusions in solid tumors: recommendations from a Latin American group of oncologists and pathologists. Future Oncol 2023; 19:2669-2682. [PMID: 38088163 DOI: 10.2217/fon-2023-0552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
NTRK gene fusions have been detected in more than 25 types of tumors and their prevalence is approximately 0.3% in solid tumors. This low prevalence makes identifying patients who could benefit from TRK inhibitors a considerable challenge. Furthermore, while numerous papers on the evaluation of NTRK fusion genes are available, not all countries have guidelines that are suitable for their setting, as is the case with Latin America. Therefore, a group of oncologists and pathologists from several countries in Latin America (Argentina, Chile, Ecuador, Mexico, Peru and Uruguay) met to discuss and reach consensus on how to identify patients with NTRK gene fusions in solid tumors. To do so, they developed a practical algorithm, considering their specific situation and limitations.
Collapse
Affiliation(s)
- Mauricio Cuello
- Academic Unit of Oncology, Hospital de Clínicas Dr. Manuel Quintela, Montevideo, Uruguay
| | - Hernán García-Rivello
- Departmento of Clinical Pathology, Instituto de Medicina Traslacional e Ingeniería Biomédica (IMTIB), Hospital Italiano, Buenos Aires, Argentina
| | - Fuad Huamán-Garaicoa
- Instituto de Salud Integral (ISAIN), Universidad Católica, Santiago de Guayaquil (Ecuador), Department of Pathology, Sociedad de Lucha Contra el Cáncer del Ecuador (SOLCA), Guayaquil, Ecuador
| | | | - César O Lara-Torres
- Laboratory of Molecular Pathology, Instituto Nacional de Cancerología, Ciudad de México, México
| | - Manglio M Rizzo
- Cancer Immunobiology Laboratory, Instituto de Investigaciones en Medicina Traslacional (IIMT), Facultad de Ciencias Biomédicas, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Universidad Austral, Derqui-Pilar, Argentina
- Department of Medical Oncology, Hospital Universitario Austral, Derqui-Pilar, Argentina
| | - Miguel Ticona-Castro
- Service of Medical Oncology, Hospital Nacional Edgardo Rebagliati Martins, EsSalud - Jesús María, Lima (Perú), Clínica Montefiori, La Molina, Lima, Perú
| | - Rogelio Trejo
- Department of Medical Oncology, Centro Médico Nacional Siglo XXI, Ciudad de México, México
| | - Pablo Zoroquiain
- Pathology Department, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
15
|
Rüschoff J, Schildhaus HU, Rüschoff JH, Jöhrens K, Bocker Edmonston T, Dietmaier W, Bläker H, Baretton G, Horst D, Dietel M, Hartmann A, Klauschen F, Merkelbach-Bruse S, Stenzinger A, Schöniger S, Tiemann M, Weichert W, Büttner R. Testing for deficient mismatch repair and microsatellite instability : A focused update. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:61-70. [PMID: 37874379 PMCID: PMC10713762 DOI: 10.1007/s00292-023-01208-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 10/25/2023]
Abstract
Testing to detect mismatch repair deficiency (dMMR) and high-grade microsatellite instability (MSI-H) has become an integral part of the routine diagnostic workup for colorectal cancer (CRC). While MSI was initially considered to be a possible indicator of a hereditary disposition to cancer (Lynch syndrome, LS), today the prediction of the therapy response to immune checkpoint inhibitors (ICI) is in the foreground. Corresponding recommendations and testing algorithms are available for use in primary diagnosis (reviewed in: Rüschoff et al. 2021).Given the increasing importance for routine use and the expanding indication spectrum of ICI therapies for non-CRCs, such as endometrial, small intestinal, gastric, and biliary tract cancers, an updated review of dMMR/MSI testing is presented. The focus is on the challenges in the assessment of immunohistochemical stains and the value of PCR-based procedures, considering the expanded ICI indication spectrum. A practice-oriented flowchart for everyday diagnostic decision-making is provided that considers new data on the frequency and type of discordances between MMR-IHC and MSI-PCR findings, and the possible role of Next Generation Sequencing in clarifying them. Reference is made to the significance of systematic quality assurance measures (e.g., QuIP MSI portal and multicenter proficiency testing), including regular continued training and education.
Collapse
Affiliation(s)
- Josef Rüschoff
- Discovery Life Sciences Biomarker GmbH and North Hesse Pathology, Germaniastr. 7, 34119, Kassel, Germany.
| | - Hans-Ulrich Schildhaus
- Discovery Life Sciences Biomarker GmbH and North Hesse Pathology, Germaniastr. 7, 34119, Kassel, Germany
| | - Jan Hendrik Rüschoff
- Institute of Pathology and Molecular Pathology, Zürich University Hospital, Schmelzbergstrasse 12, 8091, Zürich, Switzerland
| | - Korinna Jöhrens
- Institute of Pathology, Carl Gustav Carus University Hospital, Fetscherstr. 74, 01307, Dresden, Germany
| | - Tina Bocker Edmonston
- Department of Pathology, Cooper University Health Care, 401 Haddon Ave, 08103, Camden, NJ, USA
| | - Wolfgang Dietmaier
- Institute of Pathology/Center for Molecular Pathology Diagnosis, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Hendrik Bläker
- Institute for Pathology, Leipzig University Hospital, Leipzig, Germany
| | - Gustavo Baretton
- Institute of Pathology, Carl Gustav Carus University Hospital, Fetscherstr. 74, 01307, Dresden, Germany
| | - David Horst
- Institute of Pathology, Charité University Hospital, Central Campus, Charitéplatz 1, 10117, Berlin, Germany
| | - Manfred Dietel
- Institute of Pathology, Charité University Hospital, Central Campus, Charitéplatz 1, 10117, Berlin, Germany
| | - Arndt Hartmann
- Pathological Institute, University of Erlangen-Nuremberg, Krankenhausstr. 8-10, 91054, Erlangen, Germany
| | - Frederick Klauschen
- Pathological Institute, Ludwig Maximilian University of Munich, Thalkirchner Str. 36, 80337, Munich, Germany
| | - Sabine Merkelbach-Bruse
- Institute of Pathology, Cologne University Hospital, Kerpener Str. 62, 50937, Cologne, Germany
| | - Albrecht Stenzinger
- Pathological Institute, Heidelberg University Hospital, Im Neuenheimer Feld 224, 69120, Heidelberg, Germany
| | - Sandra Schöniger
- Discovery Life Sciences Biomarker GmbH and North Hesse Pathology, Germaniastr. 7, 34119, Kassel, Germany
| | - Markus Tiemann
- Hamburg Institute of Hematopathology, Fangdieckstr. 75a, 22547, Hamburg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Trogerstr. 18, 81675, Munich, Germany
| | - Reinhard Büttner
- Institute of Pathology, Cologne University Hospital, Kerpener Str. 62, 50937, Cologne, Germany
| |
Collapse
|
16
|
André T, Berton D, Curigliano G, Sabatier R, Tinker AV, Oaknin A, Ellard S, de Braud F, Arkenau HT, Trigo J, Gravina A, Kristeleit R, Moreno V, Abdeddaim C, Vano YA, Samouëlian V, Miller R, Boni V, Torres AA, Gilbert L, Brown J, Dewal N, Dabrowski C, Antony G, Zografos E, Veneris J, Banerjee S. Antitumor Activity and Safety of Dostarlimab Monotherapy in Patients With Mismatch Repair Deficient Solid Tumors: A Nonrandomized Controlled Trial. JAMA Netw Open 2023; 6:e2341165. [PMID: 37917058 PMCID: PMC10623195 DOI: 10.1001/jamanetworkopen.2023.41165] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 08/14/2023] [Indexed: 11/03/2023] Open
Abstract
Importance Mismatch repair deficiency (dMMR) occurs in various cancers, and these tumors are attractive candidates for anti-programmed cell death 1 therapies, such as dostarlimab, a recently approved immune checkpoint inhibitor. Objective To assess the antitumor activity and safety of dostarlimab in patients with advanced or recurrent dMMR solid tumors. Design, Setting, And Participants The GARNET trial was a phase 1, open-label, single-group, multicenter study that began enrolling May 8, 2017. Participants had advanced or recurrent dMMR and microsatellite instability-high (MSI-H) or polymerase epsilon (POLE)-altered solid tumors. The data cut for this interim analysis was from November 1, 2021, with median follow-up of 27.7 months. Interventions Patients received 500 mg of dostarlimab intravenously every 3 weeks for 4 doses, then 1000 mg every 6 weeks until disease progression, discontinuation, or withdrawal. Main Outcomes and Measures The primary objective was to evaluate objective response rate and duration of response in patients with dMMR solid tumors by blinded independent central review using Response Evaluation Criteria in Solid Tumors, version 1.1. Results The efficacy population included 327 patients (median [range] age, 63 [24-85] years; 235 [71.9%] female; 7 [2.1%] Asian, 6 [1.8%] Black, and 206 [63.0%] White patients), with 141 patients (43.1%) with dMMR endometrial cancer, 105 patients (32.1%) with dMMR colorectal cancer, and 81 patients (24.8%) with other dMMR tumor types. All patients had at least 1 previous line of therapy. Objective response rate assessed per blinded independent central review for dMMR solid tumors was 44.0% (95% CI, 38.6% to 49.6%). Median duration of response was not reached (range, ≥1.18 to ≥47.21 months); 72.2% of responders (104 of 144) had a response lasting 12 or more months. Median progression-free survival was 6.9 months (95% CI, 4.2 to 13.6 months); probability of progression-free survival at 24 months was 40.6% (95% CI, 35.0% to 46.1%). Median overall survival was not reached (95% CI, 31.6 months to not reached). The most frequent immune-related adverse events were hypothyroidism (25 [6.9%]), alanine aminotransferase increase (21 [5.8%]), and arthralgia (17 [4.7%]). No new safety concerns were identified. Conclusions And Relevance In this nonrandomized controlled trial, dostarlimab was a well-tolerated treatment option with rapid, robust, and durable antitumor activity in patients with diverse dMMR solid tumors. These findings suggest that dostarlimab provides meaningful long-term benefit in a population with high unmet need. Trial Registration ClinicalTrials.gov Identifier: NCT02715284.
Collapse
Affiliation(s)
- Thierry André
- Saint-Antoine Hospital, INSERM, Unité Mixte de Recherche Scientifique 938, and SIRIC CURAMUS, Sorbonne University, Paris, France
| | - Dominique Berton
- GINECO and Institut de Cancerologie de l’Ouest, Centre René Gauducheau, Saint-Herblain, France
| | - Giuseppe Curigliano
- European Institute of Oncology, IRCCS, Milan, Italy
- Department of Medical Oncology and Hemato-Oncology, University of Milano, Milan, Italy
| | - Renaud Sabatier
- Department of Medical Oncology, Institut Paoli-Calmettes, CRCM, INSERM, CNRS, Aix-Marseille University, Marseille, France
| | - Anna V. Tinker
- BC Cancer–Vancouver, Vancouver, British Columbia, Canada
| | - Ana Oaknin
- Vall d’Hebron University Hospital and Vall d’Hebron Institute of Oncology, Barcelona, Spain
| | - Susan Ellard
- BC Cancer–Kelowna, Kelowna, British Columbia, Canada
| | - Filippo de Braud
- Ordinario di Oncologia Medica Direttore Scuola di Specialità in Oncologia Medica Università di Milano, Direttore Dipartimento Oncologia e Ematoncologia Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | | | - José Trigo
- Medical Oncology Department, Hospital Virgen de la Victoria IBIMA, Malaga, Spain
| | - Adriano Gravina
- Clinical Trials Unit, Instituto Nazionale Tumori, IRCCS, Fondazione “G. Pascale,” Naples, Italy
| | | | - Victor Moreno
- START Madrid FJD, Hospital Fundación Jiménez Díaz, Madrid, Spain
| | - Cyril Abdeddaim
- Centre de Lutte Contre le Cancer–Centre Oscar Lambret, Lille, France
| | - Yann-Alexandre Vano
- Department of Medical Oncology, Hôpital Européen Georges-Pompidou, Institut du Cancer Paris CARPEM, AP-HP Centre–Université Paris Cité, Paris, France
| | - Vanessa Samouëlian
- Gynecologic Oncology Division, Centre Hospitalier de l’Université de Montréal (CHUM), Centre de Recherche du CHUM, and Université de Montréal, Montreal, Quebec, Canada
| | - Rowan Miller
- University College London, St Bartholomew’s Hospital London, London, United Kingdom
| | - Valentina Boni
- NEXT Oncology Hospital Universitario Quirónsalud Madrid, Madrid, Spain
| | - Antonio Antón Torres
- Department of Medical Oncology, Hospital Universitario Miguel Servet and IIS Aragon, Zaragoza, Spain
| | - Lucy Gilbert
- Division of Gynecologic Oncology, McGill University Health Centre, Montreal, Quebec, Canada
| | - Jubilee Brown
- Division of Gynecologic Oncology, Levine Cancer Institute, Atrium Health, Charlotte, North Carolina
| | | | | | | | | | | | - Susana Banerjee
- The Royal Marsden NHS Foundation Trust and Institute of Cancer Research, London, United Kingdom
| |
Collapse
|
17
|
Chen J, Yu N, Ou S, Wang X, Li H, Zhu H. Integrated analysis reveals SMARCD1 is a potential biomarker and therapeutic target in skin cutaneous melanoma. J Cancer Res Clin Oncol 2023; 149:11619-11634. [PMID: 37401939 DOI: 10.1007/s00432-023-05064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 06/29/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVE SMARCD1 is a part of the SWI/SNF chromatin remodeling complex family, which consists of transcription factors that are implicated in various types of cancer. Examining SMARCD1 expression in human cancers can provide valuable insights into the development and progression of skin cutaneous melanoma (SKCM). METHODS Our study comprehensively examined the association between SMARCD1 expression and numerous factors, including prognosis, tumor microenvironment (TME), immune infiltration, tumor mutational burden (TMB), and microsatellite instability (MSI) in SKCM. Then we utilized immunohistochemical staining to measure the SMARCD1 expression in both SKCM tissues and normal skin tissues. Furthermore, we conducted in vitro experimentation to evaluate the effects of SMARCD1 knockdown on SKCM cells. RESULTS We found that aberrant expression of SMARCD1 across 16 cancers was strongly correlated with overall survival (OS) and progression-free survival (PFS). In addition, our research revealed that SMARCD1 expression is associated with multiple factors in different types of cancer, including immune infiltration, TME, immune-related genes, MSI, TMB, and sensitivity to anti-cancer drugs. SMARCD1 is likely involved in various SKCM signaling pathways and biological processes. Additionally, our research revealed that an SMARCD1-based risk factor model accurately predicted OS in SKCM patients. Furthermore, the downregulation of SMARCD1 expression demonstrated a significant inhibition of SKCM cell proliferation and migration, as well as an increase in apoptosis and cell cycle arrest. CONCLUSION We conclude that SMARCD1 is a promising diagnostic, prognostic, and therapeutic biomarker for SKCM, and its expression has significant clinical implications for the development of novel treatment strategies.
Collapse
Affiliation(s)
- Jiaoquan Chen
- Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, 510095, Guangdong, China
| | - Nanji Yu
- Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, 510095, Guangdong, China
| | - Shanshan Ou
- Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, 510095, Guangdong, China
| | - Xue Wang
- Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, 510095, Guangdong, China
| | - Huaping Li
- Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, 510095, Guangdong, China
| | - Huilan Zhu
- Department of Dermatology, Guangzhou Institute of Dermatology, Guangzhou, 510095, Guangdong, China.
| |
Collapse
|
18
|
Simbolo M, Silvestris N, Malleo G, Mafficini A, Maggino L, Cocomazzi A, Veghini L, Mombello A, Pezzini F, Sereni E, Martelli FM, Gkountakos A, Ciaparrone C, Piredda ML, Ingravallo G, Paolino G, Nappo F, Rapposelli IG, Frassinetti L, Saragoni L, Lonardi S, Pea A, Paiella S, Fassan M, Brunetti O, Cingarlini S, Salvia R, Milella M, Corbo V, Lawlor RT, Scarpa A, Luchini C. Clinical and Genomic Characterization of Pancreatic Ductal Adenocarcinoma with Signet-Ring/Poorly Cohesive Cells. Mod Pathol 2023; 36:100251. [PMID: 37355152 DOI: 10.1016/j.modpat.2023.100251] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Revised: 05/30/2023] [Accepted: 06/15/2023] [Indexed: 06/26/2023]
Abstract
Signet-ring cell (SRC)/poorly cohesive cell carcinoma is an aggressive variant of pancreatic ductal adenocarcinoma (PDAC). This study aimed to clarify its clinicopathologic and molecular profiles based on a multi-institutional cohort of 20 cases. The molecular profiles were investigated using DNA and RNA sequencing. The clinicopathologic parameters and molecular alterations were analyzed based on survival indices and using a validation/comparative cohort of 480 conventional PDAC patients. The primary findings were as follows: (1) clinicopathologic features: SRC carcinomas are highly aggressive neoplasms with poor prognosis, and the lungs are elective metastatic sites; (2) survival analysis: a higher SRC component was indicative of poorer prognosis. In particular, the most clinically significant threshold of SRC was 80%, showing statistically significant differences in both disease-specific and disease-free survival; (3) genomic profiles: SRC carcinomas are similar to conventional PDAC with the most common alterations affecting the classic PDAC drivers KRAS (70% of cases), TP53 (55%), SMAD4 (25%), and CDKN2A (20%). EGFR alterations, RET::CCDC6 fusion gene, and microsatellite instability (3 different cases, 1 alteration per case) represent novel targets for precision oncology. The occurrence of SMAD4 mutations was associated with poorer prognosis; (4) pancreatic SRC carcinomas are genetically different from gastric SRC carcinomas: CDH1, the classic driver gene of gastric SRC carcinoma, is not altered in pancreatic SRC carcinoma; (5) transcriptome analysis: the cases clustered into 2 groups, one classical/exocrine-like, and the other squamous-like; and (6) SRC carcinoma-derived organoids can be successfully generated, and their cultures preserve the histologic and molecular features of parental SRC carcinoma. Although pancreatic SRC carcinoma shares similarities with conventional PDAC regarding the most important genetic drivers, it also exhibits important differences. A personalized approach for patients with this tumor type should consider the clinical relevance of histologic determination of the SRC component and the presence of potentially actionable molecular targets.
Collapse
Affiliation(s)
- Michele Simbolo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology "G. Barresi", University of Messina, Messina, Italy
| | - Giuseppe Malleo
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
| | - Andrea Mafficini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Laura Maggino
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
| | | | - Lisa Veghini
- 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
| | - Francesco Pezzini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Elisabetta Sereni
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
| | - Filippo M Martelli
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | | | - Chiara Ciaparrone
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Maria L Piredda
- ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Giuseppe Ingravallo
- Department of Emergency and Transplantation, Pathology Section, University of Bari Medical School, Bari, Italy
| | - Gaetano Paolino
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Floriana Nappo
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | | | - Luca Saragoni
- Department of Pathological Anatomy, AUSL Romagna, Morgagni-Pierantoni Hospital, Forlì, Italy
| | - Sara Lonardi
- Medical Oncology 1, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Antonio Pea
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
| | - Salvatore Paiella
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
| | - Matteo Fassan
- Surgical Pathology Unit, Department of Medicine (DIMED), University of Padua, and Veneto Institute of Oncology, IOV-IRCCS, Padua, Italy
| | - Oronzo Brunetti
- IRCCS Istituto Tumori "Giovanni Paolo II" of Bari, Vari, Italy
| | - Sara Cingarlini
- Department of Medicine, Section of Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - Roberto Salvia
- Department of Surgery, The Pancreas Institute, University and Hospital Trust of Verona, Verona, Italy
| | - Michele Milella
- Department of Medicine, Section of Oncology, University and Hospital Trust of Verona, Verona, Italy
| | - Vincenzo Corbo
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy
| | - Rita T Lawlor
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Aldo Scarpa
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy
| | - Claudio Luchini
- Department of Diagnostics and Public Health, Section of Pathology, University and Hospital Trust of Verona, Verona, Italy; ARC-Net Research Center, University and Hospital Trust of Verona, Verona, Italy.
| |
Collapse
|
19
|
Rüschoff J, Schildhaus HU, Rüschoff JH, Jöhrens K, Bocker-Edmonston T, Dietmaier W, Bläker H, Baretton G, Horst D, Dietel M, Hartmann A, Klauschen F, Merkelbach-Bruse S, Stenzinger A, Schöniger S, Tiemann M, Weichert W, Büttner R. [Testing deficient mismatch repair and microsatellite instability : A focused update. German version]. PATHOLOGIE (HEIDELBERG, GERMANY) 2023; 44:301-310. [PMID: 37548948 PMCID: PMC10457237 DOI: 10.1007/s00292-023-01209-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/18/2023] [Indexed: 08/08/2023]
Abstract
Testing to detect mismatch repair deficiency (dMMR) and high-grade microsatellite instability (MSI-H) has become an integral part of the routine diagnostic workup for colorectal cancer (CRC). While MSI was initially considered to be a possible indicator of a hereditary disposition to cancer (Lynch syndrome, LS), today the prediction of the therapy response to immune checkpoint inhibitors (ICI) is in the foreground. Corresponding recommendations and testing algorithms are available for use in primary diagnosis (reviewed in: Rüschoff et al. 2021).Given the increasing importance for routine use and the expanding indication spectrum of ICI therapies for non-CRCs, such as endometrial, small intestinal, gastric, and biliary tract cancers, an updated review of dMMR/MSI testing is presented. The focus is on the challenges in the assessment of immunohistochemical stains and the value of PCR-based procedures, considering the expanded ICI indication spectrum. A practice-oriented flowchart for everyday diagnostic decision-making is provided that considers new data on the frequency and type of discordances between MMR-IHC and MSI-PCR findings, and the possible role of Next Generation Sequencing in clarifying them. Reference is made to the significance of systematic quality assurance measures (e.g., QuIP MSI portal and multicenter proficiency testing), including regular continued training and education.
Collapse
Affiliation(s)
- Josef Rüschoff
- Discovery Life Sciences Biomarker GmbH und Pathologie Nordhessen, Germaniastr. 7, 34119, Kassel, Deutschland.
| | - Hans-Ulrich Schildhaus
- Discovery Life Sciences Biomarker GmbH und Pathologie Nordhessen, Germaniastr. 7, 34119, Kassel, Deutschland
| | - Jan Hendrik Rüschoff
- Institut für Pathologie und Molekularpathologie, Universitätsspital Zürich, Zürich, Schweiz
| | - Korinna Jöhrens
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus Dresden, Dresden, Deutschland
| | | | - Wolfgang Dietmaier
- Institut für Pathologie/Zentrum für molekularpathologische Diagnostik, Universität Regensburg, Regensburg, Deutschland
| | - Hendrik Bläker
- Institut für Pathologie, Universitätsklinikum Leipzig, Leipzig, Deutschland
| | - Gustavo Baretton
- Institut für Pathologie, Universitätsklinikum Carl Gustav Carus Dresden, Dresden, Deutschland
| | - David Horst
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Deutschland
| | - Manfred Dietel
- Institut für Pathologie, Charité - Universitätsmedizin Berlin, Campus Mitte, Berlin, Deutschland
| | - Arndt Hartmann
- Pathologisches Institut, Universität Erlangen-Nürnberg, Erlangen, Deutschland
| | - Frederick Klauschen
- Pathologisches Institut, Ludwig-Maximilians-Universität München, München, Deutschland
| | | | - Albrecht Stenzinger
- Pathologisches Institut, Universitätsklinikum Heidelberg, Heidelberg, Deutschland
| | - Sandra Schöniger
- Discovery Life Sciences Biomarker GmbH und Pathologie Nordhessen, Germaniastr. 7, 34119, Kassel, Deutschland
| | - Markus Tiemann
- Institut für Hämatopathologie Hamburg, Hamburg, Deutschland
| | - Wilko Weichert
- Institut für Pathologie, Technische Universität München, München, Deutschland
| | - Reinhard Büttner
- Institut für Pathologie, Universitätsklinikum Köln, Köln, Deutschland
| |
Collapse
|
20
|
Mishima S, Naito Y, Akagi K, Hayashi N, Hirasawa A, Hishiki T, Igarashi A, Ikeda M, Kadowaki S, Kajiyama H, Kato M, Kenmotsu H, Kodera Y, Komine K, Koyama T, Maeda O, Miyachi M, Nishihara H, Nishiyama H, Ohga S, Okamoto W, Oki E, Ono S, Sanada M, Sekine I, Takano T, Tao K, Terashima K, Tsuchihara K, Yatabe Y, Yoshino T, Baba E. Japanese Society of Medical Oncology/Japan Society of Clinical Oncology/Japanese Society of Pediatric Hematology/Oncology-led clinical recommendations on the diagnosis and use of immunotherapy in patients with high tumor mutational burden tumors. Int J Clin Oncol 2023; 28:941-955. [PMID: 37300720 PMCID: PMC10390617 DOI: 10.1007/s10147-023-02360-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 05/18/2023] [Indexed: 06/12/2023]
Abstract
The development of novel antitumor agents and accompanying biomarkers has improved survival across several tumor types. Previously, we developed recommendations for tumor-agnostic treatments in patients with solid tumors with DNA mismatch repair deficient or neurotrophic receptor tyrosine kinase fusions. Recently, immune checkpoint inhibitors have shown efficacy in patient with tumor mutation burden-high (TMB-H) solid tumors and have been established as a third tumor-agnostic agent, making it necessary to develop the guideline prioritized for these patients. Clinical questions regarding medical care were formulated for patients with TMB-H advanced solid tumors. Relevant publications were searched by PubMed and Cochrane Database. Critical publications and conference reports were added manually. Systematic reviews were performed for each clinical question for the purpose of developing clinical recommendations. The committee members identified by Japan Society of Clinical Oncology (JSCO), Japanese Society of Medical Oncology (JSMO), and Japanese society of pediatric hematology/oncology (JSPHO) voted to determine the level of each recommendation considering the strength of evidence, expected risks and benefits to patients, and other related factors. Thereafter, a peer review by experts nominated from JSCO, JSMO, and JSPHO, and the public comments among all societies' members was done. The current guideline describes three clinical questions and seven recommendations for whom, when, and how TMB should be tested, and what is recommended for patients with TMB-H advanced solid tumors. In this guideline, the committee proposed seven recommendations for performing TMB testing properly to select patients who are likely to benefit from immunotherapy.
Collapse
Affiliation(s)
- Saori Mishima
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Yoichi Naito
- National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Naomi Hayashi
- The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | | | - Ataru Igarashi
- Yokohama City University School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eiji Oki
- Kyushu University, Fukuoka, Japan
| | | | - Masashi Sanada
- National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | | | | | - Kayoko Tao
- National Cancer Center Hospital, Tokyo, Japan
| | - Keita Terashima
- National Center for Child Health and Development, Tokyo, Japan
| | | | | | | | | |
Collapse
|
21
|
Ukkola I, Nummela P, Heiskanen A, Holm M, Zafar S, Kero M, Haglund C, Satomaa T, Kytölä S, Ristimäki A. N-Glycomic Profiling of Microsatellite Unstable Colorectal Cancer. Cancers (Basel) 2023; 15:3571. [PMID: 37509233 PMCID: PMC10376987 DOI: 10.3390/cancers15143571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/06/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023] Open
Abstract
Aberrant glycosylation affects cancer progression and immune evasion. Approximately 15% of colorectal cancers (CRCs) demonstrate microsatellite instability (MSI) and display major differences in outcomes and therapeutic responses, as compared to corresponding microsatellite stable (MSS) tumors. We compared the N-glycan profiles of stage II and IV MSI CRC tumors, further subdivided into BRAFV600E wild-type and mutated subgroups (n = 10 in each subgroup), with each other and with those of paired non-neoplastic mucosal samples using mass spectrometry. Further, the N-glycans of BRAFV600E wild-type stage II MSI tumors were compared to corresponding MSS tumors (n = 9). Multiple differences in N-glycan profiles were identified between the MSI CRCs and control tissues, as well as between the stage II MSI and MSS samples. The MSI CRC tumors showed a lower relative abundance of high-mannose N-glycans than did the control tissues or the MSS CRCs. Among MSI CRC subgroups, acidic N-glycans showed tumor stage and BRAF mutation status-dependent variation. Specifically, the large, sulfated/phosphorylated, and putative terminal N-acetylhexosamine-containing acidic N-glycans differed between the MSI CRC subgroups, showing opposite changes in stages II and IV, when comparing BRAF mutated and wild-type tumors. Our results show that molecular subgroups of CRC exhibit characteristic glycan profiles that may explain certain carcinogenic properties of MSI tumors.
Collapse
Affiliation(s)
- Iiris Ukkola
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| | - Pirjo Nummela
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| | | | - Matilda Holm
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Department of Surgery, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Sadia Zafar
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| | - Mia Kero
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Caj Haglund
- Translational Cancer Medicine Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
- Department of Surgery, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Tero Satomaa
- Glykos Finland Co., Ltd., 00790 Helsinki, Finland
| | - Soili Kytölä
- HUSLAB, Department of Genetics, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
| | - Ari Ristimäki
- HUSLAB, Department of Pathology, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, 00029 Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Helsinki University Hospital and University of Helsinki, 00014 Helsinki, Finland
| |
Collapse
|
22
|
Matsubara J, Mukai K, Kondo T, Yoshioka M, Kage H, Oda K, Kudo R, Ikeda S, Ebi H, Muro K, Hayashi R, Tokudome N, Yamamoto N, Muto M. First-Line Genomic Profiling in Previously Untreated Advanced Solid Tumors for Identification of Targeted Therapy Opportunities. JAMA Netw Open 2023; 6:e2323336. [PMID: 37459099 DOI: 10.1001/jamanetworkopen.2023.23336] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
IMPORTANCE Precision oncology using comprehensive genomic profiling (CGP) by next-generation sequencing is aimed at companion diagnosis and genomic profiling. The clinical utility of CGP before the standard of care (SOC) is still not resolved, and more evidence is needed. OBJECTIVE To investigate the clinical utility of next-generation CGP (FoundationOne CDx [F1CDx]) in patients with previously untreated metastatic or recurrent solid tumors. DESIGN, Setting, and Participants This multicenter, prospective, observational cohort study enrolled patients with previously untreated advanced solid tumors between May 18, 2021, and February 16, 2022, with follow-up through August 16, 2022. The study was conducted at 6 hospitals in Japan. Eligible patients were aged 20 years or older and had Eastern Cooperative Oncology Group performance status of 0 to 1 with previously untreated metastatic or recurrent cancers in the gastrointestinal or biliary tract; pancreas, lung, breast, uterus, or ovary; and malignant melanoma. EXPOSURE Comprehensive genomic profiling testing before SOC for advanced solid tumors. MAIN OUTCOMES AND MEASURES Proportion of patients with actionable or druggable genomic alterations and molecular-based recommended therapy (MBRT). RESULTS A total of 183 patients met the inclusion criteria and 180 patients (92 men [51.1%]) with a median age of 64 years (range, 23-88 years) subsequently underwent CGP (lung [n = 28], colon/small intestine [n = 27], pancreas [n = 27], breast [n = 25], biliary tract [n = 20], gastric [n = 19], uterus [n = 12], esophagus [n = 10], ovary [n = 6], and skin melanoma [n = 6]). Data from 172 patients were available for end point analyses. Actionable alterations were found in 172 patients (100.0%; 95% CI, 97.9%-100.0%) and druggable alternations were identified in 109 patients (63.4%; 95% CI, 55.7%-70.6%). The molecular tumor board identified MBRT for 105 patients (61.0%; 95% CI, 53.3%-68.4%). Genomic alterations included in the companion diagnostics list of the CGP test were found in 49 patients (28.5%; 95% CI, 21.9%-35.9%) in a tumor-agnostic setting. After a median follow-up of 7.9 months (range, 0.5-13.2 months), 34 patients (19.8%; 95% CI, 14.1%-26.5%) received MBRT. CONCLUSIONS AND RELEVANCE The findings of this study suggest that CGP testing before SOC for patients with advanced solid tumors may be clinically beneficial to guide the subsequent anticancer therapies, including molecularly matched treatments.
Collapse
Affiliation(s)
- Junichi Matsubara
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Kumi Mukai
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Masahiro Yoshioka
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Hidenori Kage
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Katsutoshi Oda
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Ryo Kudo
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Ryuji Hayashi
- Department of Clinical Oncology, Toyama University Hospital, Toyama, Japan
| | - Nahomi Tokudome
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | | | - Manabu Muto
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
23
|
Yang AT, Laetsch TW. Safety of current treatment options for NTRK fusion-positive cancers. Expert Opin Drug Saf 2023; 22:1073-1089. [PMID: 37869783 PMCID: PMC10842066 DOI: 10.1080/14740338.2023.2274426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/19/2023] [Indexed: 10/24/2023]
Abstract
INTRODUCTION Oncogenic NTRK fusions have been found in multiple cancer types affecting adults and/or children, including rare tumors with pathognomonic fusions and common cancers in which fusions are rare. The tropomyosin receptor kinase inhibitors (TRKi) larotrectinib and entrectinib are among the first agents with tissue agnostic FDA approvals for cancer treatment, and additional TRKi are undergoing development. As experience with TRKi grow, novel mechanisms of resistance and on/off target side effects have become increasingly important considerations. AREAS COVERED Authors reviewed literature published through July 2023 on platforms such as PubMed, clinicaltrials.gov, and manufacturer/FDA drug labels, focusing on the development of TRKi, native functions of TRK, phenotype of congenital TRK aberrancies, efficacy, and safety profile of TRKi in clinical trials and investigator reports, and on/off target adverse effects associated with TRKi (Appendix A). EXPERT OPINION TRKi have histology-agnostic activity against tumors with NTRK gene fusions. TRKi are generally well tolerated with a side effect profile that compares favorably to cytotoxic chemotherapy. There are numerous ongoing studies investigating TRKi as frontline, adjuvant, and salvage therapy. It will be critical to continue to gather long-term safety data on the use of these agents, particularly in children.
Collapse
Affiliation(s)
- Adeline T. Yang
- Division of Oncology, Children’s Hospital of Philadelphia, and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| | - Theodore Willis Laetsch
- Division of Oncology, Children’s Hospital of Philadelphia, and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
24
|
Schraa SJ, Stelloo E, Laclé MM, Swennenhuis JF, Brosens LAA, Fijneman RJA, Feitsma H, Koopman M, de Leng WW, Vink GR, Bol GM. Comparison of NTRK fusion detection methods in microsatellite-instability-high metastatic colorectal cancer. Virchows Arch 2023; 482:983-992. [PMID: 37067589 PMCID: PMC10247849 DOI: 10.1007/s00428-023-03538-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/15/2023] [Accepted: 03/27/2023] [Indexed: 04/18/2023]
Abstract
Tropomyosin receptor kinase (TRK) inhibitors have been approved for metastatic solid tumors harboring NTRK fusions, but the detection of NTRK fusions is challenging. International guidelines recommend pan-TRK immunohistochemistry (IHC) screening followed by next generation sequencing (NGS) in tumor types with low prevalence of NTRK fusions, including metastatic colorectal cancer (mCRC). RNA-based NGS is preferred, but is expensive, time-consuming, and extracting good-quality RNA from FFPE tissue is challenging. Alternatives in daily clinical practice are warranted. We assessed the diagnostic performance of RNA-NGS, FFPE-targeted locus capture (FFPE-TLC), fluorescence in situ hybridization (FISH), and the 5'/3' imbalance quantitative RT-PCR (qRT-PCR) after IHC screening in 268 patients with microsatellite-instability-high mCRC, the subgroup in which NTRK fusions are most prevalent (1-5%). A consensus result was determined after review of all assay results. In 16 IHC positive tumors, 10 NTRK fusions were detected. In 33 IHC negative samples, no additional transcribed NTRK fusions were found, underscoring the high sensitivity of IHC. Sensitivity of RNA-NGS, FFPE-TLC, FISH, and qRT-PCR was 90%, 90%, 78%, and 100%, respectively. Specificity was 100% for all assays. Robustness, defined as the percentage of samples that provided an interpretable result in the first run, was 100% for FFPE-TLC, yet more limited for RNA-NGS (85%), FISH (70%), and qRT-PCR (70%). Overall, we do not recommend FISH for the detection of NTRK fusions in mCRC due to its low sensitivity and limited robustness. We conclude that RNA-NGS, FFPE-TLC, and qRT-PCR are appropriate assays for NTRK fusion detection, after enrichment with pan-TRK IHC, in routine clinical practice.
Collapse
Affiliation(s)
- Suzanna J Schraa
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | - Miangela M Laclé
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | | | - Lodewijk A A Brosens
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Remond J A Fijneman
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, Netherlands
| | | | - Miriam Koopman
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Wendy W de Leng
- Department of Pathology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
| | - Geraldine R Vink
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands
- Department of Research and Development, Netherlands Comprehensive Cancer Organisation (IKNL), Utrecht, Netherlands
| | - Guus M Bol
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht University, Utrecht, Netherlands.
| |
Collapse
|
25
|
Crescenzi A, Baloch Z. Immunohistochemistry in the pathologic diagnosis and management of thyroid neoplasms. Front Endocrinol (Lausanne) 2023; 14:1198099. [PMID: 37324272 PMCID: PMC10266214 DOI: 10.3389/fendo.2023.1198099] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
The use of immunohistochemistry cannot be underestimated in the everyday practice of thyroid pathology. It has evolved over the years beyond the traditional confirmation of thyroid origin to molecular profiling and the prediction of clinical behavior. In addition, immunohistochemistry has served to implement changes in the current thyroid tumor classification scheme. It is prudent to perform a panel of immunostains, and the immunoprofile should be interpreted in light of the cytologic and architectural features. Immunohistochemistry can also be easily performed in the limited cellularity specimen preparation generated from thyroid fine-needle aspiration and core biopsy; however, it will require laboratory validation of immunostains specific to these preparations to avoid diagnostic pitfalls. This review discusses the application of immunohistochemistry in thyroid pathology with a focus on limited cellularity preparations.
Collapse
Affiliation(s)
- Anna Crescenzi
- Pathology, University Campus Bio-Medico of Rome, Fondazione Policlinico, Rome, Italy
| | - Zubair Baloch
- Pathology & Laboratory Medicine, University of Pennsylvania Medical Center, Perelman School of Medicine, Philadelphia, PA, United States
| |
Collapse
|
26
|
Naito Y, Mishima S, Akagi K, Hayashi N, Hirasawa A, Hishiki T, Igarashi A, Ikeda M, Kadowaki S, Kajiyama H, Kato M, Kenmotsu H, Kodera Y, Komine K, Koyama T, Maeda O, Miyachi M, Nishihara H, Nishiyama H, Ohga S, Okamoto W, Oki E, Ono S, Sanada M, Sekine I, Takano T, Tao K, Terashima K, Tsuchihara K, Yatabe Y, Yoshino T, Baba E. Japanese Society of Medical Oncology/Japan Society of Clinical Oncology/Japanese Society of Pediatric Hematology/Oncology-led clinical recommendations on the diagnosis and use of tropomyosin receptor kinase inhibitors in adult and pediatric patients with neurotrophic receptor tyrosine kinase fusion-positive advanced solid tumors. Int J Clin Oncol 2023:10.1007/s10147-023-02345-7. [PMID: 37212982 DOI: 10.1007/s10147-023-02345-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Accepted: 04/13/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Clinical trials have reported the efficacy of tropomyosin receptor kinase (TRK) inhibitors against neurotrophic receptor tyrosine kinase (NTRK) fusion gene-positive advanced solid tumors. The accumulated evidence of tumor-agnostic agent has made since TRK inhibitors were approved and used in clinical practice. Therefore, we have revised the 'Japan Society of Clinical Oncology (JSCO)/Japanese Society of Medical Oncology (JSMO)-led clinical recommendations on the diagnosis and use of tropomyosin receptor kinase inhibitors in adult and pediatric patients with neurotrophic receptor tyrosine kinase fusion-positive advanced solid tumors, cooperated by the Japanese Society of Pediatric Hematology/Oncology (JSPHO)'. METHODS Clinical questions regarding medical care were formulated for patients with NTRK fusion-positive advanced solid tumors. Relevant publications were searched by PubMed and Cochrane Database. Critical publications and conference reports were added manually. Systematic reviews were performed for each clinical question for the purpose of developing clinical recommendations. The committee members identified by JSCO, JSMO, and JSPHO voted to determine the level of each recommendation considering the strength of evidence, expected risks and benefits to patients, and other related factors. Thereafter, a peer review by experts nominated from JSCO, JSMO, and JSPHO, and the public comments among all societies' members was done. RESULTS The current guideline describes 3 clinical questions and 14 recommendations for whom, when, and how NTRK fusion should be tested, and what is recommended for patients with NTRK fusion-positive advanced solid tumors. CONCLUSION The committee proposed 14 recommendations for performing NTRK testing properly to select patients who are likely to benefit from TRK inhibitors.
Collapse
Affiliation(s)
- Yoichi Naito
- National Cancer Center Hospital East, Kashiwa, Japan
| | - Saori Mishima
- National Cancer Center Hospital East, Kashiwa, Japan
| | | | - Naomi Hayashi
- The Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | | | | | - Ataru Igarashi
- Yokohama City University School of Medicine, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Eiji Oki
- Kyushu University, Fukuoka, Japan
| | | | - Masashi Sanada
- National Hospital Organization Nagoya Medical Center, Aichi, Japan
| | | | | | - Kayoko Tao
- National Cancer Center Hospital, Tokyo, Japan
| | - Keita Terashima
- National Center for Child Health and Development, Tokyo, Japan
| | | | | | | | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
27
|
Ukkola I, Nummela P, Kero M, Ristimäki A. Diagnostic performance of Idylla MSI test in colorectal cancer biopsies. Diagn Pathol 2023; 18:39. [PMID: 36978094 PMCID: PMC10053848 DOI: 10.1186/s13000-023-01328-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 03/17/2023] [Indexed: 03/30/2023] Open
Abstract
Universal testing for microsatellite instability (MSI) is recommended in colorectal cancer (CRC) to screen for Lynch syndrome and to guide optimal treatment and follow-up of the patients. Especially in neoadjuvant setting, where immuno-oncological treatments have recently shown excellent responses, identification of MSI status at biopsy is a prerequisite. Idylla MSI test offers a rapid and automated test to assess MSI-status from formalin-fixed paraffin-embedded tumor tissue sections. In this study, we compared the performance of the Idylla MSI test to mismatch repair (MMR) protein immunohistochemistry (IHC) using 117 CRC biopsies with previously known deficient MMR status. The concordance between Idylla and IHC was 99.0% (95/96) for biopsies with the recommended ≥ 20% tumor cell content. Further, 85.7% (18/21) of suboptimal CRC biopsy specimens (tumor cell content 5-15%) were diagnosed as MSI. Overall, we identified four discrepant cases of which three had tumor cell content less than 20%, explaining the discordant result. Our study shows that the Idylla MSI test offers a competent tool for MSI screening in CRC biopsy specimens.
Collapse
Affiliation(s)
- Iiris Ukkola
- Department of Pathology, HUS Diagnostic Center, HUSLAB, Helsinki University Hospital, University of Helsinki, P.O. Box 400, FI-00029 HUS, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Pirjo Nummela
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki University Hospital, Helsinki, Finland
| | - Mia Kero
- Department of Pathology, HUS Diagnostic Center, HUSLAB, Helsinki University Hospital, University of Helsinki, P.O. Box 400, FI-00029 HUS, Helsinki, Finland
| | - Ari Ristimäki
- Department of Pathology, HUS Diagnostic Center, HUSLAB, Helsinki University Hospital, University of Helsinki, P.O. Box 400, FI-00029 HUS, Helsinki, Finland.
- Applied Tumor Genomics Research Program, Research Programs Unit, University of Helsinki, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
28
|
O'Haire S, Franchini F, Kang YJ, Steinberg J, Canfell K, Desai J, Fox S, IJzerman M. Systematic review of NTRK 1/2/3 fusion prevalence pan-cancer and across solid tumours. Sci Rep 2023; 13:4116. [PMID: 36914665 PMCID: PMC10011574 DOI: 10.1038/s41598-023-31055-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/06/2023] [Indexed: 03/16/2023] Open
Abstract
NTRK gene fusions are rare somatic mutations found across cancer types with promising targeted therapies emerging. Healthcare systems face significant challenges in integrating these treatments, with uncertainty in prevalence and optimal testing methods to identify eligible patients. We performed a systematic review of NTRK fusion prevalence to inform efficient diagnostic screening and scale of therapeutic uptake. We searched Medline, Embase and Cochrane databases on 31/03/2021. Inclusion criteria were studies reporting fusion rates in solid tumours, English language, post-2010 publication and minimum sample size. Critical appraisal was performed using a custom 11-item checklist. Rates were collated by cancer type and pooled if additional synthesis criteria were met. 160 studies were included, with estimates for 15 pan-cancer and 429 specific cancer types (63 paediatric). Adult pan-cancer estimates ranged 0.03-0.70%, with higher rates found in RNA-based assays. In common cancers, rates were consistently below 0.5%. Rare morphological subtypes, colorectal microsatellite instability, and driver mutation exclusion cancers had higher rates. Only 35.6% of extracted estimates used appropriate methods and sample size to identify NTRK fusions. NTRK fusion-positive cancers are rare and widely distributed across solid tumours. Small-scale, heterogeneous data confound prevalence prediction. Further large-scale, standardised genomic data are needed to characterise NTRK fusion epidemiology.
Collapse
Affiliation(s)
- Sophie O'Haire
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia.
| | - Fanny Franchini
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
| | - Yoon-Jung Kang
- The Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council New South Wales, Sydney, Australia
| | - Julia Steinberg
- The Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council New South Wales, Sydney, Australia
| | - Karen Canfell
- The Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council New South Wales, Sydney, Australia
| | - Jayesh Desai
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Stephen Fox
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Melbourne, Australia
| | - Maarten IJzerman
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, Australia
- Erasmus School of Health Policy and Management, Rotterdam, The Netherlands
| |
Collapse
|
29
|
Medford AJ, Oshry L, Boyraz B, Kiedrowski L, Menshikova S, Butusova A, Dai CS, Gogakos T, Keenan JC, Occhiogrosso RH, Ryan P, Lennerz JK, Spring LM, Moy B, Ellisen LW, Bardia A. TRK inhibitor in a patient with metastatic triple-negative breast cancer and NTRK fusions identified via cell-free DNA analysis. Ther Adv Med Oncol 2023; 15:17588359231152844. [PMID: 36743521 PMCID: PMC9893401 DOI: 10.1177/17588359231152844] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 01/05/2023] [Indexed: 02/04/2023] Open
Abstract
Tissue-agnostic indications for targeted therapies have expanded options for patients with advanced solid tumors. The Food and Drug Administration approvals of the programmed death-ligand 1 inhibitor pembrolizumab and the TRK inhibitors larotrectinib and entrectinib provide rationale for next-generation sequencing (NGS) in effectively all advanced solid tumor patients given potential for clinical responses even in otherwise refractory disease. As proof of concept, this case report describes a 64-year-old woman with triple-negative breast cancer refractory to multiple lines of therapy, found to have a rare mutation on NGS which led to targeted therapy with meaningful response. She initially presented with metastatic recurrence 5 years after treatment for a localized breast cancer, with rapid progression through four lines of therapy in the metastatic setting, including immunotherapy, antibody-drug conjugate-based therapy, and chemotherapy. Germline genetic testing was normal. Ultimately, NGS evaluation of cell-free DNA via an 83-gene assay (Guardant Health, Inc.) identified two NTRK3 fusions: an ETV6-NTRK3 fusion associated with the rare secretory breast carcinoma, and CRTC3-NTRK3, a novel fusion partner not previously described in breast cancer. Liver biopsy was sent for whole exome sequencing and RNA-seq analysis of tissue (BostonGene, Inc., Boston, MA, USA), which provided orthogonal confirmation of both the ETV6-NTRK3 and CRTC3-NTRK3 fusions. She was started on the TRK inhibitor larotrectinib with a marked clinical and radiographic response after only 2 months of therapy. The patient granted verbal consent to share her clinical story, images, and data in this case report. This case demonstrates the significant potential benefits of NGS testing in advanced cancer and the lessons we may learn from individual patient experiences.
Collapse
Affiliation(s)
| | - Lauren Oshry
- Boston Medical Center, Boston, MA, USA,Boston University School of Medicine, Boston, MA, USA
| | - Baris Boyraz
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | | | | | | | - Charles S. Dai
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Dana Farber Cancer Institute, Boston, MA, USA
| | - Tasos Gogakos
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | | | - Rachel H. Occhiogrosso
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Dana Farber Cancer Institute, Boston, MA, USA
| | - Phoebe Ryan
- Massachusetts General Hospital Cancer Center, Boston, MA, USA
| | - Jochen K. Lennerz
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Laura M. Spring
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Beverly Moy
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Leif W. Ellisen
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| | - Aditya Bardia
- Massachusetts General Hospital Cancer Center, Boston, MA, USA,Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Zhao H, Gao J, Bai B, Wang R, Yu J, Lu H, Cheng M, Liang P. Development and external validation of a non-invasive imaging biomarker to estimate the microsatellite instability status of gastric cancer and its prognostic value: The combination of clinical and quantitative CT-imaging features. Eur J Radiol 2023; 162:110719. [PMID: 36764010 DOI: 10.1016/j.ejrad.2023.110719] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/08/2023] [Accepted: 01/25/2023] [Indexed: 01/28/2023]
Abstract
PURPOSE Molecular testing for microsatellite instability (MSI) status plays a vital role in the clinical management of gastric cancer (GC). Nevertheless, challenges of routinely applied technology for MSI determination exist. This study aimed to develop and validate a non-invasive imaging biomarker for MSI assessment in GC and explore its prognostic value. METHODS We retrospectively recruited 396 GC patients with pretreatment CT images from a single center and a public database and divided them into an original cohort (n = 356) and an external validation cohort (n = 40). The SMOTE algorithm was used to generate a balanced training cohort (n = 192) and the independent radiomics model, clinical model, and radiomics-clinic combined model were constructed for determining MSI status. The models' discrimination, calibration, clinical usefulness, and prognosis significance were evaluated by AUC, calibration, decision curve analyses, and Kaplan-Meier curve analysis, respectively. RESULTS The radiomics-clinic combined model derived from clinical and quantitative CT-based "Radscore" exhibited the best discriminatory abilities of MSI status in all cohorts, with AUCs of 0.836 (95% CI, 0.780-0.893) in the training cohort, 0.834 (95% CI, 0.688-0.981) in the external validation cohort, and 0.750 (95% CI, 0.682-0.819) in the original cohort, respectively. Meanwhile, the combined model demonstrated goodness of fitness, higher clinical net benefits, and significant positive integrated discrimination improvement compared with any independent model. While it showed no significant overall survival- or progression-free survival-based risk stratification ability (p > 0.05). CONCLUSIONS The radiomics-clinic combined model could be a potential non-invasive biomarker for MSI status in GC, which help clinical decision-making, nevertheless, provided limited prognostic ability.
Collapse
Affiliation(s)
- Huiping Zhao
- Department of CT, Shaanxi Provincial People's Hospital, No. 256, Youyi West Road, Xi'an 710068, Shaanxi Province, China.
| | - Jianbo Gao
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor & Henan International Joint Laboratory of Medical Imaging & Henan Engineering Laboratory of Tumor Imaging & Henan Key Laboratory of CT Imaging & Zhengzhou Key Laboratory of Medical Imaging Technology and Diagnosis, Zhengzhou 450052, Henan Province, China
| | - Biaosheng Bai
- Department of Radiotherapy, People's Hospital of Bayingolin Mongol Autonomous Prefecture, Korla 841000, Xinjiang, China
| | - Rui Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor & Henan International Joint Laboratory of Medical Imaging & Henan Engineering Laboratory of Tumor Imaging & Henan Key Laboratory of CT Imaging & Zhengzhou Key Laboratory of Medical Imaging Technology and Diagnosis, Zhengzhou 450052, Henan Province, China
| | - Juan Yu
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Hao Lu
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor & Henan International Joint Laboratory of Medical Imaging & Henan Engineering Laboratory of Tumor Imaging & Henan Key Laboratory of CT Imaging & Zhengzhou Key Laboratory of Medical Imaging Technology and Diagnosis, Zhengzhou 450052, Henan Province, China
| | - Ming Cheng
- Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor & Henan International Joint Laboratory of Medical Imaging & Henan Engineering Laboratory of Tumor Imaging & Henan Key Laboratory of CT Imaging & Zhengzhou Key Laboratory of Medical Imaging Technology and Diagnosis, Zhengzhou 450052, Henan Province, China; Department of Medical Information, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Pan Liang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China; Henan Key Laboratory of Image Diagnosis and Treatment for Digestive System Tumor & Henan International Joint Laboratory of Medical Imaging & Henan Engineering Laboratory of Tumor Imaging & Henan Key Laboratory of CT Imaging & Zhengzhou Key Laboratory of Medical Imaging Technology and Diagnosis, Zhengzhou 450052, Henan Province, China
| |
Collapse
|
31
|
van der Sluis K, van Sandick JW, van Dieren JM, Vollebergh MA, Grootscholten C, van den Berg JG, Snaebjornsson P, Hartemink KJ, Veenhof AAFA, Chalabi M, Kodach LL. The clinical impact of testing for biomarkers in gastric cancer patients: a real-world cohort. Histopathology 2023; 82:826-836. [PMID: 36694277 DOI: 10.1111/his.14869] [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: 11/04/2022] [Revised: 01/16/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023]
Abstract
BACKGROUND AND AIMS In gastric cancer (GC), HER2 was the first biomarker for guided therapy registered for clinical use. Considering the recent approvals of immune check-point blockade (ICB) in gastro-oesophageal cancers, testing for mismatch repair deficiency (dMMR), Epstein-Barr virus (EBV) and PD-L1 combined positive score (CPS) is becoming increasingly important. Here we describe a real-world cohort on biomarker assessment in GC patients. METHODS Patients diagnosed with GC between 2017 and 2021 were included. Biomarker results were retrieved from electronic patient files. PD-L1 CPS was determined retrospectively on dMMR and EBV-positive (EBV+) tumours. Data on genomic sequencing were analysed separately. RESULTS Of 363 patients identified, 45% had metastatic disease. In 335 patients (92%) at least one biomarker was tested. The prevalence of HER2+, dMMR and EBV+ tumours was 10% (32 of 319), 7% (20 of 294) and 1% (three of 235), respectively. Of the dMMR and EBV+ tumours, 95% had a PD-L1 CPS ≥ 5. Therapeutic strategy was adjusted in 31 of 55 patients and consisted of anti-HER2 therapies as well as ICB in clinical trials. Genomic alterations were found in 44 of 60 tested patients. TP53 (73%) and PIK3CA (20%) mutations were most common, followed by KRAS mutations (11%) and amplifications (11%). CONCLUSIONS In this real-world cohort, testing for HER2, dMMR and EBV status affected treatment decisions in 56% of the patients. Although most dMMR and EBV+ tumours had a PD-L1 CPS ≥ 5, not all patients with a high probability of treatment response are identified. Based on these results, a stepwise diagnostic strategy is proposed.
Collapse
Affiliation(s)
- Karen van der Sluis
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands.,Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Johanna W van Sandick
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Jolanda M van Dieren
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Marieke A Vollebergh
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Cecile Grootscholten
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - José G van den Berg
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Petur Snaebjornsson
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Koen J Hartemink
- Department of Surgical Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | | | - Myriam Chalabi
- Department of Gastrointestinal Oncology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Liudmila L Kodach
- Department of Pathology, Netherlands Cancer Institute, Amsterdam, the Netherlands
| |
Collapse
|
32
|
Bokemeyer C, Paracha N, Lassen U, Italiano A, Sullivan SD, Marian M, Brega N, Garcia-Foncillas J. Survival Outcomes of Patients With Tropomyosin Receptor Kinase Fusion-Positive Cancer Receiving Larotrectinib Versus Standard of Care: A Matching-Adjusted Indirect Comparison Using Real-World Data. JCO Precis Oncol 2023; 7:e2200436. [PMID: 36689698 PMCID: PMC9928633 DOI: 10.1200/po.22.00436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
PURPOSE Larotrectinib, a highly specific tropomyosin receptor kinase (TRK) inhibitor, previously demonstrated high response rates in single-arm trials of patients with TRK fusion-positive cancer, but there are limited data on comparative effectiveness against standard-of-care (SoC) regimens used in routine health care practice, before widespread adoption of TRK inhibitors as SoC for TRK fusion-positive cancers. Matching-adjusted indirect comparison, a validated methodology that balances population characteristics to facilitate cross-trial comparisons, was used to compare the overall survival (OS) of larotrectinib versus non-TRK-inhibitor SoC. MATERIALS AND METHODS Individual patient data from three larotrectinib trials (ClinicalTrials.gov identifiers: NCT02122913, NCT02637687, and NCT02576431) were compared with published aggregate real-world data from patients with locally advanced/metastatic TRK fusion-positive cancer identified in the Flatiron Health/Foundation Medicine database. OS was defined as the time from advanced/metastatic disease diagnosis to death. After matching population characteristics, the analyses included (1) a log-rank test of equality to test whether the two groups were similar before larotrectinib initiation; and (2) estimation of treatment effect of larotrectinib versus non-TRK-inhibitor SoC. These analyses are limited to prognostic variables available in real-world data. RESULTS Eighty-five larotrectinib patients and 28 non-TRK-inhibitor SoC patients were included in the analyses. After matching, log-rank testing showed no difference in baseline characteristics between the two groups (P = .31). After matching, larotrectinib was associated with a 78% lower risk of death, compared with non-TRK-inhibitor SoC (adjusted hazard ratio, 0.22 [95% CI, 0.09 to 0.52]; P = .001); median OS was 39.7 months (95% CI: 16.4, NE [not estimable]) for larotrectinib and 10.2 months (95% CI: 7.2, 14.1) for SoC. CONCLUSION Matching-adjusted indirect comparison analyses suggest longer OS with larotrectinib, compared with non-TRK-inhibitor SoC, in adult patients with TRK fusion-positive cancer.
Collapse
Affiliation(s)
- Carsten Bokemeyer
- University Medical Centre Hamburg Eppendorf, Hamburg, Germany,Carsten Bokemeyer, MD, Department Oncology, Hematology and BMT with Section of Pneumology, Universitaetsklinikum Hamburg—Eppendorf, Martinistrasse 52, D 20246 Hamburg, Germany; e-mail:
| | | | | | | | - Sean D. Sullivan
- CHOICE Institute, School of Pharmacy, University of Washington, Seattle, WA
| | | | | | - Jesus Garcia-Foncillas
- University Cancer Institute and the Department of Oncology, University Hospital Fundacion Jimenez Diaz, Autonomous University, Madrid, Spain
| |
Collapse
|
33
|
Bando H, Ohtsu A, Yoshino T. Therapeutic landscape and future direction of metastatic colorectal cancer. Nat Rev Gastroenterol Hepatol 2023; 20:306-322. [PMID: 36670267 DOI: 10.1038/s41575-022-00736-1] [Citation(s) in RCA: 69] [Impact Index Per Article: 34.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/16/2022] [Indexed: 01/22/2023]
Abstract
In the era of targeted therapy based on genomic alterations, the treatment strategy for metastatic colorectal cancer (mCRC) has been changing. Before systemic treatment initiation, determination of tumour genomic status for KRAS and NRAS, BRAFV600E mutations, ERBB2, and microsatellite instability and/or mismatch repair (MMR) status is recommended. In patients with deficient MMR and BRAFV600E mCRC, randomized phase III trials have established the efficacy of pembrolizumab as first-line therapy and the combination of encorafenib and cetuximab as second-line or third-line therapy. In addition, new agents have been actively developed in other rare molecular fractions such as ERBB2 alterations and KRASG12C mutations. In March 2022, the combination of pertuzumab and trastuzumab for ERBB2-positive mCRC was approved in Japan, thereby combining real-world evidence from the SCRUM-Japan Registry. As the populations are highly fragmented owing to rare genomic alterations, various strategies in clinical development are expected. Clinical development of a tumour-agnostic approach, such as NTRK fusion and tumour mutational burden, has successfully introduced corresponding drugs to clinical practice. Considering the difficulty of randomized trials owing to cost-benefit and rarity, a promising solution could be real-world evidence utilized as an external control from the molecular-based disease registry.
Collapse
Affiliation(s)
- Hideaki Bando
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Atsushi Ohtsu
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
| |
Collapse
|
34
|
Vingiani A, Lorenzini D, Conca E, Volpi CC, Trupia DV, Gloghini A, Perrone F, Tamborini E, Dagrada GP, Agnelli L, Capone I, Busico A, Pruneri G. Pan-TRK immunohistochemistry as screening tool for NTRK fusions: A diagnostic workflow for the identification of positive patients in clinical practice. Cancer Biomark 2023; 38:301-309. [PMID: 37545217 DOI: 10.3233/cbm-220357] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/08/2023]
Abstract
BACKGROUND Pan-TRK inhibitors Entrectinib and Larotrectinib have been recently approved as tumor-agnostic therapies in NTRK1-2-3 rearranged patients and there is therefore an urgent need to identify reliable and accessible biomarkers for capturing NTRK fusions in the real-world practice. OBJECTIVE We aim to assess the analytical validity of the recently released pan-TRK assay (Ventana), running a head-to-head comparison between immunohistochemistry and Archer FusionPlex Lung Panel (ArcherDX) that is designed to detect key fusions in 13 genes, also including NTRK1-3. METHODS Pan-TRK IHC and NGS analysis were conducted on a retrospective/prospective cohort of 124 cancer patients (carcinomas, 93 cases; soft tissue sarcomas, 19; primary central nervous system tumours, 10; and neuroblastomas, 2). FISH data were available in most of the IHC/NGS discordant cases. RESULTS A comparison between IHC and NGS results was carried out in 117 cases: among 30 pan-TRK positive cases, NTRK rearrangement by NGS was found in 11 (37%), while one of the 87 (1.1%) pan-TRK negative cases (a case of NSCLC) showed a TPM3-NRTK1 rearrangement by NGS. Accordingly, sensitivity and specificity of IHC in predicting NTRK status were 91.7% and 81.9%, respectively, while negative (NPV) and positive predictive value (PPV) were 98.8% and 36.7%, respectively. CONCLUSIONS These data lead to suggest that IHC with VENTANA pan-TRK antibody can be a reliable screening tool for the identification of patients potentially bearing NTRK rearranged tumours.
Collapse
Affiliation(s)
- Andrea Vingiani
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Daniele Lorenzini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Elena Conca
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Chiara Costanza Volpi
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Desirè Viola Trupia
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Annunziata Gloghini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Federica Perrone
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Elena Tamborini
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Gian Paolo Dagrada
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Luca Agnelli
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Iolanda Capone
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
| | - Adele Busico
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
| | - Giancarlo Pruneri
- Department of Diagnostic Innovation, Foundation IRCCS National Cancer Institute, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
- Department of Oncology and Hematoncology, University of Milan, Milan, Italy
| |
Collapse
|
35
|
Petaccia de Macedo M, Toledo Nascimento EC, Soares FA, Costa Santini F, D'Almeida Costa F, Werneck da Cunha I, Ramella Munhoz R, De Marchi P, Carnier Jorge TW, Ramos Moreira Leite K. Brazilian Expert Consensus for NTRK Gene Fusion Testing in Solid Tumors. CLINICAL PATHOLOGY (THOUSAND OAKS, VENTURA COUNTY, CALIF.) 2023; 16:2632010X231197080. [PMID: 37719804 PMCID: PMC10504829 DOI: 10.1177/2632010x231197080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 07/31/2023] [Indexed: 09/19/2023]
Abstract
Oncogenic neurotrophic tropomyosin receptor kinase gene fusions occur in less than 1% of common cancers. These mutations have emerged as new biomarkers in cancer genomic profiling with the approval of selective drugs against tropomyosin receptor kinase fusion proteins. Nevertheless, the optimal pathways and diagnostic platforms for this biomarker's screening and genomic profiling have not been defined and remain a subject of debate. A panel of national experts in molecular cancer diagnosis and treatment was convened by videoconference and suggested topics to be addressed in the literature review. The authors proposed a testing algorithm for oncogenic neurotrophic tropomyosin receptor kinase gene fusion screening and diagnosis for the Brazilian health system. This review aims to discuss the latest literature evidence and international consensus on neurotrophic tropomyosin receptor kinase gene fusion diagnosis to devise clinical guidelines for testing this biomarker. We propose an algorithm in which testing for this biomarker should be requested to diagnose advanced metastatic tumors without known driver mutations. In this strategy, Immunohistochemistry should be used as a screening test followed by confirmatory next-generation sequencing in immunohistochemistry-positive cases.
Collapse
Affiliation(s)
| | | | - Fernando Augusto Soares
- Rede D'Or São Luiz, São Paulo, Brazil
- D'Or Institute for Research and Teaching (IDOR), São Paulo, Brazil
| | | | | | - Isabela Werneck da Cunha
- Rede D'Or São Luiz, São Paulo, Brazil
- D'Or Institute for Research and Teaching (IDOR), São Paulo, Brazil
| | | | | | | | | |
Collapse
|
36
|
Abstract
INTRODUCTION Neurotrophic tyrosine receptor kinase (NTRK) gene fusions occur in ~ 0.3% of all solid tumours but are enriched in some rare tumour types. Tropomyosin receptor kinase (TRK) inhibitors larotrectinib and entrectinib are approved as tumour-agnostic therapies for solid tumours harbouring NTRK fusions. METHODS This study investigated the prevalence of NTRK fusions in Canadian patients and also aimed to help guide NTRK testing paradigms through analysis of data reported from a national clinical diagnostic testing program between September 2019 and July 2021. RESULTS Of 1,687 patients included in the final analysis, NTRK fusions were detected in 0.71% (n = 12) of patients representing salivary gland carcinoma (n = 3), soft tissue sarcoma (n = 3), CNS (n = 3), and one in each of melanoma, lung, and colorectal cancer. All three salivary gland carcinomas contained ETV6-NTRK3 fusions. Thirteen (0.77%) clinically actionable incidental findings were also detected. Two of the 13 samples containing incidental findings were NTRK fusion-positive (GFOD1-NTRK2, FGFR3-TACC3 in a glioblastoma and AFAP1-NTRK2, BRAF c.1799T>A in a glioma). The testing algorithm screened most patient samples via pan-TRK immunohistochemistry (IHC), whereas samples from the central nervous system (CNS), pathognomonic cancers, and confirmed/ putative NTRK fusion-positive samples identified under research protocols were reflexed straight to next-generation sequencing (NGS). CONCLUSION These findings highlight the benefit and practicality of a diagnostic testing program to identify patients suitable for tumour-agnostic TRK inhibitor therapies, as well as other targeted therapies, due to clinically actionable incidental findings identified. Collectively, these findings may inform future guidance on selecting the appropriate testing approach per tumour type and on optimal NTRK testing algorithms.
Collapse
|
37
|
Nakayama I, Shinozaki E, Kawachi H, Sasaki T, Yunokawa M, Tomomatsu J, Yuasa T, Kitazono S, Kobayashi K, Hayakawa K, Ueki A, Takahashi S, Yamaguchi K. Implementation of microsatellite instability testing for the assessment of solid tumors in clinical practice. Cancer Med 2022; 12:7932-7940. [PMID: 36573309 PMCID: PMC10134335 DOI: 10.1002/cam4.5569] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 12/02/2022] [Accepted: 12/16/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND In Japan, microsatellite instability (MSI) testing for solid tumors was introduced in clinical practice in December 2018. Although immune checkpoint inhibitors (ICIs) are established standards of care for patients with MSI-high tumors, the status of implementing MSI testing in clinical practice remains unclear. METHODS We retrospectively reviewed the medical records of patients with solid tumors who underwent MSI testing between January 2019 and December 2020 at our institution. RESULTS In total, 1,052 MSI tests were performed in 1,047 patients. Regardless of specimen volume and condition, the MSI status was successfully determined in 1,041 (99.0%) tests, encompassing 27 tumor types (microsatellite stable [MSS] or MSI-low: n = 991 [95.2%] and MSI-high: n = 50 [4.8%]). Patients whose specimens were fixed with 20% neutral buffered formalin (NBF) and who had specimens with prolonged storage (98.4% and 95.4%) showed lower success rates than those whose specimens were fixed with 10% NBF and who had specimens with nonprolonged storage (100.0% and 99.6%), respectively. The prolonged turnaround time (TAT) in MSI-high cases (median TAT: 24 days) was a critical issue that directly resulted in treatment delay. Of the 50 patients with MSI-high tumors, 24 (48.0%) received ICIs and 34 (68.0%) were referred to the Department of Clinical Genetic Oncology where 6 (12.0%) patients were diagnosed with Lynch syndrome. CONCLUSIONS MSI testing was successfully performed for various types of tumors and specimens in clinical practice. Our study results identified certain issues associated with the clinical implementation of MSI testing, including optimal specimen selection, extended TAT in MSI-high cases, and awareness of hereditary tumors.
Collapse
Affiliation(s)
- Izuma Nakayama
- Department of Gastroenterological Chemotherapy Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Hiroshi Kawachi
- Department of Pathology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Takashi Sasaki
- Department of Hepato‐Biliary‐Pancreatic Medicine Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Mayu Yunokawa
- Department of Medical Oncology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
- Department of Gynecologic Oncology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Junichi Tomomatsu
- Department of Medical Oncology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Takeshi Yuasa
- Division of Genitourinary Chemotherapy Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Satoru Kitazono
- Department of Thoracic Medical Oncology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Kokoro Kobayashi
- Department of Breast Medical Oncology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Keiko Hayakawa
- Department of Orthopedic Oncology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Arisa Ueki
- Department of Clinical Genetic Oncology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Shunji Takahashi
- Department of Medical Oncology Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy Cancer Institute Hospital of the Japanese Foundation for Cancer Research Tokyo Japan
| |
Collapse
|
38
|
Kang YJ, O'Haire S, Franchini F, IJzerman M, Zalcberg J, Macrae F, Canfell K, Steinberg J. A scoping review and meta-analysis on the prevalence of pan-tumour biomarkers (dMMR, MSI, high TMB) in different solid tumours. Sci Rep 2022; 12:20495. [PMID: 36443366 PMCID: PMC9705554 DOI: 10.1038/s41598-022-23319-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 10/29/2022] [Indexed: 11/29/2022] Open
Abstract
Immune checkpoint inhibitors have been approved in the USA for tumours exhibiting mismatch repair deficiency (dMMR), microsatellite instability (MSI), or high tumour mutational burden (TMB), with regulatory and reimbursement applications in multiple other countries underway. As the estimated budget impacts of future reimbursements depend on the size of the potential target population, we performed a scoping review and meta-analysis of the prevalence of these pan-tumour biomarkers in different cancers. We systematically searched Medline/Embase and included studies reporting the prevalence of dMMR/MSI/high TMB in solid tumours published 01/01/2018-31/01/2021. Meta-analyses were performed separately for the pan-cancer prevalence of each biomarker, and by cancer type and stage where possible. The searches identified 3890 papers, with 433 prevalence estimates for 32 different cancer types from 201 studies included in meta-analyses. The pooled overall prevalence of dMMR, MSI and high TMB (≥ 10 mutations/Mb) in pan-cancer studies was 2.9%, 2.7% and 14.0%, respectively. The prevalence profiles of dMMR/MSI and high TMB differed across cancer types. For example, endometrial, colorectal, small bowel and gastric cancers showed high prevalence of both dMMR and MSI (range: 8.7-26.8% and 8.5-21.9%, respectively) and high TMB (range: 8.5-43.0%), while cervical, esophageal, bladder/urothelial, lung and skin cancers showed low prevalence of dMMR and MSI (< 5%), but high prevalence of high TMB (range: 23.7-52.6%). For other cancer types, prevalence of all three biomarkers was generally low (< 5%). This structured review of dMMR/MSI/high TMB prevalence across cancers and for specific cancer types and stages provide timely evidence to inform budget impact forecasts in health technology assessments for drug approvals based on these pan-tumour biomarkers.
Collapse
Affiliation(s)
- Yoon-Jung Kang
- The Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council NSW, Sydney, NSW, 2060, Australia
| | - Sophie O'Haire
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3053, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Fanny Franchini
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3053, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - Maarten IJzerman
- Cancer Health Services Research, Centre for Health Policy, Melbourne School of Population and Global Health, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Parkville, VIC, 3053, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Sciences, The University of Melbourne, Melbourne, VIC, 3010, Australia
| | - John Zalcberg
- Department of Medical Oncology, Alfred Health and School of Public Health and Preventive Medicine, Faculty of Medicine, Monash University, Melbourne, VIC, 3004, Australia
| | - Finlay Macrae
- Colorectal Medicine and Genetics, and Department of Medicine, University of Melbourne, The Royal Melbourne Hospital, Parkville, VIC, 3050, Australia
| | - Karen Canfell
- The Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council NSW, Sydney, NSW, 2060, Australia
| | - Julia Steinberg
- The Daffodil Centre, The University of Sydney, a Joint Venture with Cancer Council NSW, Sydney, NSW, 2060, Australia.
| |
Collapse
|
39
|
Detecting mismatch repair deficiency in solid neoplasms: immunohistochemistry, microsatellite instability, or both? Mod Pathol 2022; 35:1515-1528. [PMID: 35668150 DOI: 10.1038/s41379-022-01109-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 12/25/2022]
Abstract
In managing patients with solid tumors, the value of detecting the status of tumor DNA mismatch repair function is widely recognized. Mismatch repair protein immunohistochemistry and molecular microsatellite instability testing constitute the two major test modalities currently in use, yet each is associated with caveats and limitations that can be consequential. Most notably, the traditional approach of defining mismatch repair protein immunohistochemistry abnormality by complete loss of staining in all tumor cells is evolving. Partial or clonal loss is becoming recognized as a manifestation of gene abnormality; in some cases, such clonal loss is associated with germline pathogenic variants. The current criteria and cutoff values for defining microsatellite instability-high are developed primarily according to colorectal tumors. Non-colorectal cases, and occasionally even colorectal tumors, that are mismatch repair-deficient by immunohistochemistry but not microsatellite instability-high by current standards are being recognized. Emerging data suggest that these immunohistochemistry abnormal / non-microsatellite instability-high cases warrant further genetic workup for Lynch syndrome detection. Whether these tumors respond to immunotherapy is a question still to be addressed. It is imperative that pathologists as well as clinicians and investigators be aware of such intricacies regarding routine immunohistochemistry and microsatellite instability testing and the results they generate. This review summarizes our current understanding of the advantages and limitations of these tests and offer our view on what constitutes the most optimal strategy in test selection and how best to utilize case context to enhance the interpretation of the test results.
Collapse
|
40
|
Yamada Y, Bohnenberger H, Kriegsmann M, Kriegsmann K, Sinn P, Goto N, Nakanishi Y, Seno H, Chigusa Y, Fujimoto M, Minamiguchi S, Haga H, Simon R, Sauter G, Ströbel P, Marx A. Tuft cell-like carcinomas: novel cancer subsets present in multiple organs sharing a unique gene expression signature. Br J Cancer 2022; 127:1876-1885. [PMID: 35999270 PMCID: PMC9643388 DOI: 10.1038/s41416-022-01957-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 08/06/2022] [Accepted: 08/09/2022] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Tuft cells are chemosensory epithelial cells playing a role in innate immunity. Recent studies revealed cancers with a tuft cell-like gene expression signature in the thorax. We wondered whether this signature might also occur in extrathoracic cancers. METHODS We examined mRNA expression of tuft cell markers (POU2F3, GFI1B, TRPM5, SOX9, CHAT, and AVIL) in 19 different types of cancers in multiple extrathoracic organs with The Cancer Genome Atlas (TCGA) (N = 6322). Four different extrathoracic cancers in our local archives (N = 909) were analysed by immunohistochemistry. RESULTS Twenty-two (0.35%) extrathoracic tumours with co-expression of POU2F3 and other tuft cell markers were identified in various TCGA datasets. Twelve of the 22 "tuft cell-like tumours" shared poor differentiation and a gene expression pattern, including KIT, anti-apoptotic BCL2, and ionocyte-associated genes. In our archival cases, eleven (1.21%) tumours co-expressing POU2F3, KIT, and BCL2 on immunohistochemistry, i.e., were presumable tuft cell-like cancers. In three among five TCGA cohorts, the tuft cell-like cancer subsets expressed SLFN11, a promising biomarker of PARP inhibitor susceptibility. CONCLUSIONS Tuft cell-like carcinomas form distinct subsets in cancers of many organs. It appears warranted to investigate their shared gene expression signature as a predictive biomarker for novel therapeutic strategies.
Collapse
Affiliation(s)
- Yosuke Yamada
- Institute of Pathology, University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
- Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan.
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
- German Center for Lung Cancer Research (DZL), Heidelberg, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Peter Sinn
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Norihiro Goto
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yuki Nakanishi
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Yoshitsugu Chigusa
- Department of Gynecology and Obstetrics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | - Hironori Haga
- Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Ronald Simon
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Guido Sauter
- Institute of Pathology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Philipp Ströbel
- Institute of Pathology, University Medical Center Göttingen, University of Göttingen, Göttingen, Germany
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim and Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
41
|
Khaddour K, Felipe Fernandez M, Khabibov M, Garifullin A, Dressler D, Topchu I, Patel JD, Weinberg F, Boumber Y. The Prognostic and Therapeutic Potential of DNA Damage Repair Pathway Alterations and Homologous Recombination Deficiency in Lung Cancer. Cancers (Basel) 2022; 14:5305. [PMID: 36358724 PMCID: PMC9654807 DOI: 10.3390/cancers14215305] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/19/2022] [Accepted: 10/26/2022] [Indexed: 03/28/2025] Open
Abstract
Lung cancer remains the second most commonly diagnosed cancer worldwide and the leading cause of cancer-related mortality. The mapping of genomic alterations and their role in lung-cancer progression has been followed by the development of new therapeutic options. Several novel drugs, such as targeted therapy and immunotherapy, have significantly improved outcomes. However, many patients with lung cancer do not benefit from existing therapies or develop progressive disease, leading to increased morbidity and mortality despite initial responses to treatment. Alterations in DNA-damage repair (DDR) genes represent a cancer hallmark that impairs a cell's ability to prevent deleterious mutation accumulation and repair. These alterations have recently emerged as a therapeutic target in breast, ovarian, prostate, and pancreatic cancers. The role of DDR alterations remains largely unknown in lung cancer. Nevertheless, recent research efforts have highlighted a potential role of some DDR alterations as predictive biomarkers of response to treatment. Despite the failure of PARP inhibitors (main class of DDR targeting agents) to improve outcomes in lung cancer patients, there is some evidence suggesting a role of PARP inhibitors and other DDR targeting agents in benefiting a distinct subset of lung cancer patients. In this review, we will discuss the existing literature on DDR alterations and homologous recombination deficiency (HRD) state as predictive biomarkers and therapeutic targets in both non-small cell lung and small cell lung cancer.
Collapse
Affiliation(s)
- Karam Khaddour
- Division of Hematology and Oncology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Manuel Felipe Fernandez
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Marsel Khabibov
- I. M. Sechenov First Moscow State Medical University, 119992 Moscow, Russia
| | - Airat Garifullin
- P. Hertsen Moscow Oncology Research Institute, National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, 125284 Moscow, Russia
| | - Danielle Dressler
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Iuliia Topchu
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jyoti D. Patel
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Frank Weinberg
- Division of Hematology and Oncology, Department of Medicine, University of Illinois Chicago, Chicago, IL 60612, USA
| | - Yanis Boumber
- Robert H. Lurie Comprehensive Cancer Center, Division of Hematology/Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420012 Kazan, Russia
| |
Collapse
|
42
|
Kanai M. Current Clinical Practice of Precision Medicine Using Comprehensive Genomic Profiling Tests in Biliary Tract Cancer in Japan. Curr Oncol 2022; 29:7272-7284. [PMID: 36290850 PMCID: PMC9599999 DOI: 10.3390/curroncol29100573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
With the recent advances of next generation sequencing technologies, comprehensive genomic profiling (CGP) tests, which are designed to measure more than hundreds of cancer-related genes at a time, have now been widely introduced into daily clinical practice. For the patients whose tumor samples are not fit for tissue-based CGP tests, a blood-based CGP test (liquid biopsy) is available as an alternative option. Three CGP tests, "OncoGuide NCC™Oncopanel System (124 genes)", "FoundationOne®CDx (324 genes)", and "Founda-tionOne®CDx Liquid (324 genes)", are now reimbursed by public insurance in 233 hospitals designated for cancer genomic medicine in Japan. In biliary tract cancer, the prevalence of druggable variants is relatively higher compared to other cancer types and the European Society for Medical Oncology recommends routine use of CGP tests for advanced biliary tract cancer to guide treatment options. The latest National Cancer Center Network guideline lists eight druggable markers (NTRK fusion, MSI-H, TMB-H, BRAF V600E, FGFR2 fusions/rearrangement, IDH1 mutations, RET fusion, and HER2 overexpression) and matched therapies. In Japan, matched therapies for four markers (NTRK, MSI-H, TMB-H, and FGFR2) are reimbursed by public insurance (as of September 2022). The progress of genomic profiling technology will contribute to the improvement of the dismal clinical outcomes of this disease in the future.
Collapse
Affiliation(s)
- Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
43
|
Çocukluk Çağı Kanserlerinde NTRK Somatik Füzyonları ve Tümör Agnostik Tedavi. JOURNAL OF CONTEMPORARY MEDICINE 2022. [DOI: 10.16899/jcm.1113357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Nörotrofik tirozin reseptör kinaz (NTRK) geni yeniden düzenlemeleri yakın zamanda kanser tedavisi için yeni hedefler olarak ortaya konulan biyobelirteçlerden (biyomarker) bir tanesi olarak tanımlanmış ve geliştirilmiştir. NTRK gen füzyonları öngörücü (prediktif-tanısal) bir biyobelirteç olarak kullanılmasının yanı sıra tedavi hedefi olarak da kullanılarak bireyselleştirilmiş hedef tedavide yerini almıştır. NTRK füzyon proteinlerinin selektif inhibitörleri, NTRK füzyon pozitif solid tümörlerin tedavisinde güçlü etkinliğe sahiptir (tümör-agnostik tedavi). Tümörlerinde NTRK füzyonları saptanan hastaların tedavisinde etkili olan FDA (Amerika Birleşik Devletleri Gıda ve İlaç Yönetimi) onaylı yeni tedavilerle birlikte, bu füzyonların test edilmesi önemli hale gelmiştir. Yapılan klinik çalışmalar birinci nesil tirozin reseptör kinaz (TRK) inhibitörleri olan larotrectinib ve entrectinibin NTRK füzyonu pozitif kanserlerin tedavisinde yüksek oranda başarılı olduğu görülmüştür. İlerleyen zamanlarda bu ilaçlar üzerine geniş kapsamlı araştırmaların sayısının artması bu ilaçlar hakkında daha fazla bilgiyi mevcut kılacak ve faydalı olacaktır.
Collapse
|
44
|
Lim KHT, Kong HL, Chang KTE, Tan DSW, Tan IBH, Mohamad F, Soh SY, Pang BN, Soo RA, Choo SP, Hsieh W, Aung L. Recommended testing algorithms for NTRK gene fusions in pediatric and selected adult cancers: Consensus of a Singapore Task Force. Asia Pac J Clin Oncol 2022; 18:394-403. [PMID: 34806337 PMCID: PMC9541932 DOI: 10.1111/ajco.13727] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/27/2021] [Indexed: 01/09/2023]
Abstract
The occurrence of neurotrophic tyrosine receptor kinase (NTRK) gene fusions in a wide range of tumor types presents an attractive opportunity for using a tropomyosin receptor kinase (TRK) inhibitor as cancer therapy. Recent clinical studies have demonstrated highly efficacious outcomes associated with the use of TRK inhibitors, such as larotrectinib and entrectinib in NTRK fusion-bearing cancers, in both adult and pediatric populations. While NTRK gene fusions are commonly found in some uncommon adult and pediatric malignancies, they are also found, albeit rarely, in a wide range of more common malignancies. The potential value of testing for NTRK gene fusions in practically all advanced malignancies is underpinned by the remarkable therapeutic outcomes that TRK inhibitors offer. This requirement presents practical and financial challenges in real-world oncological practice. Furthermore, different testing platforms exist to detect NTRK gene fusions, each with its advantages and disadvantages. It is, therefore, imperative to develop strategies for NTRK gene fusion testing in an attempt to optimize the use of limited tissue specimen and financial resources, and to minimize the turnaround time. A multidisciplinary task force of Singapore medical experts in both public and private sectors was convened in late 2020 to propose testing algorithms for adult colorectal tumors, sarcomas, non-small cell lung cancer, and pediatric cancers, with particular adaptation to the Singapore oncological practice. The recommendations presented here highlight the heterogeneity of NTRK-fusion positive cancers, and emphasize the need to customize the testing methods to each tumor type to optimize the workflow.
Collapse
Affiliation(s)
- Kiat Hon Tony Lim
- Division of PathologyDepartment of Anatomical PathologySingapore General HospitalSingapore
| | - Hwai Loong Kong
- ICON Cancer CentreMount Elizabeth Medical Centre OrchardSingapore
| | - Kenneth Tou En Chang
- Department of Pathology and Laboratory MedicineKK Women's and Children's HospitalSingapore
| | | | - Iain Bee Huat Tan
- Division of Medical OncologyNational Cancer Centre SingaporeSingapore
| | - Farid Mohamad
- Division of Medical OncologyNational Cancer Centre SingaporeSingapore
| | - Shui Yen Soh
- Haematology‐Oncology ServiceDepartment of Paediatrics SubspecialtiesKK Women's and Children's HospitalSingapore
| | | | - Ross Andrew Soo
- Department of Haematology‐OncologyNational University Cancer InstituteNational University Health SystemSingapore
| | | | | | - LeLe Aung
- Paediatric Haematology‐OncologyICON Cancer CentreSingapore
| |
Collapse
|
45
|
Imai M, Nakamura Y, Sunami K, Kage H, Komine K, Koyama T, Amano T, Ennishi D, Kanai M, Kenmotsu H, Maeda T, Morita S, Sakai D, Bando H, Makiyama A, Suzuki T, Hirata M, Kohsaka S, Tsuchihara K, Naito Y, Yoshino T. Expert Panel Consensus Recommendations on the Use of Circulating Tumor DNA Assays for Patients with Advanced Solid Tumors. Cancer Sci 2022; 113:3646-3656. [PMID: 35876224 PMCID: PMC9633310 DOI: 10.1111/cas.15504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/12/2022] [Accepted: 07/16/2022] [Indexed: 12/01/2022] Open
Abstract
Comprehensive genomic profiling is increasingly used to facilitate precision oncology based on molecular stratification. In addition to conventional tissue comprehensive genomic profiling, comprehensive genomic profiling of circulating tumor DNA has become widely utilized in cancer care owing on its advantages, including less invasiveness, rapid turnaround time, and capturing heterogeneity. However, circulating tumor DNA comprehensive genomic profiling has some limitations, mainly false negatives due to low levels of plasma circulating tumor deoxyribonucleic acid and false positives caused by clonal hematopoiesis. Nevertheless, no guidelines and recommendations fully address these issues. Here, an expert panel committee involving representatives from 12 Designated Core Hospitals for Cancer Genomic Medicine in Japan was organized to develop expert consensus recommendations for the use of circulating tumor deoxyribonucleic acid‐based comprehensive genomic profiling. The aim was to generate guidelines for clinicians and allied healthcare professionals on the optimal use of the circulating tumor DNA assays in advanced solid tumors and to aid the design of future clinical trials that utilize and develop circulating tumor DNA assays to refine precision oncology. Fourteen clinical questions regarding circulating tumor deoxyribonucleic acid comprehensive genomic profiling including the timing of testing and considerations for interpreting results were established by searching and curating associated literatures, and corresponding recommendations were prepared based on the literature for each clinical question. Final consensus recommendations were developed by voting to determine the level of each recommendation by the Committee members.
Collapse
Affiliation(s)
- Mitsuho Imai
- Translational Research Support Section, National Cancer Center Hospital East.,Genomics Unit, Keio University School of Medicine
| | - Yoshiaki Nakamura
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital
| | - Hidenori Kage
- Department of Next-Generation Precision Medicine Development Laboratory, Graduate School of Medicine, The University of Tokyo
| | - Keigo Komine
- Department of Medical Oncology, Tohoku University Hospital
| | - Takafumi Koyama
- Department of Experimental Therapeutics, National Cancer Center Hospital
| | - Toraji Amano
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital
| | - Daisuke Ennishi
- Center for Comprehensive Genomic Medicine, Okayama University Hospital
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University
| | | | - Takahiro Maeda
- Division of Precision Medicine, Kyushu University Graduate School of Medical Sciences
| | - Sachi Morita
- Department of Clinical Oncology and Chemotherapy, Nagoya University Hospital
| | - Daisuke Sakai
- Center for Cancer Genomics and Personalized Medicine, Osaka University Hospital
| | - Hideaki Bando
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| | | | - Tatsuya Suzuki
- Department of Hematology, National Cancer Center Hospital
| | - Makoto Hirata
- Department of Genetic Medicine and Services, National Cancer Center Hospital
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Japan
| | - Yoichi Naito
- Department of General Internal medicine/Experimental Therapeutics/Medical Oncology, National Cancer Center Hospital East
| | - Takayuki Yoshino
- Translational Research Support Section, National Cancer Center Hospital East.,Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East
| |
Collapse
|
46
|
Shonka DC, Ho A, Chintakuntlawar AV, Geiger JL, Park JC, Seetharamu N, Jasim S, Abdelhamid Ahmed AH, Bible KC, Brose MS, Cabanillas ME, Dabekaussen K, Davies L, Dias-Santagata D, Fagin JA, Faquin WC, Ghossein RA, Gopal RK, Miyauchi A, Nikiforov YE, Ringel MD, Robinson B, Ryder MM, Sherman EJ, Sadow PM, Shin JJ, Stack BC, Tuttle RM, Wirth LJ, Zafereo ME, Randolph GW. American Head and Neck Society Endocrine Surgery Section and International Thyroid Oncology Group consensus statement on mutational testing in thyroid cancer: Defining advanced thyroid cancer and its targeted treatment. Head Neck 2022; 44:1277-1300. [PMID: 35274388 DOI: 10.1002/hed.27025] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The development of systemic treatment options leveraging the molecular landscape of advanced thyroid cancer is a burgeoning field. This is a multidisciplinary evidence-based statement on the definition of advanced thyroid cancer and its targeted systemic treatment. METHODS An expert panel was assembled, a literature review was conducted, and best practice statements were developed. The modified Delphi method was applied to assess the degree of consensus for the statements developed by the author panel. RESULTS A review of the current understanding of thyroid oncogenesis at a molecular level is presented and characteristics of advanced thyroid cancer are defined. Twenty statements in topics including the multidisciplinary management, molecular evaluation, and targeted systemic treatment of advanced thyroid cancer are provided. CONCLUSIONS With the growth in targeted treatment options for thyroid cancer, a consensus definition of advanced disease and statements regarding the utility of molecular testing and available targeted systemic therapy is warranted.
Collapse
Affiliation(s)
- David C Shonka
- Department of Otolaryngology - Head and Neck Surgery, University of Virginia, Charlottesville, Virginia, USA
| | - Alan Ho
- Department of Hematology and Medical Oncology, Solid Tumor Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Jessica L Geiger
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio, USA
| | - Jong C Park
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Nagashree Seetharamu
- Division of Hematology-Oncology, Donald and Barbara Zucker School of Medicine at Hofstra University, New Hyde Park, New York, USA
| | - Sina Jasim
- Division of Endocrinology, Metabolism and Lipid Research, Department of Internal Medicine, School of Medicine, Washington University in St. Louis, Saint Louis, Missouri, USA
| | - Amr H Abdelhamid Ahmed
- Department of Otolaryngology - Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| | - Keith C Bible
- Division of Medical Oncology, Mayo Clinic, Rochester, Minnesota, USA
| | - Marcia S Brose
- Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Maria E Cabanillas
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Kirsten Dabekaussen
- Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Louise Davies
- Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire, USA
| | - Dora Dias-Santagata
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - James A Fagin
- Endocrinology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - William C Faquin
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ronald A Ghossein
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Raj K Gopal
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | | | - Yuri E Nikiforov
- Department of Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Matthew D Ringel
- Division of Endocrinology, Diabetes, and Metabolism, The Ohio State University Medical Center, Columbus, Ohio, USA
| | - Bruce Robinson
- Northern Clinical School, Sydney Medical School, Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
| | - Mabel M Ryder
- Division of Endocrinology, Diabetes, Metabolism, & Nutrition, Mayo Clinic, Rochester, Minnesota, USA
| | - Eric J Sherman
- Head and Neck Oncology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Peter M Sadow
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jennifer J Shin
- Department of Otolaryngology - Head and Neck Surgery, Center for Surgery and Public Health, Harvard Medical School, Boston, Massachusetts, USA
| | - Brendan C Stack
- Department of Otolaryngology - Head and Neck Surgery, Southern Illinois University School of Medicine, Springfield, Illinois, USA
| | - R Michael Tuttle
- Endocrinology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Lori J Wirth
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Mark E Zafereo
- Department of Head and Neck Surgery, The University of Texas M. D. Anderson Cancer Center, Houston, Texas, USA
| | - Gregory W Randolph
- Department of Otolaryngology - Head and Neck Surgery, Massachusetts Eye and Ear Infirmary, Boston, Massachusetts, USA
| |
Collapse
|
47
|
Qiao PP, Tian KS, Han LT, Ma B, Shen CK, Zhao RY, Zhang Y, Wei WJ, Chen XP. Correlation of mismatch repair deficiency with clinicopathological features and programmed death-ligand 1 expression in thyroid carcinoma. Endocrine 2022; 76:660-670. [PMID: 35366156 DOI: 10.1007/s12020-022-03031-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/24/2022] [Indexed: 12/23/2022]
Abstract
BACKGROUND Mutations in DNA mismatch repair (MMR) genes associated with thyroid carcinoma (TC) have rarely been reported, especially in East Asian populations. METHODS We examined tumor tissue from a cohort of 241 patients diagnosed with TC between 2008 and 2020. MMR proteins were detected using tissue microarray-based immunohistochemistry in order to identify MMR-protein-deficient (MMR-D) and MMR-protein-intact (MMR-I) tumors. We retrospectively summarized the clinicopathologic characteristics of patients with MMR-D TC, measured the expression of PD-L1, and recorded overall survival (OS) and other clinical outcomes. RESULTS In our cohort, there were 18 (7.5%) MMR-D (MLH1, MSH2, MSH6, and PMS2) patients, including 12 with papillary TC (PTC) (6.7%), 2 with poorly differentiated TC (PDTC) (4.7%), and 4 with anaplastic TC (ATC) (22.2%). Half of them (9/18) showed a specific deletion in MSH6, and 6 of them also carried variants in the MSH6 and PMS2 gene. Survival was significantly better in patients with MMR-D ATC than in those with MMR-I tumors (p = 0.033). Four of the 18 MMR-D patients (22%) were found to be PD-L1 positive. Their OS was much shorter than that of PD-L1-negative patients. CONCLUSIONS MMR-D and PD-L1 positivity appear to be associated with clinicopathological characteristics and prognosis in TC. The results indicate that MMR status may have important prognostic significance in TC. Therefore, immune checkpoint inhibitors that target the PD-1/PD-L1 pathway may be a treatment option for TCs.
Collapse
Affiliation(s)
- Pei-Pei Qiao
- Ningxia Medical University, Yinchuan, 750004, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Kai-Sai Tian
- Ningxia Medical University, Yinchuan, 750004, China
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Li-Tao Han
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Ben Ma
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Cen-Kai Shen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Run-Yu Zhao
- Ningxia Medical University, Yinchuan, 750004, China
| | - Yi Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China
| | - Wen-Jun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Xiao-Ping Chen
- Department of Otorhinolaryngology Head and Neck Surgery, Shanghai Pudong New Area Gongli Hospital, Shanghai, 200135, China.
| |
Collapse
|
48
|
Ukkola I, Nummela P, Kero M, Tammio H, Tuominen J, Kairisto V, Kallajoki M, Haglund C, Peltomäki P, Kytölä S, Ristimäki A. Gene fusions and oncogenic mutations in MLH1 deficient and BRAFV600E wild-type colorectal cancers. Virchows Arch 2022; 480:807-817. [PMID: 35237889 PMCID: PMC9023403 DOI: 10.1007/s00428-022-03302-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 01/09/2023]
Abstract
Gene fusions can act as oncogenic drivers and offer targets for cancer therapy. Since fusions are rare in colorectal cancer (CRC), their universal screening seems impractical. Our aim was to investigate gene fusions in 62 CRC cases with deficient MLH1 (dMLH1) and BRAFV600E wild-type (wt) status from a consecutive real-life series of 2079 CRCs. First, gene fusions were analysed using a novel FusionPlex Lung v2 RNA-based next-generation sequencing (NGS) panel, and these results were compared to a novel Idylla GeneFusion assay and pan-TRK immunohistochemistry (IHC). NGS detected seven (7/62, 11%) NTRK1 fusions (TPM3::NTRK1, PLEKHA6::NTRK1 and LMNA::NTRK1, each in two cases, and IRF2BP2::NTRK1 in one case). In addition, two ALK, four RET and seven BRAF fusions were identified. Idylla detected seven NTRK1 expression imbalances, in line with the NGS results (overall agreement 100%). Furthermore, Idylla detected the two NGS-identified ALK rearrangements as one specific ALK fusion and one ALK expression imbalance, whilst only two of the four RET fusions were discovered. However, Idylla detected several expression imbalances of ALK (n = 7) and RET (n = 1) that were found to be fusion negative with the NGS. Pan-TRK IHC showed clearly detectable, fusion partner-dependent staining patterns in the seven NTRK1 fusion cases. Overall agreement for pan-TRK antibody clone EPR17341 was 98% and for A7H6R 100% when compared to the NGS. Of the 62 CRCs, 43 were MLH1 promoter hypermethylated (MLH1ph) and 39 were RASwt. All fusion cases were both MLH1ph and RASwt. Our results show that kinase fusions (20/30, 67%) and most importantly targetable NTRK1 fusions (7/30, 23%) are frequent in CRCs with dMLH1/BRAFV600Ewt/MLH1ph/RASwt. NGS was the most comprehensive method in finding the fusions, of which a subset can be screened by Idylla or IHC, provided that the result is confirmed by NGS.
Collapse
Affiliation(s)
- Iiris Ukkola
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, P.O. Box 400, 00029, HUS, Helsinki, Finland
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Pirjo Nummela
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Mia Kero
- Department of Pathology, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, P.O. Box 400, 00029, HUS, Helsinki, Finland
| | - Hanna Tammio
- Department of Genetics, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and University of Helsinki, Helsinki, Finland
| | - Jenni Tuominen
- Department of Genomics, Laboratory of Molecular Haematology and Pathology, Turku University Central Hospital, Turku, Finland
| | - Veli Kairisto
- Department of Genomics, Laboratory of Molecular Haematology and Pathology, Turku University Central Hospital, Turku, Finland
| | - Markku Kallajoki
- Department of Pathology, University of Turku and Turku University Hospital, Turku, Finland
| | - Caj Haglund
- Department of Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Translational Cancer Medicine Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Päivi Peltomäki
- Department of Medical and Clinical Genetics, University of Helsinki, Helsinki, Finland
| | - Soili Kytölä
- Department of Genetics, HUSLAB, HUS Diagnostic Center, Helsinki University Hospital and 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, 00029, HUS, Helsinki, Finland.
- Applied Tumor Genomics Research Program, Research Programs Unit, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
49
|
Assessment of a cancer genomic profile test for patients with metastatic breast cancer. Sci Rep 2022; 12:4813. [PMID: 35315838 PMCID: PMC8938506 DOI: 10.1038/s41598-022-08925-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/15/2022] [Indexed: 11/25/2022] Open
Abstract
Comprehensive cancer genomic profile (CGP) tests are being implemented under Japanese universal health insurance system. However, the clinical usefulness of CGP test for breast cancer patients has not been evaluated. Of the 310 patients who underwent CGP testing at our institution between November 2019 and April 2021, 35 patients with metastatic breast cancer whose treatment strategy was discussed by our molecular tumor board within the study period were investigated after exclusion of 2 cases that could not be analyzed. The turn-around time, drug accessibility, and germline identification detection were evaluated. The subtype was luminal in 20 patients (57.1%), triple-negative in 12 patients (34.3%), and luminal-HER2 in 3 patients (8.6%). Actionable gene mutations were detected in 30 patients (85.7%), and 7 patients (20.0%) were recommended for clinical trial participation, with the drug administered to 2 patients (5.7%). Three patients (8.6%) died due to disease progression before the test results were disclosed. We report the results of an initial assessment of the utility of CGP testing for patients with metastatic breast cancer under Japanese universal health insurance system. Conducting CGP tests at a more appropriate time could provide patients with greater benefit from treatments based on their specific gene mutations.
Collapse
|
50
|
Koopman B, Kuijpers CCHJ, Groen HJM, Timens W, Schuuring E, Willems SM, van Kempen LC. Detection of NTRK Fusions and TRK Expression and Performance of pan-TRK Immunohistochemistry in Routine Diagnostics: Results from a Nationwide Community-Based Cohort. Diagnostics (Basel) 2022; 12:diagnostics12030668. [PMID: 35328221 PMCID: PMC8946871 DOI: 10.3390/diagnostics12030668] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/02/2022] [Accepted: 03/04/2022] [Indexed: 02/01/2023] Open
Abstract
Gene fusions involving NTRK1, NTRK2, and NTRK3 are rare drivers of cancer that can be targeted with histology-agnostic inhibitors. This study aimed to determine the nationwide landscape of NTRK/TRK testing in the Netherlands and the usage of pan-TRK immunohistochemistry (IHC) as a preselection tool to detect NTRK fusions. All pathology reports in 2017–2020 containing the search term ‘TRK’ were retrieved from the Dutch Pathology Registry (PALGA). Patient characteristics, tumor histology, NTRK/TRK testing methods, and reported results were extracted. NTRK/TRK testing was reported for 7457 tumors. Absolute testing rates increased from 815 (2017) to 3380 (2020). Tumors were tested with DNA/RNA-based molecular assay(s) (48%), IHC (47%), or in combination (5%). A total of 69 fusions involving NTRK1 (n = 22), NTRK2 (n = 6) and NTRK3 (n = 41) were identified in tumors from adult (n = 51) and pediatric (n = 18) patients. In patients tested with both IHC and a molecular assay (n = 327, of which 29 NTRK fusion-positive), pan-TRK IHC had a sensitivity of 77% (95% confidence interval (CI), 56–91) and a specificity of 84% (95% CI, 78–88%). These results showed that pan-TRK IHC has a low sensitivity in current routine practice and warrants the introduction of quality guidelines regarding the implementation and interpretation of pan-TRK IHC.
Collapse
Affiliation(s)
- Bart Koopman
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (B.K.); (W.T.); (E.S.); (S.M.W.)
| | | | - Harry J. M. Groen
- Department of Pulmonary Diseases, University of Groningen, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands;
| | - Wim Timens
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (B.K.); (W.T.); (E.S.); (S.M.W.)
| | - Ed Schuuring
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (B.K.); (W.T.); (E.S.); (S.M.W.)
| | - Stefan M. Willems
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (B.K.); (W.T.); (E.S.); (S.M.W.)
| | - Léon C. van Kempen
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, P.O. Box 30.001, 9700 RB Groningen, The Netherlands; (B.K.); (W.T.); (E.S.); (S.M.W.)
- Correspondence: ; Tel.: +31-50-361-5129
| |
Collapse
|