51
|
Momeni-Boroujeni A, Mullaney K, DiNapoli SE, Leitao MM, Hensley ML, Katabi N, Allison DHR, Park KJ, Antonescu CR, Chiang S. Expanding the Spectrum of NR4A3 Fusion-Positive Gynecologic Leiomyosarcomas. Mod Pathol 2024; 37:100474. [PMID: 38508521 DOI: 10.1016/j.modpat.2024.100474] [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: 01/25/2024] [Revised: 03/02/2024] [Accepted: 03/10/2024] [Indexed: 03/22/2024]
Abstract
Recurrent gene fusions have been observed in epithelioid and myxoid variants of uterine leiomyosarcoma. PGR::NR4A3 fusions were recently described in a subset of epithelioid leiomyosarcomas exhibiting rhabdoid morphology. In this study, we sought to expand the clinical, morphologic, immunohistochemical, and genetic features of gynecologic leiomyosarcomas harboring NR4A3 rearrangements with PGR and novel fusion partners. We identified 9 gynecologic leiomyosarcomas harboring PGR::NR4A3, CARMN::NR4A3, ACTB::NR4A3, and possible SLCO5A1::NR4A3 fusions by targeted RNA sequencing. Tumors frequently affected premenopausal women, involving the uterine corpus, uterine cervix, or pelvis. All were similarly characterized by lobules of monomorphic epithelioid and/or spindled cells arranged in sheets, cords, trabeculae, and micro- and macrocysts associated with abundant myxoid matrix and hemorrhage, creating labyrinth-like or pulmonary edema-like architecture. Myogenic differentiation with frequent estrogen receptor and progesterone receptor staining and no CD10 expression characterized all tumors. All cases showed high NR4A3 RNA expression levels and NOR1 (NR4A3) nuclear staining similar to salivary gland acinic cell carcinomas and a subset of extraskeletal myxoid chondrosarcomas harboring NR4A3 rearrangements. NOR1 (NR4A3) immunohistochemistry may serve as a useful diagnostic marker of NR4A3 fusion-positive gynecologic leiomyosarcomas.
Collapse
Affiliation(s)
- Amir Momeni-Boroujeni
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kerry Mullaney
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sara E DiNapoli
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Mario M Leitao
- Department of Surgery, Gynecologic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Martee L Hensley
- Department of Medicine, Gynecologic Medical Oncology Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nora Katabi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Douglas H R Allison
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kay J Park
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Cristina R Antonescu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sarah Chiang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|
52
|
Conde E, Hernandez S, Rodriguez Carrillo JL, Martinez R, Alonso M, Curto D, Jimenez B, Caminoa A, Benito A, Garrido P, Clave S, Arriola E, Esteban-Rodriguez I, De Castro J, Sansano I, Felip E, Rojo F, Dómine M, Abdulkader I, Garcia-Gonzalez J, Teixido C, Reguart N, Compañ D, Insa A, Mancheño N, Palanca S, Juan-Vidal O, Baixeras N, Nadal E, Cebollero M, Calles A, Martin P, Salas C, Provencio M, Aranda I, Massuti B, Lopez-Vilaro L, Majem M, Paz-Ares L, Lopez-Rios F. RET Fusion Testing in Patients With NSCLC: The RETING Study. JTO Clin Res Rep 2024; 5:100653. [PMID: 38525319 PMCID: PMC10957499 DOI: 10.1016/j.jtocrr.2024.100653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 01/31/2024] [Accepted: 02/18/2024] [Indexed: 03/26/2024] Open
Abstract
Introduction RET inhibitors with impressive overall response rates are now available for patients with NSCLC, yet the identification of RET fusions remains a difficult challenge. Most guidelines encourage the upfront use of next-generation sequencing (NGS), or alternatively, fluorescence in situ hybridization (FISH) or reverse transcriptase-polymerase chain reaction (RT-PCR) when NGS is not possible or available. Taken together, the suboptimal performance of single-analyte assays to detect RET fusions, although consistent with the notion of encouraging universal NGS, is currently widening some of the clinical practice gaps in the implementation of predictive biomarkers in patients with advanced NSCLC. Methods This situation prompted us to evaluate several RET assays in a large multicenter cohort of RET fusion-positive NSCLC (n = 38) to obtain real-world data. In addition to RNA-based NGS (the criterion standard method), all positive specimens underwent break-apart RET FISH with two different assays and were also tested by an RT-PCR assay. Results The most common RET partners were KIF5B (78.9%), followed by CCDC6 (15.8%). The two RET NGS-positive but FISH-negative samples contained a KIF5B(15)-RET(12) fusion. The three RET fusions not identified with RT-PCR were AKAP13(35)-RET(12), KIF5B(24)-RET(9) and KIF5B(24)-RET(11). All three false-negative RT-PCR cases were FISH-positive, exhibited a typical break-apart pattern, and contained a very high number of positive tumor cells with both FISH assays. Signet ring cells, psammoma bodies, and pleomorphic features were frequently observed (in 34.2%, 39.5%, and 39.5% of tumors, respectively). Conclusions In-depth knowledge of the advantages and disadvantages of the different RET testing methodologies could help clinical and molecular tumor boards implement and maintain sensible algorithms for the rapid and effective detection of RET fusions in patients with NSCLC. The likelihood of RET false-negative results with both FISH and RT-PCR reinforces the need for upfront NGS in patients with NSCLC.
Collapse
Affiliation(s)
- Esther Conde
- Hospital Universitario 12 de Octubre, Madrid, Spain
- Universidad Complutense, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
| | - Susana Hernandez
- Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | | | | | - Marta Alonso
- Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Daniel Curto
- Hospital Universitario 12 de Octubre, Madrid, Spain
| | | | | | | | - Pilar Garrido
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Sergi Clave
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Hospital del Mar, Barcelona, Spain
| | - Edurne Arriola
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Hospital del Mar, Barcelona, Spain
| | | | - Javier De Castro
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Hospital Universitario La Paz, Madrid, Spain
- Instituto de Investigacion Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain
| | - Irene Sansano
- Hospital Universitario Vall d'Hebron, Barcelona, Spain
| | | | - Federico Rojo
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Instituto de Investigacion Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Madrid, Spain
| | - Manuel Dómine
- Instituto de Investigacion Sanitaria-Fundacion Jimenez Diaz (IIS-FJD), Madrid, Spain
- Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain
| | - Ihab Abdulkader
- Hospital Clinico Universitario de Santiago, Santiago de Compostela, Spain
| | | | - Cristina Teixido
- Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | - Noemi Reguart
- Hospital Clinic, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, Barcelona, Spain
| | | | - Amelia Insa
- Hospital Clinico Universitario, Valencia, Spain
| | - Nuria Mancheño
- Hospital Universitario y Politecnico La Fe, Valencia, Spain
| | - Sarai Palanca
- Hospital Universitario y Politecnico La Fe, Valencia, Spain
| | | | - Nuria Baixeras
- Hospital Universitari de Bellvitge, L’Hospitalet, Barcelona, Spain
| | - Ernest Nadal
- Catalan Institute of Oncology, L’Hospitalet, Barcelona, Spain
| | - Maria Cebollero
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Antonio Calles
- Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Paloma Martin
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Universitario Puerta de Hierro, Madrid, Spain
| | - Clara Salas
- Hospital Universitario Puerta de Hierro, Madrid, Spain
| | | | - Ignacio Aranda
- Hospital General Universitario Dr. Balmis – Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | - Bartomeu Massuti
- Hospital General Universitario Dr. Balmis – Instituto de Investigación Sanitaria y Biomédica de Alicante (ISABIAL), Alicante, Spain
| | | | | | - Luis Paz-Ares
- Hospital Universitario 12 de Octubre, Madrid, Spain
- Universidad Complutense, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
| | - Fernando Lopez-Rios
- Hospital Universitario 12 de Octubre, Madrid, Spain
- Universidad Complutense, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
- Centro de Investigación Biomedica en Red Cancer (CIBERONC), Madrid, Spain
| |
Collapse
|
53
|
Li S, Zhang H, Chen T, Zhang X, Shang G. Current treatment and novel insights regarding ROS1-targeted therapy in malignant tumors. Cancer Med 2024; 13:e7201. [PMID: 38629293 PMCID: PMC11022151 DOI: 10.1002/cam4.7201] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Revised: 03/22/2024] [Accepted: 04/06/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND The proto-oncogene ROS1 encodes an intrinsic type I membrane protein of the tyrosine kinase/insulin receptor family. ROS1 facilitates the progression of various malignancies via self-mutations or rearrangements. Studies on ROS1-directed tyrosine kinase inhibitors have been conducted, and some have been approved by the FDA for clinical use. However, the adverse effects and mechanisms of resistance associated with ROS1 inhibitors remain unknown. In addition, next-generation ROS1 inhibitors, which have the advantage of treating central nervous system metastases and alleviating endogenous drug resistance, are still in the clinical trial stage. METHOD In this study, we searched relevant articles reporting the mechanism and clinical application of ROS1 in recent years; systematically reviewed the biological mechanisms, diagnostic methods, and research progress on ROS1 inhibitors; and provided perspectives for the future of ROS1-targeted therapy. RESULTS ROS1 is most expressed in malignant tumours. Only a few ROS1 kinase inhibitors are currently approved for use in NSCLC, the efficacy of other TKIs for NSCLC and other malignancies has not been ascertained. There is no effective standard treatment for adverse events or resistance to ROS1-targeted therapy. Next-generation TKIs appear capable of overcoming resistance and delaying central nervous system metastasis, but with a greater incidence of adverse effects. CONCLUSIONS Further research on next-generation TKIs regarding the localization of ROS1 and its fusion partners, binding sites for targeted drugs, and coadministration with other drugs is required. The correlation between TKIs and chemotherapy or immunotherapy in clinical practice requires further study.
Collapse
Affiliation(s)
- Shizhe Li
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - He Zhang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Ting Chen
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Xiaowen Zhang
- Medical Research CenterShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| | - Guanning Shang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoning ProvinceChina
| |
Collapse
|
54
|
Pelster MS, Silverman IM, Schonhoft JD, Johnson A, Selenica P, Ulanet D, Rimkunas V, Reis-Filho JS. Post-therapy emergence of an NBN reversion mutation in a patient with pancreatic acinar cell carcinoma. NPJ Precis Oncol 2024; 8:82. [PMID: 38561473 PMCID: PMC10985087 DOI: 10.1038/s41698-024-00497-x] [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: 03/06/2023] [Accepted: 12/21/2023] [Indexed: 04/04/2024] Open
Abstract
Pancreatic acinar cell carcinoma (PACC) is a rare form of pancreatic cancer that commonly harbors targetable alterations, including activating fusions in the MAPK pathway and loss-of-function (LOF) alterations in DNA damage response/homologous recombination DNA repair-related genes. Here, we describe a patient with PACC harboring both somatic biallelic LOF of NBN and an activating NTRK1 fusion. Upon disease progression following 13 months of treatment with folinic acid, fluorouracil, irinotecan, and oxaliplatin (FOLFIRINOX), genomic analysis of a metastatic liver biopsy revealed the emergence of a novel reversion mutation restoring the reading frame of NBN. To our knowledge, genomic reversion of NBN has not been previously reported as a resistance mechanism in any tumor type. The patient was treated with, but did not respond to, targeted treatment with a selective NTRK inhibitor. This case highlights the complex but highly actionable genomic landscape of PACC and underlines the value of genomic profiling of rare tumor types such as PACC.
Collapse
Affiliation(s)
| | | | | | | | - Pier Selenica
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | - Jorge S Reis-Filho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
55
|
de Traux de Wardin H, Dermawan JK, Vanoli F, Jiang SC, Singer S, Chi P, Tap W, Wexler LH, Antonescu CR. NF1-Driven Rhabdomyosarcoma Phenotypes: A Comparative Clinical and Molecular Study of NF1-Mutant Rhabdomyosarcoma and NF1-Associated Malignant Triton Tumor. JCO Precis Oncol 2024; 8:e2300597. [PMID: 38603649 PMCID: PMC11161258 DOI: 10.1200/po.23.00597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/28/2024] [Accepted: 02/16/2024] [Indexed: 04/13/2024] Open
Abstract
PURPOSE Alterations of the NF1 tumor suppressor gene is the second most frequent genetic event in embryonal rhabdomyosarcoma (ERMS), but its associations with clinicopathologic features, outcome, or coexisting molecular events are not well defined. Additionally, NF1 alterations, mostly in the setting of neurofibromatosis type I (NF1), drive the pathogenesis of most malignant peripheral nerve sheath tumor with divergent RMS differentiation (also known as malignant triton tumor [MTT]). Distinguishing between these entities can be challenging because of their pathologic overlap. This study aims to comprehensively analyze the clinicopathologic and molecular spectrum of NF1-mutant RMS compared with NF1-associated MTT for a better understanding of their pathogenesis. METHODS We investigated the clinicopathologic and molecular landscape of a cohort of 22 NF1-mutant RMS and a control group of 13 NF1-associated MTT. Cases were tested on a matched tumor-normal hybridization capture-based targeted DNA next-generation sequencing. RESULTS Among the RMS group, all except one were ERMS, with a median age of 17 years while for MTT the mean age was 39 years. Three MTTs were misdiagnosed as ERMS, having clinical impact in one. The most frequent coexisting alteration in ERMS was TP53 abnormality (36%), being mutually exclusive from NRAS mutations (14%). MTT showed coexisting CDKN2A/B and PRC2 complex alterations in 38% cases and loss of H3K27me3 expression. Patients with NF1-mutant RMS exhibited a 70% 5-year survival rate, in contrast to MTT with a 33% 5-year survival. All metastatic NF1-mutant ERMS were associated with TP53 alterations. CONCLUSION Patients with NF1-mutant ERMS lacking TP53 alterations may benefit from dose-reduction chemotherapy. On the basis of the diagnostic challenges and significant treatment and prognostic differences, molecular profiling of challenging tumors with rhabdomyoblastic differentiation is recommended.
Collapse
Affiliation(s)
- Henry de Traux de Wardin
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Pediatrics, Brussels University Hospital, Academic Children's Hospital Queen Fabiola, Université Libre de Bruxelles, Brussels, Belgium
| | - Josephine K. Dermawan
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, OH
| | - Fabio Vanoli
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Samuel C. Jiang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Samuel Singer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Ping Chi
- Department of Medicine, Sarcoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY
| | - William Tap
- Department of Medicine, Sarcoma Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Leonard H. Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Cristina R. Antonescu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
56
|
Chu YH, Mullaney K, DiNapoli SE, Cohen MA, Xu B, Ghossein R, Katabi N, Dogan S. FGFR1/2/3-rearranged carcinoma of the head and neck: expanded histological spectrum crossing path with high-risk HPV in the sinonasal tract. Histopathology 2024; 84:589-600. [PMID: 38010295 PMCID: PMC10872948 DOI: 10.1111/his.15099] [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: 09/20/2023] [Revised: 10/27/2023] [Accepted: 11/02/2023] [Indexed: 11/29/2023]
Abstract
AIMS Oncogenic FGFR1/2/3 rearrangements are found in various cancers. Reported cases in head and neck (HN) are mainly squamous cell carcinomas (SCCs) with FGFR3::TACC3 fusions, a subset of which also harbour high-risk human papillomavirus (HPV). However, the knowledge of the clinicopathological spectrum of FGFR-rearranged head and neck carcinomas (FHNC) is limited. METHODS AND RESULTS A retrospective MSK-fusion clinical sequencing cohort 2016-23 was searched to identify malignant tumours in the HN region harbouring FGFR1/2/3 fusion. FHNC were characterised by histological examination, immunohistochemistry and molecular analysis. Electronic medical records were reviewed. Three FHNC were identified. Two cases (cases 1 and 2) involved sinonasal tract and were high-grade carcinomas with squamous, basaloid, glandular and/or ductal-myoepithelial features. Case 1 arose in a 79-year-old man and harboured FGFR2::KIF1A fusion. Case 2 arose in a 58-year-old man, appeared as HPV-related multiphenotypic sinonasal carcinoma (HMSC), and was positive for FGFR2::TACC2 fusion and concurrent high-risk HPV, non-type 16/18. Case 3 was FGFR3::TACC3 fusion-positive keratinising SCCs arising in the parotid of a 60-year-old man. All three cases presented at stage T4. Clinical follow-up was available in two cases; case 1 remained disease-free for 41 months post-treatment and case 3 died of disease 2 months after the diagnosis. CONCLUSIONS FHNC include a morphological spectrum of carcinomas with squamous features and may occur in different HN locations, such as parotid gland and the sinonasal tract. Sinonasal cases can harbour FGFR2 rearrangement with or without associated high-risk HPV. Timely recognition of FHNC could help select patients potentially amenable to targeted therapy with FGFR inhibitors. Further studies are needed (1) to determine if FGFR2 rearranged/HPV-positive sinonasal carcinomas are biologically distinct from HMSC, and (2) to elucidate the biological and clinical significance of FGFR2 rearrangement in the context of high-risk HPV.
Collapse
Affiliation(s)
- Ying-Hsia Chu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Kerry Mullaney
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Sara E. DiNapoli
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Marc A. Cohen
- Department of Surgery, Head and Neck Service, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Bin Xu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Ronald Ghossein
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Nora Katabi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| | - Snjezana Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, United States
| |
Collapse
|
57
|
Hasegawa N, Hayashi T, Niizuma H, Kikuta K, Imanishi J, Endo M, Ikeuchi H, Sasa K, Sano K, Hirabayashi K, Takagi T, Ishijima M, Kato S, Kohsaka S, Saito T, Suehara Y. Detection of Novel Tyrosine Kinase Fusion Genes as Potential Therapeutic Targets in Bone and Soft Tissue Sarcomas Using DNA/RNA-based Clinical Sequencing. Clin Orthop Relat Res 2024; 482:549-563. [PMID: 38014853 PMCID: PMC10871756 DOI: 10.1097/corr.0000000000002901] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 11/15/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND Approximately 1% of clinically treatable tyrosine kinase fusions, including anaplastic lymphoma kinase, neurotrophic tyrosine receptor kinase, RET proto-oncogene, and ROS proto-oncogene 1, have been identified in soft tissue sarcomas via comprehensive genome profiling based on DNA sequencing. Histologic tumor-specific fusion genes have been reported in approximately 20% of soft tissue sarcomas; however, unlike tyrosine kinase fusion genes, these fusions cannot be directly targeted in therapy. Approximately 80% of tumor-specific fusion-negative sarcomas, including myxofibrosarcoma and leiomyosarcoma, that are defined in complex karyotype sarcomas remain genetically uncharacterized; this mutually exclusive pattern of mutations suggests that other mutually exclusive driver oncogenes are yet to be discovered. Tumor-specific, fusion-negative sarcomas may be associated with unique translocations, and oncogenic fusion genes, including tyrosine kinase fusions, may have been overlooked in these sarcomas. QUESTIONS/PURPOSES (1) Can DNA- or RNA-based analysis reveal any characteristic gene alterations in bone and soft tissue sarcomas? (2) Can useful and potential tyrosine kinase fusions in tumors from tumor-specific, fusion-negative sarcomas be detected using an RNA-based screening system? (3) Do the identified potential fusion tumors, especially in neurotrophic tyrosine receptor kinase gene fusions in bone sarcoma, transform cells and respond to targeted drug treatment in in vitro assays? (4) Can the identified tyrosine kinase fusion genes in sarcomas be useful therapeutic targets? METHODS Between 2017 and 2020, we treated 100 patients for bone and soft tissue sarcomas at five institutions. Any biopsy or surgery from which a specimen could be obtained was included as potentially eligible. Ninety percent (90 patients) of patients were eligible; a further 8% (8 patients) were excluded because they were either lost to follow-up or their diagnosis was changed, leaving 82% (82 patients) for analysis here. To answer our first and second questions regarding gene alterations and potential tyrosine kinase fusions in eight bone and 74 soft tissue sarcomas, we used the TruSight Tumor 170 assay to detect mutations, copy number variations, and gene fusions in the samples. To answer our third question, we performed functional analyses involving in vitro assays to determine whether the identified tyrosine kinase fusions were associated with oncogenic abilities and drug responses. Finally, to determine usefulness as therapeutic targets, two pediatric patients harboring an NTRK fusion and an ALK fusion were treated with tyrosine kinase inhibitors in clinical trials. RESULTS DNA/RNA-based analysis demonstrated characteristic alterations in bone and soft tissue sarcomas; DNA-based analyses detected TP53 and copy number alterations of MDM2 and CDK4 . These single-nucleotide variants and copy number variations were enriched in specific fusion-negative sarcomas. RNA-based screening detected fusion genes in 24% (20 of 82) of patients. Useful potential fusions were detected in 19% (11 of 58) of tumor-specific fusion-negative sarcomas, with nine of these patients harboring tyrosine kinase fusion genes; five of these patients had in-frame tyrosine kinase fusion genes ( STRN3-NTRK3, VWC2-EGFR, ICK-KDR, FOXP2-MET , and CEP290-MET ) with unknown pathologic significance. The functional analysis revealed that STRN3-NTRK3 rearrangement that was identified in bone had a strong transforming potential in 3T3 cells, and that STRN3-NTRK3 -positive cells were sensitive to larotrectinib in vitro. To confirm the usefulness of identified tyrosine kinase fusion genes as therapeutic targets, patients with well-characterized LMNA-NTRK1 and CLTC-ALK fusions were treated with tyrosine kinase inhibitors in clinical trials, and a complete response was achieved. CONCLUSION We identified useful potential therapeutic targets for tyrosine kinase fusions in bone and soft tissue sarcomas using RNA-based analysis. We successfully identified STRN3-NTRK3 fusion in a patient with leiomyosarcoma of bone and determined the malignant potential of this fusion gene via functional analyses and drug effects. In light of these discoveries, comprehensive genome profiling should be considered even if the sarcoma is a bone sarcoma. There seem to be some limitations regarding current DNA-based comprehensive genome profiling tests, and it is important to use RNA testing for proper diagnosis and accurate identification of fusion genes. Studies on more patients, validation of results, and further functional analysis of unknown tyrosine kinase fusion genes are required to establish future treatments. CLINICAL RELEVANCE DNA- and RNA-based screening systems may be useful for detecting tyrosine kinase fusion genes in specific fusion-negative sarcomas and identifying key therapeutic targets, leading to possible breakthroughs in the treatment of bone and soft tissue sarcomas. Given that current DNA sequencing misses fusion genes, RNA-based screening systems should be widely considered as a worldwide test for sarcoma. If standard treatments such as chemotherapy are not effective, or even if the sarcoma is of bone, RNA sequencing should be considered to identify as many therapeutic targets as possible.
Collapse
Affiliation(s)
- Nobuhiko Hasegawa
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Takuo Hayashi
- Department of Human Pathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Hidetaka Niizuma
- Department of Pediatrics, Tohoku University School of Medicine, Miyagi, Japan
| | - Kazutaka Kikuta
- Division of Musculoskeletal Oncology and Orthopaedic Surgery, Tochigi Cancer Center, Tochigi, Japan
| | - Jungo Imanishi
- Department of Orthopaedic Surgery, Teikyo University School of Medicine, Tokyo, Japan
- Department of Orthopaedic Oncology and Surgery, Saitama Medical University International Medical Center, Saitama, Japan
| | - Makoto Endo
- Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroshi Ikeuchi
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Keita Sasa
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kei Sano
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Kaoru Hirabayashi
- Division of Diagnostic Pathology, Tochigi Cancer Center, Tochigi, Japan
| | - Tatsuya Takagi
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Muneaki Ishijima
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shunsuke Kato
- Department of Clinical Oncology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Tsuyoshi Saito
- Department of Human Pathology, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Yoshiyuki Suehara
- Department of Medicine for Orthopaedics and Motor Organ, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
- Intractable Disease Research Center, Juntendo University Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
58
|
Murray NP. Biomarkers of minimal residual disease and treatment. Adv Clin Chem 2024; 119:33-70. [PMID: 38514211 DOI: 10.1016/bs.acc.2024.02.002] [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] [Indexed: 03/23/2024]
Abstract
Minimal residual disease (MRD) has been defined as a very small numbers of cancer cells that remain in the body after curative treatment. Its presence or absence will ultimately determine prognosis. With the introduction of new technologies the presence of MRD in patients with solid tumours can be detected and characterized. As MRD predicts future relapse, be it early or late treatment failure, in an otherwise asymptomatic patient its treatment and when to start treatment remains to be determined. Thus the concepts of personalized medicine using different biomarkers to classify the biological properties of MRD maybe come possible. Based on this determinations it may be possible to use targeted therapies rather than all patients with the same type of cancer receiving a standard treatment. However, it is important to understand the limitations of the different technologies, what these techniques are detecting and how they may help in the treatment of patients with cancer. The majority of published studies are in patients with metastatic cancer and there are few reports in patients with MRD. In this chapter the concept of MRD, the methods used to detect it and what treatments may be effective based on the biological characteristics of the tumour cells as determined by different biomarkers is reviewed. MRD depends on the phenotypic properties of the tumour cells to survive in their new environment and the anti-tumour immune response. This is a dynamic process and changes with time in the wake of immunosuppression caused by the tumour cells and/or the effects of treatment to select resistant tumour cells. With the use of biomarkers to typify the characteristics of MRD and the development of new drugs a personalized treatment can be designed rather than all patients given the same treatment. Patients who are initially negative for MRD may not require further treatment with liquid biopsies used to monitor the patients during follow-up in order to detect those patients who may become MRD positive. The liquid biopsy used during the follow up of MRD positive patients can be used to detect changes in the biological properties of the tumour cells and thus may need treatment changes to overcome tumour cell resistance.
Collapse
Affiliation(s)
- Nigel P Murray
- Minimal Residual Disease Laboratory, Faculty of Medicine, University Finis Terrae, Santiago, Chile.
| |
Collapse
|
59
|
Kim DW, Schram AM, Hollebecque A, Nishino K, Macarulla T, Rha SY, Duruisseaux M, Liu SV, Al Hallak MN, Umemoto K, Wesseler C, Cleary JM, Springfeld C, Neuzillet C, Joe A, Jauhari S, Ford J, Goto K. The phase I/II eNRGy trial: Zenocutuzumab in patients with cancers harboring NRG1 gene fusions. Future Oncol 2024; 20:1057-1067. [PMID: 38348690 PMCID: PMC11721935 DOI: 10.2217/fon-2023-0824] [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: 09/22/2023] [Accepted: 01/04/2024] [Indexed: 06/12/2024] Open
Abstract
Neuregulin 1 (NRG1) fusions are oncogenic drivers that have been detected in non-small-cell lung cancer (NSCLC), pancreatic ductal adenocarcinoma (PDAC) and other solid tumors. NRG1 fusions are rare, occurring in less than 1% of solid tumors. Patients with NRG1 fusion positive (NRG1+) cancer have limited therapeutic options. Zenocutuzumab is a novel, bispecific IgG1 antibody that targets both HER2 and HER3 proteins and inhibits NRG1 binding through a 'Dock & Block®' mechanism of action. Here, we describe the rationale and design of the phase II component of the eNRGy trial, part of the overall, open-label phase I/II, multicenter trial exploring the safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity and antitumor activity of zenocutuzumab in patients with NRG1+ NSCLC, PDAC or other solid tumors.
Collapse
MESH Headings
- Adult
- Female
- Humans
- Male
- Middle Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Antineoplastic Agents, Immunological/therapeutic use
- Antineoplastic Agents, Immunological/adverse effects
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/genetics
- Carcinoma, Non-Small-Cell Lung/pathology
- Carcinoma, Pancreatic Ductal/genetics
- Carcinoma, Pancreatic Ductal/drug therapy
- Carcinoma, Pancreatic Ductal/pathology
- Lung Neoplasms/drug therapy
- Lung Neoplasms/genetics
- Lung Neoplasms/pathology
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neuregulin-1/genetics
- Oncogene Proteins, Fusion/genetics
- Receptor, ErbB-2/genetics
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/metabolism
- Receptor, ErbB-3/genetics
- Clinical Trials, Phase I as Topic
- Clinical Trials, Phase II as Topic
- Multicenter Studies as Topic
Collapse
Affiliation(s)
- Dong-Wan Kim
- Department of Internal Medicine, Seoul National University College of Medicine & Seoul National University Hospital, Seoul, 03080, Republic of Korea
| | - Alison M Schram
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Antoine Hollebecque
- Drug Development (DITEP), GI Oncology, Gustave Roussy Cancer Campus, Villejuif, 94805, France
| | - Kazumi Nishino
- Department of Thoracic Oncology, Osaka International Cancer Institute, Osaka, 540-0008, Japan
| | - Teresa Macarulla
- Gastrointestinal and Endocrine Tumor Unit, Vall d´Hebrón University Hospital, Vall d´Hebrón Institute of Oncology (VHIO), Barcelona, 08035, Spain
| | - Sun Young Rha
- Department of Internal Medicine, Yonsei Cancer Center, Yonsei University Health System, Seoul, 03722, Republic of Korea
| | - Michaël Duruisseaux
- Department of Respiratory Medicine and Early Phase, Louis Pradel Hospital, Hospices Civils de Lyon Cancer Institute, Lyon, 69500, France
- Cancer Research Centre of Lyon, UMR INSERM 1052 CNRS 5286, Lyon, 69008, France
- Claude Bernard University Lyon 1, University of Lyon, Villeurbanne, 69100, France
| | - Stephen V Liu
- Thoracic Oncology and Developmental Therapeutics, Lombardi Comprehensive Cancer Center, Georgetown University, WA 20007, USA
| | - Mohammed Najeeb Al Hallak
- Department of Oncology, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Kumiko Umemoto
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, 216-8511, Japan
| | - Claas Wesseler
- Department of Pulmonology, Asklepios Tumorzentrum Hamburg, Klinikum Harburg, Hamburg, 21075, Germany
| | - James M Cleary
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Harvard Medical School, Boston, MA 02215, USA
| | - Christoph Springfeld
- Department of Medical Oncology, Heidelberg University Hospital, Department of Medical Oncology, Heidelberg, 69120, Germany
| | - Cindy Neuzillet
- GI Oncology, Medical Oncology Department, Curie Institute, Versailles-Saint Quentin University, Saint-Cloud, 92064, France
| | - Andrew Joe
- Clinical Development, Merus NV, Utrecht, 3584, The Netherlands
| | - Shekeab Jauhari
- Clinical Development, Merus NV, Utrecht, 3584, The Netherlands
| | - Jim Ford
- Clinical Trials, Merus NV, Utrecht, 3584, The Netherlands
| | - Koichi Goto
- Department of Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, 277-8577, Japan
| |
Collapse
|
60
|
Zhang M, Jiang L, Liu XY, Liu FX, Zhang H, Zhang YJ, Tang XM, Ma YS, Wu HY, Diao X, Yang C, Liu JB, Fu D, Zhang J, Yu H. KLK10/LIPH/PARD6B/SLC52A3 are promising molecular biomarkers for the prognosis of pancreatic cancer through a ceRNA network. Heliyon 2024; 10:e24287. [PMID: 38234923 PMCID: PMC10792648 DOI: 10.1016/j.heliyon.2024.e24287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 11/30/2023] [Accepted: 01/05/2024] [Indexed: 01/19/2024] Open
Abstract
Pancreatic adenocarcinoma (PAAD) remains challenging to diagnose and treat clinically due to its difficult early diagnosis, low surgical resection rate, and high risk of postoperative recurrence and metastasis. SMAD4 is a classical mutated gene in pancreatic cancer and is lost in up to 60%-90 % of PAAD patients, and its mutation often predicts a poor prognosis and treatment resistance. In this study, based on the expression profile data in The Cancer Genome Atlas database, we identified a ceRNA network composed of 2 lncRNAs, 1 miRNA, and 4 mRNAs through differential expression analysis and survival prognosis analysis. Among them, high expression of KLK10/LIPH/PARD6B/SLC52A3 influenced the prognosis and overall survival of PAAD patients. We confirmed the high expression of these target genes in pancreatic tissue of pancreatic-specific SMAD4-deficient mice. In addition, immune infiltration analysis showed that the high expression of these target genes affects the tumor immune environment and contributes to the progression of PAAD. Abnormal overexpression of these target genes may be caused by hypermethylation. In conclusion, we found that KLK10/LIPH/PARD6B/SLC52A3 is a potential prognostic marker for PAAD based on a competing endogenous RNA-mediated mechanism and revealed the potential pathogenic mechanism by which deficient expression of SMAD4 promotes pancreatic cancer progression, which provides a new pathway and theoretical basis for targeted therapy or improved prognosis of pancreatic cancer. These data will help reveal potential therapeutic targets for pancreatic cancer and improve the prognosis of pancreatic cancer patients.
Collapse
Affiliation(s)
- Meng Zhang
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226019, Jiangsu, China
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, Jiangsu, China
| | - Lin Jiang
- Department of Anesthesiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, Jiangsu, 225300, China
| | - Xin-Yun Liu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Fu-Xing Liu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Hui Zhang
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, Jiangsu, China
| | - Yan-Juan Zhang
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226019, Jiangsu, China
- Department of Clinical Laboratory, Affiliated Maternity & Child Health Care Hospital of Nantong University, Nantong, 226019, Jiangsu, China
| | - Xiao-Mei Tang
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, Shanghai, China
| | - Yu-Shui Ma
- Cancer Institute, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200032, Shanghai, China
| | - Hui-Yi Wu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| | - Xun Diao
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, Jiangsu, China
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ji-Bin Liu
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, Jiangsu, China
| | - Da Fu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
- Institute of Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, 226631, Jiangsu, China
- General Surgery, Institute of Pancreatic Diseases, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200025, Shanghai, China
| | - Jie Zhang
- Department of Immunology, School of Medicine, Nantong University, Nantong, 226019, Jiangsu, China
| | - Hong Yu
- Department of Pathology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou, 225300, Jiangsu, China
- Department of Pathology, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, Jiangsu, China
| |
Collapse
|
61
|
Tjota MY, Segal JP, Wang P. Clinical Utility and Benefits of Comprehensive Genomic Profiling in Cancer. J Appl Lab Med 2024; 9:76-91. [PMID: 38167763 DOI: 10.1093/jalm/jfad091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/28/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Comprehensive genomic profiling (CGP) with next-generation sequencing detects genetic alterations of hundreds of genes simultaneously and multiple molecular biomarkers with one test. In the personalized medicine era, CGP is increasingly used for cancer diagnosis, treatment selection, and prognosis prediction. CONTENT In this review, we summarize the benefits of CGP, clinical utility of CGP, and challenges of setting up CGP in the clinical laboratories. Besides the genetic alterations identified in the cancer-related genes, other biomarkers such as tumor mutational burden, microsatellite instability, and homologous recombination deficiency are critical for initiating targeted therapy. Compared with conventional tests, CGP uses less specimen and shortens the turnaround time if multiple biomarkers need to be tested. RNA fusion assay and liquid biopsy are helpful additions to DNA-based CGP by detecting fusions/splicing variants and complementing tissue-based CGP findings, respectively. SUMMARY Many previous hurdles for implementing CGP in the clinical laboratories have been gradually alleviated such as the decrease in sequencing cost, availability of both open-source and commercial bioinformatics tools, and improved reimbursement. These changes have helped to make CGP available to a greater population of cancer patients for improving characterization of their tumors and expanding their eligibility for clinical trials. Additionally, sequencing results of the hundreds of genes on CGP panels could be further analyzed to better understand the biology of various cancers and identify new biomarkers.
Collapse
Affiliation(s)
- Melissa Yuwono Tjota
- Department of Pathology, The University of Chicago, Chicago, IL 60637, United States
| | - Jeremy P Segal
- Department of Pathology, The University of Chicago, Chicago, IL 60637, United States
| | - Peng Wang
- Department of Pathology, The University of Chicago, Chicago, IL 60637, United States
| |
Collapse
|
62
|
Solomon JP. Practical Considerations for Oncogenic Fusion Detection and Reporting in Solid Tumors. J Appl Lab Med 2024; 9:116-123. [PMID: 38167769 DOI: 10.1093/jalm/jfad068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/15/2023] [Indexed: 01/05/2024]
Abstract
BACKGROUND Chromosomal rearrangements that result in oncogenic fusions can hold tremendous clinical significance in solid tumors, often with diagnostic or treatment implications. CONTENT Traditionally, low-throughput methods such as fluorescence in situ hybridization were used to identify fusions in the clinical laboratory. With the rise of next-generation sequencing techniques and the broad adoption of comprehensive genomic profiling, the practice of screening for fusions as part of an oncologic workup has evolved. RNA sequencing methods are increasingly used, as these comprehensive high-throughput assays have many advantages over traditional techniques. Several RNA sequencing platforms are available, each with benefits and drawbacks. Regardless of the approach, systematic evaluation of the RNA sequencing results and the fusions identified by the assay should be performed. Assessment of fusion events relies upon evaluation of quality evidence, structural evidence, and functional evidence to ensure accurate fusion reporting and interpretation. SUMMARY Given the clinical significance of gene fusions in oncology, understanding the variety of assays available for fusion detection, their benefits and drawbacks, and how they are used in the identification and interpretation of gene fusions is important for the modern precision oncology practice.
Collapse
Affiliation(s)
- James P Solomon
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
63
|
Lim SM, Lee JB, Oya Y, Nutzinger J, Soo R. Path Less Traveled: Targeting Rare Driver Oncogenes in Non-Small-Cell Lung Cancer. JCO Oncol Pract 2024; 20:47-56. [PMID: 37733983 DOI: 10.1200/op.23.00273] [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] [Received: 05/02/2023] [Revised: 07/21/2023] [Accepted: 08/11/2023] [Indexed: 09/23/2023] Open
Abstract
Over the past decade, tremendous efforts have been made in the development of targeted agents in non-small-cell lung cancer (NSCLC) with nonsquamous histology. Pivotal studies have used next-generation sequencing to select the patient population harboring oncogenic driver alterations that are targetable with targeted therapies. As treatment paradigm rapidly evolves for patients with rare oncogene-driven NSCLC, updated comprehensive overview of diagnostic approach and treatment options is paramount in clinical settings. In this review article, we discuss the epidemiology, molecular testing, and landmark clinical trials addressing the targeted agents for ROS1 rearrangement, METex14 skipping mutation, EGFR exon 20 insertion, KRAS G12C mutation, HER2 mutation, RET fusion, NTRK fusion, and BRAF mutations.
Collapse
Affiliation(s)
- Sun Min Lim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Jii Bum Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, South Korea
| | - Yuko Oya
- Department of Respiratory Disease, Fujita Health University, Toyoake, Japan
| | - Jorn Nutzinger
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| | - Ross Soo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore
| |
Collapse
|
64
|
Vos JL, Burman B, Jain S, Fitzgerald CWR, Sherman EJ, Dunn LA, Fetten JV, Michel LS, Kriplani A, Ng KK, Eng J, Tchekmedyian V, Haque S, Katabi N, Kuo F, Han CY, Nadeem Z, Yang W, Makarov V, Srivastava RM, Ostrovnaya I, Prasad M, Zuur CL, Riaz N, Pfister DG, Klebanoff CA, Chan TA, Ho AL, Morris LGT. Nivolumab plus ipilimumab in advanced salivary gland cancer: a phase 2 trial. Nat Med 2023; 29:3077-3089. [PMID: 37620627 PMCID: PMC11293616 DOI: 10.1038/s41591-023-02518-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023]
Abstract
Salivary gland cancers (SGCs) are rare, aggressive cancers without effective treatments when metastasized. We conducted a phase 2 trial evaluating nivolumab (nivo, anti-PD-1) and ipilimumab (ipi, anti-CTLA-4) in 64 patients with metastatic SGC enrolled in two histology-based cohorts (32 patients each): adenoid cystic carcinoma (ACC; cohort 1) and other SGCs (cohort 2). The primary efficacy endpoint (≥4 objective responses) was met in cohort 2 (5/32, 16%) but not in cohort 1 (2/32, 6%). Treatment safety/tolerability and progression-free survival (PFS) were secondary endpoints. Treatment-related adverse events grade ≥3 occurred in 24 of 64 (38%) patients across both cohorts, and median PFS was 4.4 months (95% confidence interval (CI): 2.4, 8.3) and 2.2 months (95% CI: 1.8, 5.3) for cohorts 1 and 2, respectively. We present whole-exome, RNA and T cell receptor (TCR) sequencing data from pre-treatment and on-treatment tumors and immune cell flow cytometry and TCR sequencing from peripheral blood at serial timepoints. Responding tumors universally demonstrated clonal expansion of pre-existing T cells and mutational contraction. Responding ACCs harbored neoantigens, including fusion-derived neoepitopes, that induced T cell responses ex vivo. This study shows that nivo+ipi has limited efficacy in ACC, albeit with infrequent, exceptional responses, and that it could be promising for non-ACC SGCs, particularly salivary duct carcinomas. ClinicalTrials.gov identifier: NCT03172624 .
Collapse
Affiliation(s)
- Joris L Vos
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Bharat Burman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Swati Jain
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Conall W R Fitzgerald
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Eric J Sherman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lara A Dunn
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - James V Fetten
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Loren S Michel
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anuja Kriplani
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenneth K Ng
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Juliana Eng
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vatche Tchekmedyian
- Department of Medicine, Maine Medical Center-Tufts University School of Medicine, Portland, ME, USA
| | - Sofia Haque
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Nora Katabi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Fengshen Kuo
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Catherine Y Han
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zaineb Nadeem
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wei Yang
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Vladimir Makarov
- Center for Immunotherapy and Precision Immuno-oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Raghvendra M Srivastava
- Center for Immunotherapy and Precision Immuno-oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Irina Ostrovnaya
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Manu Prasad
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Charlotte L Zuur
- Department of Head and Neck Oncology and Surgery, The Netherlands Cancer Institute - Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
- Department of Otorhinolaryngology Head and Neck Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Nadeem Riaz
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - David G Pfister
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Christopher A Klebanoff
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Timothy A Chan
- Center for Immunotherapy and Precision Immuno-oncology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Alan L Ho
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Luc G T Morris
- Head and Neck Service and Immunogenomic Oncology Platform, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
65
|
Wang T, Askan G, Ozcan K, Rana S, Zehir A, Bhanot UK, Yantiss RK, Rao DS, Wahl SJ, Bagci P, Balci S, Balachandran V, Jarnagin WR, Adsay NV, Klimstra DS, Basturk O. Tumoral Intraductal Neoplasms of the Bile Ducts Comprise Morphologically and Genetically Distinct Entities. Arch Pathol Lab Med 2023; 147:1390-1401. [PMID: 36821179 DOI: 10.5858/arpa.2022-0343-oa] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/16/2022] [Indexed: 02/24/2023]
Abstract
CONTEXT.— Tumoral (grossly visible) intraductal neoplasms of the bile ducts are still being characterized. OBJECTIVE.— To investigate their morphologic, immunohistochemical, and molecular features. DESIGN.— Forty-one cases were classified as gastric-, intestinal-, pancreatobiliary-type intraductal papillary neoplasm (IPN), intraductal oncocytic papillary neoplasm (IOPN), or intraductal tubulopapillary neoplasm (ITPN) on the basis of histology. All neoplasms were subjected to targeted next-generation sequencing. RESULTS.— The mean age at diagnosis was 69 years (42-81 years); male to female ratio was 1.3. Most neoplasms (n = 23, 56%) were extrahepatic/large (mean size, 4.6 cm). The majority (n = 32, 78%) contained high-grade dysplasia, and 68% (n = 28) revealed invasion. All gastric-type IPNs (n = 9) and most ITPNs/IOPNs showed consistent colabeling for CK7/MUC6, which was less common among others (P = .004). Intestinal-type IPNs (n = 5) showed higher rates of CK20 expression than others (P < .001). Overall, the most commonly mutated genes included TP53 and APC, while copy number variants affected ELF3 and CDKN2A/B. All gastric-type IPNs contained an alteration affecting the Wnt signaling pathway; 7 of 9 (78%) showed aberrations in the MAPK pathway. Mutations in APC and KRAS were common in gastric-type IPNs as compared with others (P = .01 for both). SMAD4 was more frequently mutated in intestinal-type IPNs (P = .02). Pancreatobiliary-type IPNs (n = 14) exhibited frequent alterations in tumor suppressor genes including TP53, CDKN2A/B, and ARID2 (P = .04, P = .01 and P = .002, respectively). Of 6 IOPNs analyzed, 3 (50%) revealed ATP1B1-PRKACB fusion. ITPNs (n = 6) showed relatively few recurrent genetic aberrations. Follow-up information was available for 38 patients (median, 58.5 months). The ratio of disease-related deaths was higher for the cases with invasion (56% versus 10%). CONCLUSIONS.— Tumoral intraductal neoplasms of the bile ducts, similar to their counterparts in the pancreas, are morphologically and genetically heterogeneous.
Collapse
Affiliation(s)
- Tao Wang
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Gokce Askan
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Kerem Ozcan
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Satshil Rana
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Ahmet Zehir
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Umeshkumar K Bhanot
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Rhonda K Yantiss
- Department of Pathology, Weill Cornell Medicine, New York, New York (Yantiss)
| | - Deepthi S Rao
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Samuel J Wahl
- Department of Pathology, Lenox Hill Hospital, New York, New York (Wahl)
| | - Pelin Bagci
- Department of Pathology, Marmara University Hospital, Istanbul, Turkey (Bagci)
| | - Serdar Balci
- Department of Pathology, Memorial Healthcare Group, Istanbul, Turkey (Balci)
| | - Vinod Balachandran
- The Department of Surgery (Balachandran, Jarnagin), Memorial Sloan Kettering Cancer Center, New York, New York
| | - William R Jarnagin
- The Department of Surgery (Balachandran, Jarnagin), Memorial Sloan Kettering Cancer Center, New York, New York
| | - N Volkan Adsay
- The Department of Pathology, Koç University Hospital and Koç University Research Center for Translational Medicine (KUTTAM), Istanbul, Turkey (Adsay)
| | - David S Klimstra
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| | - Olca Basturk
- From the Department of Pathology and Laboratory Medicine (Wang, Askan, Ozcan, Rana, Zehir, Bhanot, Rao, Klimstra, Basturk), Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
66
|
[Chinese Expert Consensus on the Clinical Practice of Non-small Cell Lung Cancer
Fusion Gene Detection Based on RNA-based NGS]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2023; 26:801-812. [PMID: 37985137 PMCID: PMC10714047 DOI: 10.3779/j.issn.1009-3419.2023.102.43] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Indexed: 11/22/2023]
Abstract
RNA-based next-generation sequencing (NGS) has been recommended as a method for detecting fusion genes in non-small cell lung cancer (NSCLC) according to clinical practice guidelines and expert consensus. The primary targetable alterations in NSCLC consist of gene mutations and fusions, making the detection of gene mutations and fusions indispensable for assessing the feasibility of targeted therapies. Currently, the integration of DNA-based NGS and RNA-based NGS allows for simultaneous detection of gene mutations and fusions and has been partially implemented in clinical practice. However, standardized guidelines and criteria for the significance, application scenarios, and quality control of RNA-based NGS in fusion gene detection are still lacking in China. This consensus aims to provide further clarity on the practical significance, application scenarios, and quality control measures of RNA-based NGS in fusion gene detection. Additionally, it offers guiding recommendations to facilitate the clinical implementation of RNA-based NGS in the diagnosis and treatment of NSCLC, ultimately maximizing the benefits for patients from fusion gene detection.
.
Collapse
|
67
|
Gu W, Zhuang W, Zhuang M, He M, Li Z. DNA damage response and repair gene mutations are associated with tumor mutational burden and outcomes to platinum-based chemotherapy/immunotherapy in advanced NSCLC patients. Diagn Pathol 2023; 18:119. [PMID: 37924135 PMCID: PMC10623851 DOI: 10.1186/s13000-023-01401-0] [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: 07/08/2023] [Accepted: 10/08/2023] [Indexed: 11/06/2023] Open
Abstract
BACKGROUND DNA damage response and repair (DDR) genes are crucial for maintaining the integrity of the genome. This study aims to explore the correlation of DDR gene mutations with TMB, clinical characteristics, and outcomes to platinum-based chemotherapy and platinum-based chemotherapy/immunotherapy in non-small cell lung cancer (NSCLC) without EGFR and ALK alterations. METHODS Tumor tissue from 49 patients with stage III or IV NSCLC who were without EGFR and ALK alterations were analyzed using targeted next-generation sequencing (NGS). Among them, 13 patients received first-line platinum-based chemotherapy, 32 patients received first-line platinum-based chemotherapy/immunotherapy. RESULTS In these NSCLC patients without EGFR and ALK alterations, the frequently mutated genes included TP53, KMT2D and KRAS, the most frequently mutated DDR gene was FANCG, DDR gene mutations were detected in 20 patients. The mutation frequency of homologous recombination (HR) pathway was significantly higher in lung squamous cell carcinoma (LUSC) than that in lung adenocarcinoma (LUAD) (30.8% vs. 5.7%). Among DDR positive patients, a lower percentage exhibited metastasis. Patients with DDR gene mutations, cell-cycle checkpoint pathway mutations, and BER pathway mutations had significantly higher TMB compared to those without corresponding mutations. In the patients receiving platinum-based chemotherapy/immunotherapy, the disease control rate was significantly lower in the DDR-positive group compared with that in the DDR-negative group (55.6% vs. 100.0%). Among LUAD patients receiving platinum-based chemotherapy/immunotherapy, we observed a worse overall survival (OS) in DDR-positive group, as well as poorer progression-free survival(PFS)and OS in BER-positive and FANCG mutated group. CONCLUSIONS DDR gene mutations are associated with tumor metastasis, TMB, and outcomes to platinum-based chemotherapy/immunotherapy in advanced NSCLC patients.
Collapse
Affiliation(s)
- Weiguang Gu
- Department of Oncology, Nanhai People's hospital/the Sixth Affiliated Hospital of South China University of Technology, Foshan, 528200, Guangdong province, China.
| | - Wenya Zhuang
- Department of Oncology, Nanhai People's hospital/the Sixth Affiliated Hospital of South China University of Technology, Foshan, 528200, Guangdong province, China
- The Second Clinical Medical College, Southern Medical University, Guangzhou, 510515, Guangdong province, China
| | - Mengxia Zhuang
- Department of Oncology, Nanhai People's hospital/the Sixth Affiliated Hospital of South China University of Technology, Foshan, 528200, Guangdong province, China
- The Second Clinical Medical College, Southern Medical University, Guangzhou, 510515, Guangdong province, China
| | - Minhong He
- Department of Oncology, Nanhai People's hospital/the Sixth Affiliated Hospital of South China University of Technology, Foshan, 528200, Guangdong province, China
| | - Zhihua Li
- Department of Oncology, Nanhai People's hospital/the Sixth Affiliated Hospital of South China University of Technology, Foshan, 528200, Guangdong province, China
| |
Collapse
|
68
|
Wang X, Liu LL, Li Q, Xia QY, Li R, Ye SB, Zhang RS, Fang R, Chen H, Wu N, Rao Q. Loss of YAP1 C-terminus expression as an ancillary marker for metaplastic thymoma: a potential pitfall in detecting YAP1::MAML2 gene rearrangement. Histopathology 2023; 83:798-809. [PMID: 37565303 DOI: 10.1111/his.15024] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/11/2023] [Accepted: 07/21/2023] [Indexed: 08/12/2023]
Abstract
AIMS Metaplastic thymoma is a rare thymic tumour characterized by Yes Associated Protein 1 (YAP1) and Mastermind Like Transcriptional Coactivator 2 (MAML2) gene fusions resulting from an intrachromosomal inversion of chromosome 11. Immunohistochemistry with an antibody directed against the C-terminus of YAP1 has shown loss of expression in YAP1-rearranged vascular neoplasms, poromas, and porocarcinomas. This study aimed to validate an anti-YAP1 C-terminal antibody as an ancillary immunohistochemical marker for the diagnosis of metaplastic thymoma. MATERIALS AND METHODS Ten metaplastic thymomas were selected for the current study. Fluorescence in situ hybridization (FISH), next-generation sequencing (NGS), and reverse transcription-polymerase chain reaction (RT-PCR) analyses were performed to detect YAP1::MAML2 fusions. We then performed immunohistochemistry to detect YAP1 C-terminus expression in 10 metaplastic thymomas, 50 conventional thymomas (10 each of type A thymoma, type AB thymoma, type B1 thymoma, type B2 thymoma, and type B3 thymoma) and seven thymic carcinomas. RESULTS All 10 cases showed narrow split signals with a distance of nearly two signal diameters and sometimes had false-negative results in YAP1 and MAML2 break-apart FISH (BA-FISH). Abnormal colocalized signals of the YAP1::MAML2 fusion were observed in all 10 cases using fusion FISH (F-FISH) assays. Eight of 10 cases with adequate nucleic acids were successfully sequenced and all showed YAP1::MAML2 fusions; in two cases the fusions were detected by both DNA and RNA sequencing and in six cases by RNA sequencing only. YAP1::MAML2 fusion transcripts were identified in four cases by RT-PCR. Metaplastic thymoma showed loss of YAP1 C-terminus expression in all 10 (100%) cases. All other thymic neoplasms showed retained YAP1 C-terminus expression. CONCLUSION YAP1 C-terminus immunohistochemistry is a highly sensitive and specific ancillary marker that distinguishes metaplastic thymoma from its mimics. BA-FISH assays could not effectively detect YAP1::MAML2 fusions due to the proximity of the two genes. Loss of YAP1 C-terminus expression is a reliable surrogate for the detection of YAP1::MAML2 fusions in metaplastic thymoma.
Collapse
Affiliation(s)
- Xuan Wang
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Lei-Lei Liu
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qing Li
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qiu-Yuan Xia
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Rui Li
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Sheng-Bing Ye
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ru-Song Zhang
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Ru Fang
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Hui Chen
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Nan Wu
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Qiu Rao
- Department of Pathology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
69
|
Ranghiero A, Frascarelli C, Cursano G, Pescia C, Ivanova M, Vacirca D, Rappa A, Taormina SV, Barberis M, Fusco N, Rocco EG, Venetis K. Circulating tumour DNA testing in metastatic breast cancer: Integration with tissue testing. Cytopathology 2023; 34:519-529. [PMID: 37640801 DOI: 10.1111/cyt.13295] [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: 06/16/2023] [Revised: 07/26/2023] [Accepted: 08/14/2023] [Indexed: 08/31/2023]
Abstract
Breast cancer biomarker profiling predominantly relies on tissue testing (surgical and/or biopsy samples). However, the field of liquid biopsy, particularly the analysis of circulating tumour DNA (ctDNA), has witnessed remarkable progress and continues to evolve rapidly. The incorporation of ctDNA-based testing into clinical practice is creating new opportunities for patients with metastatic breast cancer (MBC). ctDNA offers advantages over conventional tissue analyses, as it reflects tumour heterogeneity and enables multiple serial biopsies in a minimally invasive manner. Thus, it serves as a valuable complement to standard tumour tissues and, in certain instances, may even present a potential alternative approach. In the context of MBC, ctDNA testing proves highly informative in the detection of disease progression, monitoring treatment response, assessing actionable biomarkers, and identifying mechanisms of resistance. Nevertheless, ctDNA does exhibit inherent limitations, including its generally low abundance, necessitating timely blood samplings and rigorous management of the pre-analytical phase. The development of highly sensitive assays and robust bioinformatic tools has paved the way for reliable ctDNA analyses. The time has now come to establish how ctDNA and tissue analyses can be effectively integrated into the diagnostic workflow of MBC to provide patients with the most comprehensive and accurate profiling. In this manuscript, we comprehensively analyse the current methodologies employed in ctDNA analysis and explore the potential benefits arising from the integration of tissue and ctDNA testing for patients diagnosed with MBC.
Collapse
Affiliation(s)
- Alberto Ranghiero
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Chiara Frascarelli
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Giulia Cursano
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Carlo Pescia
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- School of Pathology, University of Milan, Milan, Italy
| | - Mariia Ivanova
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Davide Vacirca
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Alessandra Rappa
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | | | - Massimo Barberis
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Nicola Fusco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Elena Guerini Rocco
- Division of Pathology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | | |
Collapse
|
70
|
Yavas A, Tan J, Ozkan HS, Yilmaz F, Reid MD, Bagci P, Shi J, Shia J, Adsay V, Klimstra DS, Basturk O. Solitary Fibrous Tumor of the Pancreas: Analysis of 9 Cases With Literature Review. Am J Surg Pathol 2023; 47:1230-1242. [PMID: 37573546 PMCID: PMC10592360 DOI: 10.1097/pas.0000000000002108] [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: 08/15/2023]
Abstract
Solitary fibrous tumor (SFT) has been increasingly reported in various anatomic sites. However, it is still extremely rare in the pancreas. Herein, we present the first series of primary pancreatic SFTs. Nine cases of primary pancreatic SFTs were analyzed. The mean age was 60 years (36 to 76 y) with no sex predilection. Six tumors were in the head, 3 were in the tail. On imaging studies, tumors were described as a hypervascular mass, 2 revealed cystic areas, and 3 were favored to be neuroendocrine tumors. On biopsy, 2 cases were diagnosed as atypical spindle cell tumor; one was misdiagnosed as suspicious for sarcoma, and another case as metastatic renal cell carcinoma. Two were diagnosed as low-grade sarcoma and low-grade stromal tumor on frozen sections. Grossly, tumors were well-demarcated with a median size of 4 cm (0.9 to 15 cm). Microscopically, they were composed of ovoid to spindle tumor cells with no significant mitotic activity and were arranged in alternating hypercellular and hypocellular areas. Staghorn-like vessels and entrapped pancreatic parenchyma were also detected within all tumors. Tumor cells revealed diffuse/strong nuclear STAT6 expression in 7 of 8, CD34 in 7 of 9, and bcl-2 in 4 of 4 tested cases. One tested tumor harbored NAB2 - STAT6 fusion. Eight patients with available follow-up data were free of disease at a mean follow-up of 76 months (3 to 189 mo). SFT should be considered in the differential diagnoses of mesenchymal neoplasms of the pancreas. Immunohistochemical nuclear STAT6 expression is a characteristic feature of SFT. Primary pancreatic SFTs seem to have favorable biological behavior in our series.
Collapse
Affiliation(s)
- Aslihan Yavas
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Hulya Sahin Ozkan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Funda Yilmaz
- Department of Pathology, Ege University, Izmir, Turkey
| | - Michelle D Reid
- Department of Pathology, Emory University Hospital, Atlanta, GA, USA
| | - Pelin Bagci
- Department of Pathology, Marmara University, Istanbul, Turkey
| | - Jiaqi Shi
- Department of Pathology and Clinical Labs, University of Michigan, Ann Arbor, MI, USA
| | - Jinru Shia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Volkan Adsay
- Department of Pathology, Koc University, Istanbul, Turkey
| | - David S. Klimstra
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Olca Basturk
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
71
|
Ptashkin RN, Ewalt MD, Jayakumaran G, Kiecka I, Bowman AS, Yao J, Casanova J, Lin YTD, Petrova-Drus K, Mohanty AS, Bacares R, Benhamida J, Rana S, Razumova A, Vanderbilt C, Balakrishnan Rema A, Rijo I, Son-Garcia J, de Bruijn I, Zhu M, Lachhander S, Wang W, Haque MS, Seshan VE, Wang J, Liu Y, Nafa K, Borsu L, Zhang Y, Aypar U, Suehnholz SP, Chakravarty D, Park JH, Abdel-Wahab O, Mato AR, Xiao W, Roshal M, Yabe M, Batlevi CL, Giralt S, Salles G, Rampal R, Tallman M, Stein EM, Younes A, Levine RL, Perales MA, van den Brink MRM, Dogan A, Ladanyi M, Berger MF, Brannon AR, Benayed R, Zehir A, Arcila ME. Enhanced clinical assessment of hematologic malignancies through routine paired tumor and normal sequencing. Nat Commun 2023; 14:6895. [PMID: 37898613 PMCID: PMC10613284 DOI: 10.1038/s41467-023-42585-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 10/16/2023] [Indexed: 10/30/2023] Open
Abstract
Genomic profiling of hematologic malignancies has augmented our understanding of variants that contribute to disease pathogenesis and supported development of prognostic models that inform disease management in the clinic. Tumor only sequencing assays are limited in their ability to identify definitive somatic variants, which can lead to ambiguity in clinical reporting and patient management. Here, we describe the MSK-IMPACT Heme cohort, a comprehensive data set of somatic alterations from paired tumor and normal DNA using a hybridization capture-based next generation sequencing platform. We highlight patterns of mutations, copy number alterations, and mutation signatures in a broad set of myeloid and lymphoid neoplasms. We also demonstrate the power of appropriate matching to make definitive somatic calls, including in patients who have undergone allogeneic stem cell transplant. We expect that this resource will further spur research into the pathobiology and clinical utility of clinical sequencing for patients with hematologic neoplasms.
Collapse
Affiliation(s)
- Ryan N Ptashkin
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- C2i Genomics, New York, NY, USA
| | - Mark D Ewalt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Gowtham Jayakumaran
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Guardant Health, Palo Alto, CA, USA
| | - Iwona Kiecka
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anita S Bowman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - JinJuan Yao
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jacklyn Casanova
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yun-Te David Lin
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kseniya Petrova-Drus
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Abhinita S Mohanty
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ruben Bacares
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jamal Benhamida
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Satshil Rana
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anna Razumova
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Chad Vanderbilt
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anoop Balakrishnan Rema
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ivelise Rijo
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Julie Son-Garcia
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ino de Bruijn
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Menglei Zhu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sean Lachhander
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wei Wang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mohammad S Haque
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Venkatraman E Seshan
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jiajing Wang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ying Liu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Khedoudja Nafa
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Laetitia Borsu
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yanming Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Umut Aypar
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sarah P Suehnholz
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Debyani Chakravarty
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jae H Park
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anthony R Mato
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Wenbin Xiao
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mikhail Roshal
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mariko Yabe
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Connie Lee Batlevi
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sergio Giralt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gilles Salles
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raajit Rampal
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Martin Tallman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - Eytan M Stein
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anas Younes
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Oncology R&D, AstraZeneca, New York, NY, USA
| | - Ross L Levine
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Miguel-Angel Perales
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Marcel R M van den Brink
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael F Berger
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - A Rose Brannon
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ryma Benayed
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Oncology R&D, AstraZeneca, New York, NY, USA
| | - Ahmet Zehir
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Oncology R&D, AstraZeneca, New York, NY, USA.
| | - Maria E Arcila
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| |
Collapse
|
72
|
Mitiushkina NV, Tiurin VI, Anuskina AA, Bordovskaya NA, Shestakova AD, Martianov AS, Bubnov MG, Shishkina AS, Semina MV, Romanko AA, Kuligina ES, Imyanitov EN. Molecular Analysis of Biliary Tract Cancers with the Custom 3' RACE-Based NGS Panel. Diagnostics (Basel) 2023; 13:3168. [PMID: 37891989 PMCID: PMC10605186 DOI: 10.3390/diagnostics13203168] [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: 09/02/2023] [Revised: 10/05/2023] [Accepted: 10/08/2023] [Indexed: 10/29/2023] Open
Abstract
The technique 3' rapid amplification of cDNA ends (3' RACE) allows for detection of translocations with unknown gene partners located at the 3' end of the chimeric transcript. We composed a 3' RACE-based RNA sequencing panel for the analysis of FGFR1-4 gene rearrangements, detection of activating mutations located within FGFR1-4, IDH1/2, ERBB2 (HER2), KRAS, NRAS, BRAF, and PIK3CA genes, and measurement of the expression of ERBB2, PD-L1, and FGFR1-4 transcripts. This NGS panel was utilized for the molecular profiling of 168 biliary tract carcinomas (BTCs), including 83 intrahepatic cholangiocarcinomas (iCCAs), 44 extrahepatic cholangiocarcinomas (eCCAs), and 41 gallbladder adenocarcinomas (GBAs). The NGS failure rate was 3/168 (1.8%). iCCAs, but not other categories of BTCs, were characterized by frequent FGFR2 alterations (17/82, 20.7%) and IDH1/2 mutations (23/82, 28%). Other potentially druggable events included ERBB2 amplifications or mutations (7/165, 4.2% of all successfully analyzed BTCs) and BRAF p.V600E mutations (3/165, 1.8%). In addition to NGS, we analyzed microsatellite instability (MSI) using the standard five markers and revealed this event in 3/158 (1.9%) BTCs. There were no instances of ALK, ROS1, RET, and NTRK1-3 gene rearrangements or MET exon 14 skipping mutations. Parallel analysis of 47 iCCA samples with the Illumina TruSight Tumor 170 kit confirmed good performance of our NGS panel. In conclusion, targeted RNA sequencing coupled with the 3' RACE technology is an efficient tool for the molecular diagnostics of BTCs.
Collapse
Affiliation(s)
- Natalia V. Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Vladislav I. Tiurin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Aleksandra A. Anuskina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Natalia A. Bordovskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Anna D. Shestakova
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Aleksandr S. Martianov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Mikhail G. Bubnov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Anna S. Shishkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Maria V. Semina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Aleksandr A. Romanko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
| | - Ekaterina S. Kuligina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St. Petersburg, Russia; (N.V.M.); (N.A.B.); (A.S.S.); (M.V.S.); (A.A.R.); (E.S.K.)
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, 194100 St. Petersburg, Russia
| |
Collapse
|
73
|
Chen H, Wang B, Zhang Y, Shu Y, Dong H, Zhao Q, Yang C, Li J, Duan X, Zhou Q. A unified DNA- and RNA-based NGS strategy for the analysis of multiple types of variants at the dual nucleic acid level in solid tumors. J Clin Lab Anal 2023; 37:e24977. [PMID: 37877443 PMCID: PMC10681543 DOI: 10.1002/jcla.24977] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/27/2023] [Accepted: 10/15/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Targeted next-generation sequencing (NGS) is a powerful and suitable approach to comprehensively identify multiple types of variants in tumors. RNA-based NGS is increasingly playing an important role in precision oncology. Both parallel and sequential DNA- and RNA-based approaches are expensive, burdensome, and have long turnaround times, which can be impractical in clinical practice. A streamlined, unified DNA- and RNA-based NGS approach is urgently needed in clinical practice. METHODS A DNA/RNA co-hybrid capture sequencing (DRCC-Seq) approach was designed to capture pre-capture DNA and RNA libraries in a single tube and convert them into one NGS library. The performance of the DRCC-Seq approach was evaluated by a panel of reference standards and clinical samples. RESULTS The average depth, DNA data ratio, capture ratio, and target coverage 250 (×) of the DNA panel data had a negative correlation with an increase in the proportion of RNA probes. The SNVs, indels, fusions, and MSI status were not affected by the proportion of RNA probes, but the copy numbers of the target genes were higher than expected in the standard materials, and many unexpected gene amplifications were found using D:R (1:2) and D:R (1:4) probe panels. The optimal ratio of DNA and RNA probes in the combined probe panel was 1:1 using the DRCC-Seq approach. The DRCC-Seq approach was feasible and reliable for detecting multiple types of variants in reference standards and real-world clinical samples. CONCLUSIONS The DRCC-Seq approach is more cost-effective, with a shorter turnaround time and lower labor requirements than either parallel or sequential targeted DNA NGS and RNA NGS. It is feasible to identify multiple genetic variations at the DNA and RNA levels simultaneously in clinical practice.
Collapse
Affiliation(s)
- Huijuan Chen
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
- Computer Network Information Center, Chinese Academy of SciencesBeijingChina
- WillingMed Technology Beijing Co., Ltd.BeijingChina
| | - Bing Wang
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
| | - Yiran Zhang
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
| | - Yingshuang Shu
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
| | - Henan Dong
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
| | - Qian Zhao
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
| | - Chunyan Yang
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
| | - Jianji Li
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
| | - Xiaohong Duan
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
- ChosenMed Technology (Zhejiang) Co. Ltd.ZhejiangChina
- Institute of Disaster and Emergency Medicine, Medical CollegeTianjin UniversityTianJinChina
| | - Qiming Zhou
- ChosenMed Clinical Laboratory (Beijing) Co. Ltd.BeijingChina
- ChosenMed Technology (Zhejiang) Co. Ltd.ZhejiangChina
| |
Collapse
|
74
|
Zhou S, Zhang F, Xu M, Zhang L, Liu Z, Yang Q, Wang C, Wang B, Ma T, Feng J. Novel insights into molecular patterns of ROS1 fusions in a large Chinese NSCLC cohort: a multicenter study. Mol Oncol 2023; 17:2200-2212. [PMID: 37584407 PMCID: PMC10552890 DOI: 10.1002/1878-0261.13509] [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: 06/24/2023] [Revised: 08/01/2023] [Accepted: 08/14/2023] [Indexed: 08/17/2023] Open
Abstract
ROS proto-oncogene 1, receptor tyrosine kinase (ROS1) rearrangements are a crucial therapeutic target in non-small cell lung cancer (NSCLC). However, there is limited comprehensive analysis of the molecular patterns of ROS1 fusions. This study aimed to address this gap by analysing 135 ROS1 fusions from 134 Chinese NSCLC patients using next-generation sequencing (NGS). The fusions were categorized into common and uncommon based on their incidence. Our study revealed, for the first time, a unique distribution preference of breakpoints within ROS1, with common fusions occurring in introns 31-33 and uncommon fusions occurring in introns 34 and 35. Additionally, we identified previously unknown breakpoints within intron 28 of ROS1. Furthermore, we identified a close association between the distribution patterns of fusion partners and breakpoints on ROS1, providing important insights into the molecular landscape of ROS1 fusions. We also confirmed the presence of inconsistent breakpoints in ROS1 fusions between DNA-based NGS and RNA-based NGS through rigorous validation methods. These inconsistencies were attributed to alternative splicing resulting in out-of-frame or exonic ROS1 fusions. These findings significantly contribute to our understanding of the molecular characteristics of ROS1 fusions, which have implications for panel design and the treatment of NSCLC patients with ROS1 rearrangements.
Collapse
Affiliation(s)
- Shengyu Zhou
- Clinical Nursing Department, School of Nursing and Rehabilitation, Cheeloo College of MedicineShandong UniversityJinanChina
- Department of Respiratory and Critical Care Medicine, Qilu Hospital, Cheeloo College of MedicineShandong UniversityJinanChina
| | - Fayan Zhang
- College of Traditional Chinese MedicineShandong University of Traditional Chinese MedicineJinanChina
| | - Mengxiang Xu
- Jichenjunchuang Clinical LaboratoryHangzhouChina
- Genecn‐Biotech Co.LtdHangzhouChina
| | - Lei Zhang
- Cancer Center, Daping HospitalArmy Medical UniversityChongqingChina
| | - Zhengchuang Liu
- Key Laboratory of Gastroenterology of Zhejiang ProvinceZhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical CollegeChina
| | - Qiong Yang
- General Surgery, Cancer CenterZhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College)China
| | - Chunyang Wang
- Jichenjunchuang Clinical LaboratoryHangzhouChina
- Genecn‐Biotech Co.LtdHangzhouChina
| | - Baoming Wang
- Jichenjunchuang Clinical LaboratoryHangzhouChina
- Genecn‐Biotech Co.LtdHangzhouChina
| | - Tonghui Ma
- Jichenjunchuang Clinical LaboratoryHangzhouChina
- Genecn‐Biotech Co.LtdHangzhouChina
- Key Laboratory of Gastroenterology of Zhejiang ProvinceZhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical CollegeChina
| | - Jiao Feng
- Cancer Center, Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang ProvinceZhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical CollegeChina
- General Surgery, Department of Gastrointestinal and Pancreatic Surgery, Cancer CenterZhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical CollegeChina
- School of PharmacyHangzhou Normal UniversityChina
| |
Collapse
|
75
|
Shreenivas A, Janku F, Gouda MA, Chen HZ, George B, Kato S, Kurzrock R. ALK fusions in the pan-cancer setting: another tumor-agnostic target? NPJ Precis Oncol 2023; 7:101. [PMID: 37773318 PMCID: PMC10542332 DOI: 10.1038/s41698-023-00449-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Accepted: 09/05/2023] [Indexed: 10/01/2023] Open
Abstract
Anaplastic lymphoma kinase (ALK) alterations (activating mutations, amplifications, and fusions/rearrangements) occur in ~3.3% of cancers. ALK fusions/rearrangements are discerned in >50% of inflammatory myofibroblastic tumors (IMTs) and anaplastic large cell lymphomas (ALCLs), but only in ~0.2% of other cancers outside of non-small cell lung cancer (NSCLC), a rate that may be below the viability threshold of even large-scale treatment trials. Five ALK inhibitors -alectinib, brigatinib, ceritinb, crizotinib, and lorlatinib-are FDA approved for ALK-aberrant NSCLCs, and crizotinib is also approved for ALK-aberrant IMTs and ALCL, including in children. Herein, we review the pharmacologic tractability of ALK alterations, focusing beyond NSCLC. Importantly, the hallmark of approved indications is the presence of ALK fusions/rearrangements, and response rates of ~50-85%. Moreover, there are numerous reports of ALK inhibitor activity in multiple solid and hematologic tumors (e.g., histiocytosis, leiomyosarcoma, lymphoma, myeloma, and colorectal, neuroendocrine, ovarian, pancreatic, renal, and thyroid cancer) bearing ALK fusions/rearrangements. Many reports used crizotinib or alectinib, but each of the approved ALK inhibitors have shown activity. ALK inhibitor activity is also seen in neuroblastoma, which bear ALK mutations (rather than fusions/rearrangements), but response rates are lower (~10-20%). Current data suggests that ALK inhibitors have tissue-agnostic activity in neoplasms bearing ALK fusions/rearrangements.
Collapse
Affiliation(s)
- Aditya Shreenivas
- Medical College of Wisconsin (MCW) Cancer Center, Milwaukee, WI, USA.
| | | | - Mohamed A Gouda
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui-Zi Chen
- Medical College of Wisconsin (MCW) Cancer Center, Milwaukee, WI, USA
| | - Ben George
- Medical College of Wisconsin (MCW) Cancer Center, Milwaukee, WI, USA
| | - Shumei Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, UC San Diego Moores Cancer Center, La Jolla, CA, USA
| | - Razelle Kurzrock
- Medical College of Wisconsin (MCW) Cancer Center, Milwaukee, WI, USA.
- University of Nebraska, Omaha, NE, USA.
- Worldwide Innovative Network (WIN) for Personalized Cancer Therapy, Chevilly-Larue, France.
| |
Collapse
|
76
|
Romanko AA, Mulkidjan RS, Tiurin VI, Saitova ES, Preobrazhenskaya EV, Krivosheyeva EA, Mitiushkina NV, Shestakova AD, Belogubova EV, Ivantsov AO, Iyevleva AG, Imyanitov EN. Cost-Efficient Detection of NTRK1/2/3 Gene Fusions: Single-Center Analysis of 8075 Tumor Samples. Int J Mol Sci 2023; 24:14203. [PMID: 37762506 PMCID: PMC10531831 DOI: 10.3390/ijms241814203] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/26/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
The majority of NTRK1, NTRK2, and NTRK3 rearrangements result in increased expression of the kinase portion of the involved gene due to its fusion to an actively transcribed gene partner. Consequently, the analysis of 5'/3'-end expression imbalances is potentially capable of detecting the entire spectrum of NTRK gene fusions. Archival tumor specimens obtained from 8075 patients were subjected to manual dissection of tumor cells, DNA/RNA isolation, and cDNA synthesis. The 5'/3'-end expression imbalances in NTRK genes were analyzed by real-time PCR. Further identification of gene rearrangements was performed by variant-specific PCR for 44 common NTRK fusions, and, whenever necessary, by RNA-based next-generation sequencing (NGS). cDNA of sufficient quality was obtained in 7424/8075 (91.9%) tumors. NTRK rearrangements were detected in 7/6436 (0.1%) lung carcinomas, 11/137 (8.0%) pediatric tumors, and 13/851 (1.5%) adult non-lung malignancies. The highest incidence of NTRK translocations was observed in pediatric sarcomas (7/39, 17.9%). Increased frequency of NTRK fusions was seen in microsatellite-unstable colorectal tumors (6/48, 12.5%), salivary gland carcinomas (5/93, 5.4%), and sarcomas (7/143, 4.9%). None of the 1293 lung carcinomas with driver alterations in EGFR/ALK/ROS1/RET/MET oncogenes had NTRK 5'/3'-end expression imbalances. Variant-specific PCR was performed for 744 tumors with a normal 5'/3'-end expression ratio: there were no rearrangements in 172 EGFR/ALK/ROS1/RET/MET-negative lung cancers and 125 pediatric tumors, while NTRK3 fusions were detected in 2/447 (0.5%) non-lung adult malignancies. In conclusion, this study describes a diagnostic pipeline that can be used as a cost-efficient alternative to conventional methods of NTRK1-3 analysis.
Collapse
Affiliation(s)
- Aleksandr A. Romanko
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Rimma S. Mulkidjan
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Vladislav I. Tiurin
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Evgeniya S. Saitova
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Elena V. Preobrazhenskaya
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Elena A. Krivosheyeva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Natalia V. Mitiushkina
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Anna D. Shestakova
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Evgeniya V. Belogubova
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Alexandr O. Ivantsov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
| | - Aglaya G. Iyevleva
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, 194100 St.-Petersburg, Russia
| | - Evgeny N. Imyanitov
- Department of Tumor Growth Biology, N.N. Petrov Institute of Oncology, 197758 St.-Petersburg, Russia (V.I.T.)
- Department of Medical Genetics, St.-Petersburg Pediatric Medical University, 194100 St.-Petersburg, Russia
| |
Collapse
|
77
|
Valery M, Vasseur D, Fachinetti F, Boilève A, Smolenschi C, Tarabay A, Antoun L, Perret A, Fuerea A, Pudlarz T, Boige V, Hollebecque A, Ducreux M. Targetable Molecular Alterations in the Treatment of Biliary Tract Cancers: An Overview of the Available Treatments. Cancers (Basel) 2023; 15:4446. [PMID: 37760415 PMCID: PMC10526255 DOI: 10.3390/cancers15184446] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 08/27/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Biliary tract cancers (BTCs) are rare tumours, most often diagnosed at an unresectable stage, associated with poor prognosis, with a 5-year survival rate not exceeding 10%. Only first- and second-line treatments are well codified with the combination of cisplatin-gemcitabine chemotherapy and immunotherapy followed by 5-FU and oxaliplatin chemotherapy, respectively. Many studies have shown that BTC, and more particularly intrahepatic cholangiocarcinoma (iCCA), have a high rate of targetable somatic alteration. To date, the FDA has approved several drugs. Ivosidenib targeting IDH1 mutations, as well as futibatinib and pemigatinib targeting FGFR2 fusions, are approved for pre-treated advanced CCA. The combination of dabrafenib and trametinib are approved for BRAFV600E mutated advanced tumours, NTRK inhibitors entrectinib and larotrectinib for tumours bearing NTRK fusion and prembrolizumab for MSI-H advanced tumours, involving a small percentage of BTC in these three settings. Several other potentially targetable alterations are found in BTC, such as HER2 mutations or amplifications or KRASG12C mutations and mutations in genes involved in DNA repair mechanisms. This review aims to clarify the specific diagnostic modalities for gene alterations and to summarize the results of the main trials and developments underway for the management of advanced BTC with targetable alterations.
Collapse
Affiliation(s)
- Marine Valery
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Damien Vasseur
- Medical Biology and Pathology Department, Gustave Roussy, F-94805 Villejuif, France;
| | - Francesco Fachinetti
- Dana-Farber Institute, Lowe Center for Thoracic Oncology, Boston, MA 02215, USA;
| | - Alice Boilève
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, F-94805 Villejuif, France
| | - Cristina Smolenschi
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Département d’Innovation Thérapeutique, Gustave Roussy, F-94805 Villejuif, France
| | - Anthony Tarabay
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Leony Antoun
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Audrey Perret
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Alina Fuerea
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Thomas Pudlarz
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Valérie Boige
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
| | - Antoine Hollebecque
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Département d’Innovation Thérapeutique, Gustave Roussy, F-94805 Villejuif, France
| | - Michel Ducreux
- Medical Oncology Department, Gustave Roussy, F-94805 Villejuif, France; (A.B.); (C.S.); (A.T.); (L.A.); (A.P.); (A.F.); (T.P.); (V.B.); (A.H.); (M.D.)
- Université Paris-Saclay, Gustave Roussy, Inserm Unité Dynamique des Cellules Tumorales, F-94805 Villejuif, France
| |
Collapse
|
78
|
Cufer T, Kosty MP. ESMO/ASCO Recommendations for a Global Curriculum in Medical Oncology Edition 2023. JCO Glob Oncol 2023; 9:e2300277. [PMID: 37867478 PMCID: PMC10664856 DOI: 10.1200/go.23.00277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 08/24/2023] [Indexed: 10/24/2023] Open
Abstract
The European Society for Medical Oncology (ESMO) and ASCO are publishing a new edition of the ESMO/ASCO Global Curriculum (GC) with contributions from more than 150 authors. The purpose of the GC is to provide recommendations for the training of physicians in medical oncology and to establish a set of educational standards for trainees to qualify as medical oncologists. This edition builds on prior ones in 2004, 2010, and 2016 and incorporates scientific advances and input from an ESMO ASCO survey on GC adoption conducted in 2019, which revealed that GC has been adopted or adapted in as many as two thirds of the countries surveyed. To make GC even more useful and applicable, certain subchapters were rearranged into stand-alone chapters, that is, cancer epidemiology, diagnostics, and research. In line with recent progress in the field of multidisciplinary cancer care new (sub)chapters, such as image-guided therapy, cell-based therapy, and nutritional support, were added. Moreover, this edition includes an entirely new chapter dedicated to cancer control principles, aiming to ensure that medical oncologists are able to identify and implement sustainable and equitable cancer care, tailored to local needs and resources. Besides content renewal, modern didactic principles were introduced. GC content is presented using two chapter templates (cancer-specific and non-cancer-specific), with three didactic points (objectives, key concepts, and skills). The next step is promoting GC as a contemporary and comprehensive document applicable all over the world, particularly due to its capacity to harmonize education in medical oncology and, in so doing, help to reduce global disparities in cancer care.
Collapse
Affiliation(s)
- Tanja Cufer
- Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Michael P. Kosty
- Division of Hematology and Oncology, Scripps MD Anderson Cancer Center, La Jolla, CA
| |
Collapse
|
79
|
Mazieres J, Vioix H, Pfeiffer BM, Campden RI, Chen Z, Heeg B, Cortot AB. MET Exon 14 Skipping in NSCLC: A Systematic Literature Review of Epidemiology, Clinical Characteristics, and Outcomes. Clin Lung Cancer 2023; 24:483-497. [PMID: 37451931 DOI: 10.1016/j.cllc.2023.06.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 05/30/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
INTRODUCTION MET exon 14 (METex14) skipping is a rare oncogenic driver in non-small-cell lung cancer (NSCLC) for which targeted therapy with MET tyrosine kinase inhibitors (TKIs) was recently approved. Given the heterogeneity in published data of METex14 skipping NSCLC, we conducted a systematic literature review to evaluate its frequency, patient characteristics, and outcomes. METHODS On June 13, 2022 we conducted a systematic literature review of publications and conference abstracts reporting frequency, patient characteristics, or outcomes of patients with METex14 skipping NSCLC. RESULTS We included 139 studies reporting frequency or patient characteristics (350,997 patients), and 39 studies reporting clinical outcomes (3989 patients). Median METex14 skipping frequency was 2.0% in unselected patients with NSCLC, with minimal geographic variation. Median frequency was 2.4% in adenocarcinoma or nonsquamous subgroups, 12.0% in sarcomatoid, and 1.3% in squamous histology. Patients with METex14 skipping NSCLC were more likely to be elderly, have adenocarcinoma histology; there was no marked sex or smoking status distribution. In first line of treatment, median objective response rate ranged from 50.7% to 68.8% with targeted therapies (both values correspond to MET TKIs), was 33.3% with immunotherapy, and ranged from 23.1% to 27.0% with chemotherapy. CONCLUSIONS Patients with METex14 skipping are more likely to have certain characteristics, but no patient subgroup can be ruled out; thus, it is crucial to test all patients with NSCLC to identify suitable candidates for MET inhibitor therapy. MET TKIs appeared to result in higher efficacy outcomes, although no direct comparison with chemotherapy or immunotherapy regimens was found.
Collapse
Affiliation(s)
- Julien Mazieres
- CHU de Toulouse, Université Paul Sabatier, Toulouse, France.
| | - Helene Vioix
- The Healthcare Business of Merck KGaA, Darmstadt, Germany
| | | | | | - Zhiyuan Chen
- Ingress Health, A Cytel Company, Rotterdam, The Netherlands
| | - Bart Heeg
- Ingress Health, A Cytel Company, Rotterdam, The Netherlands
| | - Alexis B Cortot
- Université Lille, Centre Hospitalier Universitaire de Lille, Centre national de la recherche scientifique, Inserm, Institute Pasteur de Lille, Lille, France
| |
Collapse
|
80
|
Saldivar JS, Harris J, Ayash E, Hong M, Tandon P, Sinha S, Hebron PM, Houghton EE, Thorne K, Goodman LJ, Li C, Marfatia TR, Anderson J, Morra M, Lyle J, Bartha G, Chen R. Analytic validation of NeXT Dx™, a comprehensive genomic profiling assay. Oncotarget 2023; 14:789-806. [PMID: 37646774 PMCID: PMC10467627 DOI: 10.18632/oncotarget.28490] [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: 05/31/2023] [Accepted: 08/19/2023] [Indexed: 09/01/2023] Open
Abstract
We describe the analytic validation of NeXT Dx, a comprehensive genomic profiling assay to aid therapy and clinical trial selection for patients diagnosed with solid tumor cancers. Proprietary methods were utilized to perform whole exome and whole transcriptome sequencing for detection of single nucleotide variants (SNVs), insertions/deletions (indels), copy number alterations (CNAs), and gene fusions, and determination of tumor mutation burden and microsatellite instability. Variant calling is enhanced by sequencing a patient-specific normal sample from, for example, a blood specimen. This provides highly accurate somatic variant calls as well as the incidental reporting of pathogenic and likely pathogenic germline alterations. Fusion detection via RNA sequencing provides more extensive and accurate fusion calling compared to DNA-based tests. NeXT Dx features the proprietary Accuracy and Content Enhanced technology, developed to optimize sequencing and provide more uniform coverage across the exome. The exome was validated at a median sequencing depth of >500x. While variants from 401 cancer-associated genes are currently reported from the assay, the exome/transcriptome assay is broadly validated to enable reporting of additional variants as they become clinically relevant. NeXT Dx demonstrated analytic sensitivities as follows: SNVs (99.4%), indels (98.2%), CNAs (98.0%), and fusions (95.8%). The overall analytic specificity was >99.0%.
Collapse
Affiliation(s)
| | - Jason Harris
- Personalis, Inc., Fremont, CA 94555, USA
- These authors contributed equally to this work
| | - Erin Ayash
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | | | | | | | | | | | | - Conan Li
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | | | | - John Lyle
- Personalis, Inc., Fremont, CA 94555, USA
| | | | | |
Collapse
|
81
|
Desilets A, Repetto M, Yang SR, Sherman EJ, Drilon A. RET-Altered Cancers-A Tumor-Agnostic Review of Biology, Diagnosis and Targeted Therapy Activity. Cancers (Basel) 2023; 15:4146. [PMID: 37627175 PMCID: PMC10452615 DOI: 10.3390/cancers15164146] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 08/11/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
RET alterations, such as fusions or mutations, drive the growth of multiple tumor types. These alterations are found in canonical (lung and thyroid) and non-canonical (e.g., gastrointestinal, breast, gynecological, genitourinary, histiocytic) cancers. RET alterations are best identified via comprehensive next-generation sequencing, preferably with DNA and RNA interrogation for fusions. Targeted therapies for RET-dependent cancers have evolved from older multikinase inhibitors to selective inhibitors of RET such as selpercatinib and pralsetinib. Prospective basket trials and retrospective reports have demonstrated the activity of these drugs in a wide variety of RET-altered cancers, notably those with RET fusions. This paved the way for the first tumor-agnostic selective RET inhibitor US FDA approval in 2022. Acquired resistance to RET kinase inhibitors can take the form of acquired resistance mutations (e.g., RET G810X) or bypass alterations.
Collapse
Affiliation(s)
- Antoine Desilets
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.D.); (M.R.); (S.-R.Y.); (E.J.S.)
| | - Matteo Repetto
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.D.); (M.R.); (S.-R.Y.); (E.J.S.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20133 Milan, Italy
| | - Soo-Ryum Yang
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.D.); (M.R.); (S.-R.Y.); (E.J.S.)
| | - Eric J. Sherman
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.D.); (M.R.); (S.-R.Y.); (E.J.S.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Alexander Drilon
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (A.D.); (M.R.); (S.-R.Y.); (E.J.S.)
- Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| |
Collapse
|
82
|
Treichler G, Hoeller S, Rueschoff JH, Rechsteiner M, Britschgi C, Arnold F, Zoche M, Hiltbrunner S, Moch H, Akhoundova D, Opitz I, Curioni-Fontecedro A. Improving the turnaround time of molecular profiling for advanced non-small cell lung cancer: Outcome of a new algorithm integrating multiple approaches. Pathol Res Pract 2023; 248:154660. [PMID: 37413876 DOI: 10.1016/j.prp.2023.154660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/08/2023]
Abstract
BACKGROUND Molecular tumor profiling to identify oncogenic drivers and actionable mutations has a profound impact on how lung cancer is treated. Especially in the subgroup of non-small cell lung cancer (NSCLC), molecular testing for certain mutations is crucial in daily clinical practice and is recommended by international guidelines. To date, a standardized approach to identify druggable genetic alterations are lacking. We have developed and implemented a new diagnostic algorithm to harmonize the molecular testing of NSCLC. PATIENTS AND METHODS In this retrospective analysis, we reviewed 119 patients diagnosed with NSCLC at the University Hospital Zurich. Tumor samples were analyzed using our standardized diagnostic algorithm: After the histological diagnosis was made, tissue samples were further analyzed by immunohistochemical stainings as well as the real-time PCR test Idylla™. Extracted DNA was further utilized for comprehensive genomic profiling (FoundationOne®CDx, F1CDx). RESULTS Out of the 119 patients were included in this study, 100 patients were diagnosed with non-squamous NSCLC (nsqNSCLC) and 19 with squamous NSCLC (sqNSCLC). The samples from the nsqNSCLC patients underwent testing by Idylla™ and were evaluated by immunohistochemistry (IHC). F1CDx analysis was run on 67 samples and 46 potentially actionable genomic alterations were detected. Ten patients received the indicated targeted treatment. The median time to test results was 4 days for the Idylla test, 5 days for IHC and 13 days for the F1CDx. CONCLUSION In patients with NSCLC, the implementation of a standardized molecular testing algorithm provided information on predictive markers for NSCLC within a few working days. The implementation of broader genomic profiling led to the identification of actionable targets, which would otherwise not have been discovered.
Collapse
Affiliation(s)
- G Treichler
- Department of Medical Oncology and Hematology University Hospital Zurich, Switzerland; Comprehensive Cancer Center Zurich, Switzerland; Department of Medical Oncology and Hematology, Cantonal Hospital Winterthur, Switzerland
| | - S Hoeller
- Department of Medical Oncology and Hematology University Hospital Zurich, Switzerland; Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland; University of Zurich, Switzerland; Comprehensive Cancer Center Zurich, Switzerland
| | - J H Rueschoff
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - M Rechsteiner
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - C Britschgi
- Department of Medical Oncology and Hematology University Hospital Zurich, Switzerland; University of Zurich, Switzerland; Comprehensive Cancer Center Zurich, Switzerland
| | - F Arnold
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - M Zoche
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - S Hiltbrunner
- Department of Medical Oncology and Hematology University Hospital Zurich, Switzerland; Comprehensive Cancer Center Zurich, Switzerland; Faculty of Science and Medicine, University of Fribourg, Switzerland
| | - H Moch
- Department of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland
| | - D Akhoundova
- Department of Medical Oncology and Hematology University Hospital Zurich, Switzerland; Comprehensive Cancer Center Zurich, Switzerland; Department of Oncology, University Hospital Bern, Switzerland
| | - I Opitz
- University of Zurich, Switzerland; Department of Thoracic Surgery, University Hospital Zurich, Switzerland
| | - A Curioni-Fontecedro
- Department of Medical Oncology and Hematology University Hospital Zurich, Switzerland; University of Zurich, Switzerland; Comprehensive Cancer Center Zurich, Switzerland; Faculty of Science and Medicine, University of Fribourg, Switzerland; Department of Oncology, Cantonal Hospital Fribourg, Switzerland.
| |
Collapse
|
83
|
Hernandez S, Conde E, Alonso M, Illarramendi A, Bote de Cabo H, Zugazagoitia J, Paz-Ares L, Lopez-Rios F. A narrative review of methods for the identification of ALK fusions in patients with non-small cell lung carcinoma. Transl Lung Cancer Res 2023; 12:1549-1562. [PMID: 37577307 PMCID: PMC10413037 DOI: 10.21037/tlcr-22-855] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Accepted: 06/25/2023] [Indexed: 08/15/2023]
Abstract
Background and Objective This narrative review is intended to provide pragmatic knowledge of current methods for the search of anaplastic lymphoma kinase (ALK) fusions in patients with non-small cell lung carcinoma (NSCLC). This information is very timely, because a recent survey has identified that almost 50% of patients with advanced NSCLC were not candidates for targeted therapies because of biomarker testing issues. Methods PubMed was searched from January 1st, 2012 to February 28th, 2023 using the following keywords: "ALK" and "lung", including reviews and our own work. Key Content and Findings Testing rates have not reached 85% among patients' candidates to ALK inhibition. The advantages and disadvantages of the different analytical options [immunohistochemistry (IHC), fluorescence in situ hybridization (FISH), real-time polymerase chain reaction and next-generation sequencing (NGS)] are discussed. The key factor for success in ALK testing is a deep understanding of the concept of "molecular redundancy". This notion has been recommended and endorsed by all the major professional organizations in the field and can be summarized as follows: "laboratories should ensure that test results that are unexpected, discordant, equivocal, or otherwise of low confidence are confirmed or resolved using an alternative method or sample". In-depth knowledge of the different ALK testing methodologies can help clinical and molecular tumor boards implement and maintain sensible algorithms for a rapid and effective detection of predictive biomarkers in patients with NSCLC. Conclusions Multimodality testing has the potential to increase both the testing rate and the accuracy of ALK fusion identification.
Collapse
Affiliation(s)
- Susana Hernandez
- Pathology Department, Hospital Universitario 12 de Octubre, Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Esther Conde
- Pathology Department, Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i+12), Ciberonc, Madrid, Spain
| | - Marta Alonso
- Pathology Department, Hospital Universitario 12 de Octubre, Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | | | - Helena Bote de Cabo
- Oncology Department, Hospital Universitario 12 de Octubre, H12O-CNIO Lung Cancer Clinical Research Unit, Research Institute Hospital 12 de Octubre (i+12)/Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Jon Zugazagoitia
- Oncology Department, Hospital Universitario 12 de Octubre, H12O-CNIO Lung Cancer Clinical Research Unit, Research Institute Hospital 12 de Octubre (i+12)/Spanish National Cancer Research Center (CNIO), Ciberonc, Madrid, Spain
| | - Luis Paz-Ares
- Oncology Department, Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, H12O-CNIO Lung Cancer Clinical Research Unit, Research Institute Hospital 12 de Octubre (i+12)/Spanish National Cancer Research Center (CNIO), Ciberonc, Madrid, Spain
| | - Fernando Lopez-Rios
- Pathology Department, Hospital Universitario 12 de Octubre, Universidad Complutense de Madrid, Research Institute Hospital 12 de Octubre (i+12), Ciberonc, Madrid, Spain
| |
Collapse
|
84
|
Wang Y, Chen D, Liu Y, Shi D, Duan C, Li J, Shi X, Zhang Y, Yu Z, Sun N, Wang W, Ma Y, Xu X, Otkur W, Liu X, Xia T, Qi H, Piao HL, Liu HX. Multidirectional characterization of cellular composition and spatial architecture in human multiple primary lung cancers. Cell Death Dis 2023; 14:462. [PMID: 37488117 PMCID: PMC10366158 DOI: 10.1038/s41419-023-05992-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/26/2023]
Abstract
Multiple primary lung cancers (MPLCs) pose diagnostic and therapeutic challenges in clinic. Here, we orchestrated the cellular and spatial architecture of MPLCs by combining single-cell RNA-sequencing and spatial transcriptomics. Notably, we identified a previously undescribed sub-population of epithelial cells termed as CLDN2+ alveolar type II (AT2) which was specifically enriched in MPLCs. This subtype was observed to possess a relatively stationary state, play a critical role in cellular communication, aggregate spatially in tumor tissues, and dominate the malignant histopathological patterns. The CLDN2 protein expression can help distinguish MPLCs from intrapulmonary metastasis and solitary lung cancer. Moreover, a cell surface receptor-TNFRSF18/GITR was highly expressed in T cells of MPLCs, suggesting TNFRSF18 as one potential immunotherapeutic target in MPLCs. Meanwhile, high inter-lesion heterogeneity was observed in MPLCs. These findings will provide insights into diagnostic biomarkers and therapeutic targets and advance our understanding of the cellular and spatial architecture of MPLCs.
Collapse
Affiliation(s)
- Yawei Wang
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- Department of Thoracic Surgery, Affiliated Hospital of Qingdao University, 266000, Qingdao, China
| | - Di Chen
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Yu Liu
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Daiwang Shi
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Chao Duan
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Jinghan Li
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Xiang Shi
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Yong Zhang
- Department of Pathology, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Zhanwu Yu
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Nan Sun
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Wei Wang
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Yegang Ma
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China
| | - Xiaohan Xu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, 110122, Shenyang, China
| | - Wuxiyar Otkur
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Xiaolong Liu
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Tian Xia
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Huan Qi
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, 116023, Dalian, China.
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China.
- Department of Biochemistry & Molecular Biology, School of Life Sciences, China Medical University, 110122, Shenyang, China.
| | - Hong-Xu Liu
- Department of Thoracic Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of China Medical University, 110042, Shenyang, China.
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, 110042, Shenyang, China.
| |
Collapse
|
85
|
Stanzione B, Del Conte A, Bertoli E, De Carlo E, Revelant A, Spina M, Bearz A. Therapeutical Options in ROS1-Rearranged Advanced Non Small Cell Lung Cancer. Int J Mol Sci 2023; 24:11495. [PMID: 37511255 PMCID: PMC10380455 DOI: 10.3390/ijms241411495] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Revised: 07/11/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
ROS proto-oncogene 1 (ROS1) rearrangements occur in 0.9-2.6% of patients with non small cell lung cancer (NSCLC), conferring sensitivity to treatment with specific tyrosine-kinase inhibitors (TKI). Crizotinib, a first-generation TKI, was the first target-therapy approved for the first-line treatment of ROS1-positive NSCLC. Recently, entrectinib, a multitarget inhibitor with an anti-ROS1 activity 40 times more potent than crizotinib and better activity on the central nervous system (CNS), received approval for treatment-naive patients. After a median time-to-progression of 5.5-20 months, resistance mechanisms can occur, leading to tumor progression. Therefore, newer generation TKI with greater potency and brain penetration have been developed and are currently under investigation. This review summarizes the current knowledge on clinicopathological characteristics of ROS1-positive NSCLC and its therapeutic options.
Collapse
Affiliation(s)
- Brigida Stanzione
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessandro Del Conte
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Elisa Bertoli
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
- Department of Medicine (DAME), University of Udine, 33100 Udine, Italy
| | - Elisa De Carlo
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alberto Revelant
- Department of Radiotherapy, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Michele Spina
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| | - Alessandra Bearz
- Department of Medical Oncology, Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy
| |
Collapse
|
86
|
Doe-Tetteh SA, Camp SY, Reales D, Crowdis J, Noronha AM, Wolff B, Alano T, Galle J, Duygu Selcuklu S, Viale A, Socci ND, Liu YL, Tew WP, Aghajanian C, Ladanyi M, He MX, AlDubayan SH, Mazor RD, Shpilberg O, Hershkovitz-Rokah O, Riancho JA, Hernandez JL, Gonzalez-Vela MC, Buthorn JJ, Wilson M, Webber AE, Yabe M, Petrova-Drus K, Rosenblum M, Durham BH, Abdel-Wahab O, Berger MF, Donoghue MT, Kung AL, Bender JG, Shukla NN, Funt SA, Dogan A, Soslow RA, Al-Ahmadie H, Feldman DR, Van Allen EM, Diamond EL, Solit DB. Overcoming Barriers to Tumor Genomic Profiling through Direct-to-Patient Outreach. Clin Cancer Res 2023; 29:2445-2455. [PMID: 36862133 PMCID: PMC10330105 DOI: 10.1158/1078-0432.ccr-22-3247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 01/05/2023] [Accepted: 02/28/2023] [Indexed: 03/03/2023]
Abstract
PURPOSE To overcome barriers to genomic testing for patients with rare cancers, we initiated a program to offer free clinical tumor genomic testing worldwide to patients with select rare cancer subtypes. EXPERIMENTAL DESIGN Patients were recruited through social media outreach and engagement with disease-specific advocacy groups, with a focus on patients with histiocytosis, germ cell tumors (GCT), and pediatric cancers. Tumors were analyzed using the MSK-IMPACT next-generation sequencing assay with the return of results to patients and their local physicians. Whole-exome recapture was performed for female patients with GCTs to define the genomic landscape of this rare cancer subtype. RESULTS A total of 333 patients were enrolled, and tumor tissue was received for 288 (86.4%), with 250 (86.8%) having tumor DNA of sufficient quality for MSK-IMPACT testing. Eighteen patients with histiocytosis have received genomically guided therapy to date, of whom 17 (94%) have had clinical benefit with a mean treatment duration of 21.7 months (range, 6-40+). Whole-exome sequencing of ovarian GCTs identified a subset with haploid genotypes, a phenotype rarely observed in other cancer types. Actionable genomic alterations were rare in ovarian GCT (28%); however, 2 patients with ovarian GCTs with squamous transformation had high tumor mutational burden, one of whom had a complete response to pembrolizumab. CONCLUSIONS Direct-to-patient outreach can facilitate the assembly of cohorts of rare cancers of sufficient size to define their genomic landscape. By profiling tumors in a clinical laboratory, results could be reported to patients and their local physicians to guide treatment. See related commentary by Desai and Subbiah, p. 2339.
Collapse
Affiliation(s)
- Seyram A. Doe-Tetteh
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Sabrina Y. Camp
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
| | - Dalicia Reales
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Jett Crowdis
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
| | - Anne Marie Noronha
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Bernadette Wolff
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Department of Nursing, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Tina Alano
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Department of Nursing, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Jesse Galle
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - S. Duygu Selcuklu
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Agnes Viale
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Nicholas D. Socci
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Ying L. Liu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - William P. Tew
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Carol Aghajanian
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Marc Ladanyi
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Meng Xiao He
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
- Harvard Graduate Program in Biophysics, Boston, MA, 02115, USA
| | - Saud H. AlDubayan
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
- College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Roei David Mazor
- Clinic of Histiocytic Neoplasms, Institute of Hematology, Assuta Medical Center, Tel Aviv, Israel
| | - Ofer Shpilberg
- Clinic of Histiocytic Neoplasms, Institute of Hematology, Assuta Medical Center, Tel Aviv, Israel
- Adelson School of Medicine, Ariel University, Ariel, Israel
| | - Oshrat Hershkovitz-Rokah
- Department of Molecular Biology, Faculty of Natural Sciences, Ariel University, Ariel, Israel
- Translational Research Lab, Assuta Medical Center, Tel-Aviv, Israel
| | - Jose A. Riancho
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Jose L. Hernandez
- Department of Internal Medicine, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - M. Carmen Gonzalez-Vela
- Department of Pathology, Hospital U.M. Valdecilla, University of Cantabria, IDIVAL, Santander, Spain
| | - Justin J. Buthorn
- Department of Neurology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Manda Wilson
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Amy E. Webber
- Bioinformatics Core, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Mariko Yabe
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Kseniya Petrova-Drus
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Marc Rosenblum
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Benjamin H. Durham
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael F. Berger
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Mark T.A. Donoghue
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Andrew L. Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Julia Glade Bender
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Neerav N. Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Samuel A. Funt
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Ahmet Dogan
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Robert A. Soslow
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Hikmat Al-Ahmadie
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
| | - Darren R. Feldman
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - Eliezer M. Van Allen
- Department of Medical Oncology, Dana Farber Cancer Institute
- Cancer Program, Broad Institute of MIT and Harvard
| | - Eli L. Diamond
- Department of Neurology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| | - David B. Solit
- Marie-Josée and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, NY, New York, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Joan & Sanford I. Weill Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
87
|
Qiu T, Zhi X, Ren S. Recent advance of next-generation sequencing in patients with lung cancer. Expert Rev Mol Diagn 2023; 23:959-970. [PMID: 37750512 DOI: 10.1080/14737159.2023.2260755] [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: 04/07/2023] [Accepted: 09/15/2023] [Indexed: 09/27/2023]
Abstract
INTRODUCTION Precision medicine based on the driver genes mutation status is the current systemic therapeutic paradigm in patients with lung cancer. Next-generation sequencing (NGS) has emerged as a powerful platform for molecular diagnosis by virtue of high-throughput and massively parallel sequencing. Liquid biopsy also enabled the dynamic monitoring and comprehensive profiling of lung cancer in a noninvasive manner. However, challenges remain in the field of technology and clinical applications, especially in the era of immunotherapy. AREAS COVERED Here, we update the role of NGS in the context of lung cancer screening, molecular diagnosis, predictive and prognostic biomarkers, and guiding personalized treatment. EXPERT OPINION The NGS application for actable genomic alternation has greatly changed the therapeutic landscape in patients with lung cancer including perioperative setting and advanced stage. Meanwhile, emerging evidence has shown the potential of other applications such as early screening and detection, and MRD. However, challenges remain such as the lack of standardized protocols across different platforms and bioinformatics analysis pipelines, and the complexity of interpreting and leveraging numerous genomic mutation messages for therapy selection. Future research is needed to overcome these challenges and expand the applications of NGS to other aspects such as immunotherapy.
Collapse
Affiliation(s)
- Tianyu Qiu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinxin Zhi
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shengxiang Ren
- Department of Medical Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
88
|
Rao W, Liu Y, Li Y, Guo L, Qiu T, Dong L, Ying J, Li W. Potential unreliability of ALK variant allele frequency in the efficacy prediction of targeted therapy in NSCLC. Front Med 2023; 17:493-502. [PMID: 37010729 DOI: 10.1007/s11684-022-0946-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/28/2022] [Indexed: 04/04/2023]
Abstract
Anaplastic lymphoma kinase (ALK) is the most common fusion gene involved in non-small cell lung cancer (NSCLC), and remarkable response has been achieved with the use of ALK tyrosine kinase inhibitors (ALK-TKIs). However, the clinical efficacy is highly variable. Pre-existing intratumoral heterogeneity (ITH) has been proven to contribute to the poor treatment response and the resistance to targeted therapies. In this work, we investigated whether the variant allele frequencies (VAFs) of ALK fusions can help assess ITH and predict targeted therapy efficacy. Through the application of next-generation sequencing (NGS), 7.2% (326/4548) of patients were detected to be ALK positive. On the basis of the adjusted VAF (adjVAF, VAF normalization for tumor purity) of four different threshold values (adjVAF < 50%, 40%, 30%, or 20%), the association of ALK subclonality with crizotinib efficacy was assessed. Nonetheless, no statistical association was observed between median progression-free survival (PFS) and ALK subclonality assessed by adjVAF, and a poor correlation of adjVAF with PFS was found among the 85 patients who received first-line crizotinib. Results suggest that the ALK VAF determined by hybrid capture-based NGS is probably unreliable for ITH assessment and targeted therapy efficacy prediction in NSCLC.
Collapse
Affiliation(s)
- Wei Rao
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yutao Liu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yan Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Lin Dong
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| | - Weihua Li
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
89
|
Gibbs SN, Peneva D, Cuyun Carter G, Palomares MR, Thakkar S, Hall DW, Dalglish H, Campos C, Yermilov I. Comprehensive Review on the Clinical Impact of Next-Generation Sequencing Tests for the Management of Advanced Cancer. JCO Precis Oncol 2023; 7:e2200715. [PMID: 37285561 DOI: 10.1200/po.22.00715] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Revised: 03/14/2023] [Accepted: 04/05/2023] [Indexed: 06/09/2023] Open
Abstract
PURPOSE This review summarizes the published evidence on the clinical impact of using next-generation sequencing (NGS) tests to guide management of patients with cancer in the United States. METHODS We performed a comprehensive literature review to identify recent English language publications that presented progression-free survival (PFS) and overall survival (OS) of patients with advanced cancer receiving NGS testing. RESULTS Among 6,475 publications identified, 31 evaluated PFS and OS among subgroups of patients who received NGS-informed cancer management. PFS and OS were significantly longer among patients who were matched to targeted treatment in 11 and 16 publications across tumor types, respectively. CONCLUSION Our review indicates that NGS-informed treatment can have an impact on survival across tumor types.
Collapse
Affiliation(s)
- Sarah N Gibbs
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| | - Desi Peneva
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| | | | | | | | | | - Hannah Dalglish
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| | - Cynthia Campos
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| | - Irina Yermilov
- Partnership for Health Analytic Research (PHAR), LLC, Beverly Hills, CA
| |
Collapse
|
90
|
de Traux de Wardin H, Xu B, Dermawan JK, Smith MH, Wolden SL, Antonescu CR, Wexler LH. Extremity Rhabdomyosarcoma-An Integrated Clinicopathologic and Genomic Study to Improve Risk Stratification. JCO Precis Oncol 2023; 7:e2200705. [PMID: 37315267 PMCID: PMC10309566 DOI: 10.1200/po.22.00705] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/30/2023] [Accepted: 05/08/2023] [Indexed: 06/16/2023] Open
Abstract
PURPOSE Extremity rhabdomyosarcoma (RMS) is associated with a very poor outcome compared with other sites, mainly because of its high incidence of alveolar histology and regional lymph node involvement. To better define prognostic markers in this clinical subset, we investigated our experience of 61 patients with extremity RMS treated at our tertiary cancer center for the past 2 decades. PATIENTS AND METHODS The patients had a median age of 8 years at diagnosis, equal gender distribution, and two-thirds occurred in the lower extremity. Most (85%) patients had FOXO1 fusion-positive alveolar RMS (ARMS), with 70% having a PAX3::FOXO1 transcript. Remaining were seven patients with fusion-negative embryonal RMS (ERMS) and two with MYOD1-mutant spindle cell/sclerosing RMS (SRMS). In 40% of the patients, material was available for DNA-based targeted sequencing using MSK-IMPACT cancer gene panel. RESULTS One-third of patients presented with localized disease at diagnosis while the remaining had regional nodal (18%) or distant metastases (51%). Metastatic disease, high-risk group, and age 10 years or older significantly affected the overall survival (OS; hazard ratio [HR], 2.68 [P = .004], 2.78 [P = .010] and 2.26 [P = .034], respectively). Although the presence of metastatic disease had a dismal impact on 5-year EFS and OS (19% and 29%, respectively), nodal involvement had a comparatively lower impact on 5-year EFS and 5-year OS (43% and 66%, respectively). PAX3::FOXO1 ARMS had worse prognosis and afflicted older children compared with PAX7::FOXO1 (HR = 3.45, P = .016). The most common events in the ARMS group included MED12 alterations, CDK4 amplifications, and CDKN2A deletions (8%-17%). The latter two abnormalities were mutually exclusive, enriched for acral and high-risk lesions, and correlated with poor outcome on OS (P = .02). CONCLUSION Our data provide rationale for considering the integration of molecular abnormalities to refine risk stratification in extremity RMS.
Collapse
Affiliation(s)
| | - Bin Xu
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Mariel H. Smith
- Pediatric Hematology/Oncology, Massachusetts General Hospital for Children, Boston, MA
| | - Suzanne L. Wolden
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY
| | | | - Leonard H. Wexler
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
91
|
Dyrbekk APH, Warsame AA, Suhrke P, Ludahl MO, Moe JO, Eide IJZ, Lund-Iversen M, Brustugun OT. "Evaluation of ROS1 expression and rearrangements in a large cohort of early-stage lung cancer". Diagn Pathol 2023; 18:70. [PMID: 37237384 DOI: 10.1186/s13000-023-01357-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 05/18/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND ROS1 fusion is an infrequent, but attractive target for therapy in patients with metastatic non- small-cell lung cancer. In studies on mainly late-stage disease, the prevalence of ROS1 fusions is about 1-3%. In early-stage lung cancer ROS1 might also provide a fruitful target for neoadjuvant or adjuvant therapy. In the present study, we investigated the prevalence of ROS1 fusion in a Norwegian cohort of early-stage lung cancer. We also explored whether positive ROS1 immunohistochemical (IHC) stain was associated with certain mutations, clinical characteristics and outcomes. METHODS The study was performed using biobank material from 921 lung cancer patients including 542 patients with adenocarcinoma surgically resected during 2006-2018. Initially, we screened the samples with two different IHC clones (D4D6 and SP384) targeting ROS1. All samples that showed more than weak or focal staining, as well as a subgroup of negative samples, were analyzed with ROS1 fluorescence in situ hybridization (FISH) and next-generation sequencing (NGS) with a comprehensive NGS DNA and RNA panel. Positive ROS1-fusion was defined as those samples positive in at least two of the three methods (IHC, FISH, NGS). RESULTS Fifty cases were IHC positive. Of these, three samples were both NGS and FISH-positive and considered positive for ROS1 fusion. Two more samples were FISH positive only, and whilst IHC and NGS were negative. These were also negative with Reverse Transcription quantitative real time Polymerase Chain Reaction (RT-qPCR). The prevalence of ROS1 fusion in adenocarcinomas was 0.6%. All cases with ROS1 fusion had TP53 mutations. IHC-positivity was associated with adenocarcinoma. Among SP384-IHC positive cases we also found an association with never smoking status. There was no association between positive IHC and overall survival, time to relapse, age, stage, sex or pack-year of smoking. CONCLUSIONS ROS1 seems to be less frequent in early-stage disease than in advanced stages. IHC is a sensitive, but less specific method and the results need to be confirmed with another method like FISH or NGS.
Collapse
Affiliation(s)
- Anne Pernille Harlem Dyrbekk
- University of Oslo, NO-0316, Oslo, Norway.
- Department of Pathology, Vestfold Hospital Trust, NO-3103, Tonsberg, Norway.
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, NO-0310, Oslo, Norway.
| | - Abdirashid Ali Warsame
- Department of Pathology, Oslo University Hospital, The Norwegian Radium Hospital, NO-0310, Oslo, Norway
| | - Pål Suhrke
- Department of Pathology, Vestfold Hospital Trust, NO-3103, Tonsberg, Norway
| | - Marianne Odnakk Ludahl
- Department of Microbiology/ Division for Genetechnology, Vestfold Hospital Trust, NO-3103, Tonsberg, Norway
| | - Joakim Oliu Moe
- Department of Internal Medicine, Vestfold Hospital Trust, NO-3103, Tonsberg, Norway
| | - Inger Johanne Zwicky Eide
- University of Oslo, NO-0316, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, NO-0310, Oslo, Norway
- Department of Oncology, Vestre Viken Hospital Trust, NO-3004, Drammen, Norway
| | - Marius Lund-Iversen
- Department of Pathology, Oslo University Hospital, The Norwegian Radium Hospital, NO-0310, Oslo, Norway
| | - Odd Terje Brustugun
- University of Oslo, NO-0316, Oslo, Norway
- Department of Cancer Genetics, Institute for Cancer Research, The Norwegian Radium Hospital, NO-0310, Oslo, Norway
- Department of Oncology, Vestre Viken Hospital Trust, NO-3004, Drammen, Norway
| |
Collapse
|
92
|
Heydt C, Ihle MA, Merkelbach-Bruse S. Overview of Molecular Detection Technologies for MET in Lung Cancer. Cancers (Basel) 2023; 15:cancers15112932. [PMID: 37296895 DOI: 10.3390/cancers15112932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/12/2023] Open
Abstract
MET tyrosine kinase receptor pathway activation has become an important actionable target in solid tumors. Aberrations in the MET proto-oncogene, including MET overexpression, the activation of MET mutations, MET mutations that lead to MET exon 14 skipping, MET gene amplifications, and MET fusions, are known to be primary and secondary oncogenic drivers in cancer; these aberrations have evolved as predictive biomarkers in clinical diagnostics. Thus, the detection of all known MET aberrations in daily clinical care is essential. In this review, current molecular technologies for the detection of the different MET aberrations are highlighted, including the benefits and drawbacks. In the future, another focus will be on the standardization of detection technologies for the delivery of reliable, quick, and affordable tests in clinical molecular diagnostics.
Collapse
Affiliation(s)
- Carina Heydt
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Michaela Angelika Ihle
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Sabine Merkelbach-Bruse
- Faculty of Medicine, Institute of Pathology, University Hospital Cologne, University of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| |
Collapse
|
93
|
Wu S, Liu Y, Li K, Liang Z, Zeng X. Molecular and cytogenetic features of NTRK fusions enriched in BRAF and RET double-negative papillary thyroid cancer. J Mol Diagn 2023:S1525-1578(23)00106-X. [PMID: 37236546 DOI: 10.1016/j.jmoldx.2023.04.007] [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] [Revised: 02/17/2023] [Accepted: 04/10/2023] [Indexed: 05/28/2023] Open
Abstract
Rare NTRK-driven malignant neoplasms can be effectively inhibited by anti-TRK agents. The discovery of NTRK1/2/3-rich tumours in papillary thyroid cancer (PTC) patients is a precondition for the rapid identification of NTRK fusion tumours. Knowledge of NTRK gene activation is critical to accurately detect NTRK status. A total of 229 BRAF V600E-negative samples from PTC patients were analysed in this study. Break-apart fluorescence in situ hybridisation (FISH) was performed to detect RET fusion. NTRK status was analysed using FISH, DNA- and RNA-based next-generation sequencing (NGS), and quantitative reverse transcription-polymerase chain reaction (RT-qPCR). In 128 BRAF and RET double-negative cases, 56 (43.8%, 56/128) NTRK rearrangement tumours were found, including 1 NTRK2, 16 NTRK1, and 39 NTRK3 fusions. Two novel NTRK fusions, EZR::NTRK1 and EML4::NTRK2, was found in the NTRK rearrangement tumors.Dominant break-apart and extra 3' signal patterns accounted for 89.3% (50/56) and 5.4% (3/56) of all NTRK-positive cases, respectively, as determined by FISH. In our cohort, there were 2.3% (3/128) FISH false-negative and 3.1% (4/128) FISH false-positive cases identified. NTRK fusions are highly recurrent in BRAF and RET double-negative PTCs. FISH or RNA-based NGS is a reliable detection approach. NTRK rearrangement can be precisely, rapidly, and economically detected based on the developed optimal algorithm.
Collapse
Affiliation(s)
- Shafei Wu
- Department of Pathology, Peking Union Medical College Hospital, and Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yuanyuan Liu
- Department of Pathology, Peking Union Medical College Hospital, and Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Kaimi Li
- Department of Pathology, Peking Union Medical College Hospital, and Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zhiyong Liang
- Department of Pathology, Peking Union Medical College Hospital, and Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| | - Xuan Zeng
- Department of Pathology, Peking Union Medical College Hospital, and Molecular Pathology Research Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
94
|
Hofman V, Goffinet S, Bontoux C, Long-Mira E, Lassalle S, Ilié M, Hofman P. A Real-World Experience from a Single Center (LPCE, Nice, France) Highlights the Urgent Need to Abandon Immunohistochemistry for ROS1 Rearrangement Screening of Advanced Non-Squamous Non-Small Cell Lung Cancer. J Pers Med 2023; 13:jpm13050810. [PMID: 37240980 DOI: 10.3390/jpm13050810] [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: 03/20/2023] [Revised: 05/04/2023] [Accepted: 05/08/2023] [Indexed: 05/28/2023] Open
Abstract
The detection of ROS1 rearrangements in metastatic non-squamous non-small cell lung carcinoma (NS-NSCLC) permits administration of efficient targeted therapy. Detection is based on a testing algorithm associated with ROS1 immunohistochemistry (IHC) screening followed by ROS1 FISH and/or next generation sequencing (NGS) to confirm positivity. However, (i) ROS1 rearrangements are rare (1-2% of NS-NSCLC), (ii) the specificity of ROS1 IHC is not optimal, and (iii) ROS1 FISH is not widely available, making this algorithm challenging to interpret time-consuming. We evaluated RNA NGS, which was used as reflex testing for ROS1 rearrangements in NS-NSCLC with the aim of replacing ROS1 IHC as a screening method. ROS1 IHC and RNA NGS were prospectively performed in 810 NS-NSCLC. Positive results were analyzed by ROS1 FISH. ROS1 IHC was positive in 36/810 (4.4%) cases that showed variable staining intensity while NGS detected ROS1 rearrangements in 16/810 (1.9%) cases. ROS1 FISH was positive in 15/810 (1.8%) of ROS1 IHC positive cases and in all positive ROS1 NGS cases. Obtaining both ROS1 IHC and ROS1 FISH reports took an average of 6 days, while obtaining ROS1 IHC and RNA NGS reports took an average of 3 days. These results showed that systematic screening for the ROS1 status using IHC must be replaced by NGS reflex testing.
Collapse
Affiliation(s)
- Véronique Hofman
- Laboratory of Clinical and Experimental Pathology, University Côte d'Azur, FHU OncoAge, Pasteur Hospital, 06000 Nice, France
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, 06000 Nice, France
- Team 4, IRCAN Inserm U1081, CNRS 7284, Université Côte d'Azur, 06100 Nice, France
| | - Samantha Goffinet
- Laboratory of Clinical and Experimental Pathology, University Côte d'Azur, FHU OncoAge, Pasteur Hospital, 06000 Nice, France
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, 06000 Nice, France
| | - Christophe Bontoux
- Laboratory of Clinical and Experimental Pathology, University Côte d'Azur, FHU OncoAge, Pasteur Hospital, 06000 Nice, France
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, 06000 Nice, France
- Team 4, IRCAN Inserm U1081, CNRS 7284, Université Côte d'Azur, 06100 Nice, France
| | - Elodie Long-Mira
- Laboratory of Clinical and Experimental Pathology, University Côte d'Azur, FHU OncoAge, Pasteur Hospital, 06000 Nice, France
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, 06000 Nice, France
- Team 4, IRCAN Inserm U1081, CNRS 7284, Université Côte d'Azur, 06100 Nice, France
| | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, University Côte d'Azur, FHU OncoAge, Pasteur Hospital, 06000 Nice, France
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, 06000 Nice, France
- Team 4, IRCAN Inserm U1081, CNRS 7284, Université Côte d'Azur, 06100 Nice, France
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, University Côte d'Azur, FHU OncoAge, Pasteur Hospital, 06000 Nice, France
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, 06000 Nice, France
- Team 4, IRCAN Inserm U1081, CNRS 7284, Université Côte d'Azur, 06100 Nice, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, University Côte d'Azur, FHU OncoAge, Pasteur Hospital, 06000 Nice, France
- Hospital-Integrated Biobank (BB-0033-00025), Pasteur Hospital, 06000 Nice, France
- Team 4, IRCAN Inserm U1081, CNRS 7284, Université Côte d'Azur, 06100 Nice, France
| |
Collapse
|
95
|
Albitar M, Zhang H, Charifa A, Ip A, Ma W, McCloskey J, Donato M, Siegel D, Waintraub S, Gutierrez M, Pecora A, Goy A. Combining cell-free RNA with cell-free DNA in liquid biopsy for hematologic and solid tumors. Heliyon 2023; 9:e16261. [PMID: 37251903 PMCID: PMC10208940 DOI: 10.1016/j.heliyon.2023.e16261] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 05/07/2023] [Accepted: 05/11/2023] [Indexed: 05/31/2023] Open
Abstract
Current use of liquid biopsy is based on cell-free DNA (cfDNA) and the evaluation of mutations or methylation pattern. However, expressed RNA can capture mutations, changes in expression levels due to methylation, and provide information on cell of origin, growth, and proliferation status. We developed an approach to isolate cell-free total nucleic acid (cfDNA) and used targeted next generation sequencing to sequence cell-free RNA (cfRNA) and cfDNA as new approach in liquid biopsy. We demonstrate that cfRNA is overall more sensitive than cfDNA in detecting mutations. We show that cfRNA is reliable in detecting fusion genes and cfDNA is reliable in detecting chromosomal gains and losses. cfRNA levels of various solid tumor biomarkers were significantly higher (P < 0.0001) in samples from solid tumors as compared with normal control. Similarly, cfRNA lymphoid markers and cfRNA myeloid markers were all higher in lymphoid and myeloid neoplasms, respectively as compared with control (P < 0.0001). Using machine learning we demonstrate cfRNA was highly predictive of diagnosis (AUC >0.98) of solid tumors, B-cell lymphoid neoplasms, T-cell lymphoid neoplasms, and myeloid neoplasms. In evaluating the host immune system, cfRNA CD4:CD8B and CD3D:CD19 ratios in normal controls were as expected (median: 5.92 and 6.87, respectively) and were significantly lower in solid tumors (P < 0.0002). This data suggests that liquid biopsy combining analysis of cfRNA with cfDNA is practical and may provide helpful information in predicting genomic abnormalities, diagnosis of neoplasms and evaluating both the tumor biology and the host response.
Collapse
Affiliation(s)
- Maher Albitar
- Genomic Testing Cooperative, LCA, Irvine, CA, 92618, USA
| | - Hong Zhang
- Genomic Testing Cooperative, LCA, Irvine, CA, 92618, USA
| | - Ahmad Charifa
- Genomic Testing Cooperative, LCA, Irvine, CA, 92618, USA
| | - Andrew Ip
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Wanlong Ma
- Genomic Testing Cooperative, LCA, Irvine, CA, 92618, USA
| | - James McCloskey
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Michele Donato
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - David Siegel
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Stanley Waintraub
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Martin Gutierrez
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Andrew Pecora
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| | - Andre Goy
- John Theurer Cancer Center at Hackensack University Medical Center, Hackensack, NJ, 07601, USA
| |
Collapse
|
96
|
Yu Y, Zhou J, Li X, Goto K, Min X, Nishino K, Cui J, Wu L, Sakakibara J, Shu Y, Dong X, Li L, Yoneshima Y, Zhou C, Li X, Zhang Y, Huang D, Zang A, Zhang W, Wang X, Zhang L, Bai C, Fang J, Cao L, Zhao Y, Yu Y, Shi M, Zhong D, Li F, Li M, Wu Q, Zhou J, Sun M, Lu S. Gumarontinib in patients with non-small-cell lung cancer harbouring MET exon 14 skipping mutations: a multicentre, single-arm, open-label, phase 1b/2 trial. EClinicalMedicine 2023; 59:101952. [PMID: 37096188 PMCID: PMC10121392 DOI: 10.1016/j.eclinm.2023.101952] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 03/21/2023] [Accepted: 03/21/2023] [Indexed: 04/26/2023] Open
Abstract
Background Approximately 3-4% of patients with non-small-cell lung cancer (NSCLC) have MET exon 14 (METex14) skipping mutations. We report primary results from the phase 2 stage of a phase 1b/2 study of gumarontinib, a selective, potent, oral MET inhibitor, in patients with METex14 skipping mutation-positive (METex14-positive) NSCLC. Methods The single-arm, multicentre, open-label, phase 2 stage of the GLORY study was conducted at 42 centres across China and Japan. Adults with locally advanced or metastatic METex14-positive NSCLC received oral gumarontinib 300 mg once daily in continuous 21-day cycles until disease progression, intolerable toxicity, or withdrawal of consent. Eligible patients had failed one or two prior lines of therapy (not including a MET inhibitor), were ineligible for/refused chemotherapy, and had no genetic alterations targetable with standard therapies. The primary endpoint was objective response rate in patients with a valid baseline tumour assessment, by blinded independent review. The study was registered at ClinicalTrials.gov (NCT04270591). Findings Between Aug 2, 2019 and Apr 28, 2021, 84 patients were enrolled and received gumarontinib (median follow-up 13.5 months [IQR 8.7-17.1]), at data cut-off (Apr 28, 2022) five patients whose METex14 status could not be confirmed by a central laboratory were excluded from the efficacy analysis. The objective response rate was 66% (95% CI 54-76) overall (n = 79), 71% (95% CI 55-83) in treatment-naïve patients (n = 44), and 60% (95% CI 42-76) in previously-treated patients (n = 35). The most common treatment-related adverse events (any grade) were oedema (67/84 patients, 80%) and hypoalbuminuria (32/84, 38%). Grade ≥3 treatment-emergent adverse events occurred in 45 (54%) patients. Treatment-related adverse events leading to permanent discontinuation occurred in 8% (7/84) of patients. Interpretation Gumarontinib monotherapy had durable antitumour activity with manageable toxicity in patients with locally advanced or metastatic METex14-positive NSCLC when used in first line or later. Funding Haihe Biopharma Co., Ltd. Supported in part by grants from the National Science and Technology Major Project of China for "Clinical Research of Gumarontinib, a highly selective MET inhibitor" (2018ZX09711002-011-003); the National Natural Science Foundation of China (82030045 to S.L. and 82172633 to YF.Y); Shanghai Municipal Science & Technology Commission Research Project (19411950500 to S.L.); Shanghai Shenkang Action Plan (16CR3005A to S.L.) and Shanghai Chest Hospital Project of Collaborative Innovation (YJXT20190105 to S.L.).
Collapse
Affiliation(s)
- Yongfeng Yu
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianya Zhou
- Respiratory Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xingya Li
- Oncology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Koichi Goto
- Thoracic Oncology, National Cancer Center Hospital East, Kashiwa, Chiba, Japan
| | - Xuhong Min
- Radiology Intervention Department, Anhui Chest Hospital, Hefei, China
| | - Kazumi Nishino
- Thoracic Oncology, Osaka International Cancer Institute, Osaka, Japan
| | - Jiuwei Cui
- Oncology Department, The First Bethune Hospital of Jilin University, Changchun, China
| | - Lin Wu
- Second Department of Thoracic Medicine, Hunan Cancer Hospital, Changsha, China
| | - Jun Sakakibara
- Respiratory Medicine, Hokkaido University Hospital, Sapporo, Japan
| | - Yongqian Shu
- Oncology Department, Jiangsu Province Hospital, Nanjing, China
| | - Xiaorong Dong
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lu Li
- Lung Cancer Center, West China Hospital of Sichuan University, Chengdu, China
| | - Yasuto Yoneshima
- Respiratory Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Chengzhi Zhou
- Oncology, The First Affiliate Hospital of Guangzhou Medical University, Guangzhou, China
| | - Xiaoling Li
- Thoracic Medicine, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yiping Zhang
- Thoracic Oncology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Dingzhi Huang
- Oncology, Tianjin Medical University Cancer Institute & Hospital, Tianjin, China
| | - Aimin Zang
- Medical Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | - Wei Zhang
- Pneumology Department/Institute Office, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiuwen Wang
- Oncology, Qilu Hospital of Shandong University, Jinan, China
| | - Li Zhang
- Department of Respiratory and Critical Care Medicine, Peking Union Medical College Hospital, Beijing, China
| | - Chong Bai
- Pneumology Department, Changhai Hospital of Shanghai, Shanghai, China
| | - Jian Fang
- Thoracic Oncology Second Department, Beijing Cancer Hospital, Beijing, China
| | - Lejie Cao
- Pneumology Department, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Yanqiu Zhao
- Respiratory Department of Internal Medicine, Henan Cancer Hospital, Zhengzhou, China
| | - Yan Yu
- The Third Ward of Respiratory Medicine Department, Harbin Medical University Cancer Hospital, Harbin, China
| | - Meiqi Shi
- Oncology Department, Jiangsu Cancer Hospital, Nanjing, China
| | - Diansheng Zhong
- Internal Medicine-Oncology, Tianjin Medical University General Hospital, Tianjin, China
| | - Fugen Li
- Clinical Science Department, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Meng Li
- Biostatistic and Data Science, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Qiuxia Wu
- Clinical Science Department, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Jun Zhou
- Clinical Science Department, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Minghui Sun
- Clinical Science Department, Haihe Biopharma Co., Ltd, Shanghai, China
| | - Shun Lu
- Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Corresponding author. Department of Medical Oncology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No. 241, Huaihai West Road, Shanghai 200030, China.
| |
Collapse
|
97
|
Chen J, Xu C, Lv J, Lu W, Zhang Y, Wang D, Song Y. Clinical characteristics and targeted therapy of different gene fusions in non-small cell lung cancer: a narrative review. Transl Lung Cancer Res 2023; 12:895-908. [PMID: 37197619 PMCID: PMC10183389 DOI: 10.21037/tlcr-22-566] [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: 07/31/2022] [Accepted: 02/17/2023] [Indexed: 04/07/2023]
Abstract
Background and Objective Lung cancer is the most fatal malignant tumor in the world. Since the discovery of driver genes, targeted therapy has been demonstrated to be superior to traditional chemotherapy and has revolutionized the therapeutic landscape of non-small cell lung cancer (NSCLC). The remarkable success of tyrosine kinase inhibitors (TKIs) in patients with epidermal growth factor receptor (EGFR) mutations and anaplastic lymphoma kinase (ALK) fusions has shifted the treatment from platinum-based combination chemotherapy to targeted therapy. Although the incidence rate of gene fusion is low in NSCLC, it is of great significance in advanced refractory patients. However, the clinical characteristics and the latest treatment progress of patients with gene fusions in lung cancer have not been thoroughly explored. The objective of this narrative review was to summarize the latest research progress of targeted therapy for gene fusion variants in NSCLC to improve understanding for clinicians. Methods We conducted a search of PubMed database and American Society of Clinical Oncology (ASCO), the European Society for Medical Oncology (ESMO), and World Conference on Lung Cancer (WCLC) abstracts meeting proceedings from 1 January 2005 to 31 August 2022 with the following keywords "non-small cell lung cancer", "fusion", "rearrangement", "targeted therapy" and "tyrosine kinase inhibitor". Key Content and Findings We comprehensively listed the targeted therapy of various gene fusions in NSCLC. Fusions of ALK, ROS proto-oncogene 1 (ROS1), and rearranged during transfection proto-oncogene (RET) are relatively more common than others (NTRK fusions, NRG1 fusions, FGFR fusions, etc.). Among ALK-rearranged NSCLC patients treated with crizotinib, alectinib, brigatinib, or ensartinib, the Asian population exhibited a slightly better effect than the non-Asian population in first-line therapy. It was revealed that ceritinib may have a slightly better effect in the non-Asian ALK-rearranged population as first-line therapy. The effect of crizotinib might be similar in Asians and non-Asians with ROS1-fusion-positive NSCLC in first-line therapy. The non-Asian population were shown to be more likely to be treated with selpercatinib and pralsetinib for RET-rearranged NSCLC than the Asian population. Conclusions The present report summarizes the current state of fusion gene research and the associated therapeutic methods to improve understanding for clinicians, but how to better overcome drug resistance remains a problem that needs to be explored.
Collapse
Affiliation(s)
- Jiayan Chen
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
| | - Chunwei Xu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Jiawen Lv
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Wanjun Lu
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yixue Zhang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Dong Wang
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yong Song
- Department of Respiratory Medicine, Jinling Hospital, Nanjing Medical University, Nanjing, China
- Department of Respiratory Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
98
|
Brea E, Rotow J. Targeted Therapy for Non–Small Cell Lung Cancer. Hematol Oncol Clin North Am 2023; 37:575-594. [PMID: 37024384 DOI: 10.1016/j.hoc.2023.02.009] [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: 04/08/2023]
Abstract
This article provides an updated review of the management of oncogene-driven non-small cell lung cancer. The use of targeted therapies for lung cancer driven by EGFR, ALK, ROS1, RET, NTRK, HER2, BRAF, MET, and KRAS are discussed, both in the first-line setting and in the setting of acquired resistance.
Collapse
Affiliation(s)
- Elliott Brea
- Department of Medical Oncology, Dana-Farber Cancer Institute, SM353, 450 Brookline Avenue, Boston, MA 02215, USA
| | - Julia Rotow
- Dana-Farber Cancer Institute, 450 Brookline Avenue, DA1240, Boston, MA 02215, USA.
| |
Collapse
|
99
|
Choudhury NJ, Marra A, Sui JSY, Flynn J, Yang SR, Falcon CJ, Selenica P, Schoenfeld AJ, Rekhtman N, Gomez D, Berger MF, Ladanyi M, Arcila M, Rudin CM, Riely GJ, Kris MG, Heller G, Reis-Filho JS, Yu HA. Molecular Biomarkers of Disease Outcomes and Mechanisms of Acquired Resistance to First-Line Osimertinib in Advanced EGFR-Mutant Lung Cancers. J Thorac Oncol 2023; 18:463-475. [PMID: 36494075 PMCID: PMC10249779 DOI: 10.1016/j.jtho.2022.11.022] [Citation(s) in RCA: 57] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/14/2022] [Accepted: 11/18/2022] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Preferred first-line treatment for patients with metastatic EGFR-mutant lung cancer is osimertinib, yet it is not known whether patient outcomes may be improved by identifying and intervening on molecular markers associated with therapeutic resistance. METHODS All patients with metastatic EGFR-mutant lung cancer treated with first-line osimertinib at the Memorial Sloan Kettering Cancer Center (n = 327) were identified. Available pretreatment and postprogression tumor samples underwent targeted gene panel sequencing and mutational signature analysis using SigMA algorithm. Progression-free survival (PFS) and overall survival were estimated using the Kaplan-Meier method. RESULTS Using multivariate analysis, baseline atypical EGFR (median PFS = 5.8 mo, p < 0.001) and concurrent TP53/RB1 alterations (median PFS = 10.5 mo, p = 0.015) were associated with shorter PFS on first-line osimertinib. Of 95 patients with postprogression biopsies, acquired resistance mechanisms were identified in 52% (off-target, n = 24; histologic transformation, n = 14; on-target, n = 12), with MET amplification (n = 9), small cell lung transformation (n = 7), and acquired EGFR amplification (n = 7), the most frequently identified mechanisms. Although there was no difference in postprogression survival on the basis of identified resistance (p = 0.07), patients with subsequent second-line therapy tailored to postprogression biopsy results had improved postprogression survival (hazard ratio = 0.09, p = 0.006). The paired postprogression tumors had higher tumor mutational burden (p = 0.008) and further dominant APOBEC mutational signatures (p = 0.07) compared with the pretreatment samples. CONCLUSIONS Patients with EGFR-mutant lung cancer treated with first-line osimertinib have improved survival with treatment adaptation on the basis of identified mechanisms of resistance at time of progression using tissue-based genomic analysis. Further survival gains may be achieved using risk-based treatment adaptation of pretreatment genomic alterations.
Collapse
Affiliation(s)
- Noura J Choudhury
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Antonio Marra
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jane S Y Sui
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jessica Flynn
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Soo-Ryum Yang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Christina J Falcon
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pier Selenica
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Adam J Schoenfeld
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Natasha Rekhtman
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Daniel Gomez
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Michael F Berger
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Marie-Josee and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Marc Ladanyi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Marie-Josee and Henry R. Kravis Center for Molecular Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Maria Arcila
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Charles M Rudin
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Gregory J Riely
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Mark G Kris
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Glenn Heller
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jorge S Reis-Filho
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Helena A Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York.
| |
Collapse
|
100
|
Markham JF, Fellowes AP, Green T, Leal JL, Legaie R, Cullerne D, Morris T, John T, Solomon B, Fox SB. Predicting response to immune checkpoint blockade in NSCLC with tumour-only RNA-seq. Br J Cancer 2023; 128:1148-1154. [PMID: 36572732 PMCID: PMC10006283 DOI: 10.1038/s41416-022-02105-w] [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: 07/07/2022] [Revised: 12/04/2022] [Accepted: 12/06/2022] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND Targeted RNA sequencing (RNA-seq) from FFPE specimens is used clinically in cancer for its ability to estimate gene expression and to detect fusions. Using a cohort of NSCLC patients, we sought to determine whether targeted RNA-seq could be used to measure tumour mutational burden (TMB) and the expression of immune-cell-restricted genes from FFPE specimens and whether these could predict response to immune checkpoint blockade. METHODS Using The Cancer Genome Atlas LUAD dataset, we developed a method for determining TMB from tumour-only RNA-seq and showed a correlation with DNA sequencing derived TMB calculated from tumour/normal sample pairs (Spearman correlation = 0.79, 95% CI [0.73, 0.83]. We applied this method to targeted sequencing data from our patient cohort and validated these results against TMB estimates obtained using an orthogonal assay (Spearman correlation = 0.49, 95% CI [0.24, 0.68]). RESULTS We observed that the RNA measure of TMB was significantly higher in responders to immune blockade treatment (P = 0.028) and that it was predictive of response (AUC = 0.640 with 95% CI [0.493, 0.786]). By contrast, the expression of immune-cell-restricted genes was uncorrelated with patient outcome. CONCLUSION TMB calculated from targeted RNA sequencing has a similar diagnostic ability to TMB generated from targeted DNA sequencing.
Collapse
Affiliation(s)
- John F Markham
- Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC, 3000, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew P Fellowes
- Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC, 3000, Australia.
- Department of Pathology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia.
| | - Thomas Green
- Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC, 3000, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Jose Luis Leal
- Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Roxane Legaie
- Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC, 3000, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
| | - Darren Cullerne
- Murdoch Children's Research Institute, Flemington Road, Parkville, VIC, 3052, Australia
| | - Tessa Morris
- Southern Blood and Cancer Service, Te Whatu Ora Southern, Dunedin, New Zealand
- Mercy Cancer Care, Mercy Hospital, Dunedin, New Zealand
| | - Tom John
- Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Ben Solomon
- Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC, 3000, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| | - Stephen B Fox
- Peter MacCallum Cancer Centre, 305 Grattan Street, Parkville, VIC, 3000, Australia
- Department of Pathology, Peter MacCallum Cancer Centre, Parkville, VIC, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|