1
|
Satgunaseelan L, Lee M, Iannuzzi S, Hallal S, Deang K, Stanceski K, Wei H, Mason S, Shivalingam B, Sim HW, Buckland ME, Alexander KL. 'The Reports of My Death Are Greatly Exaggerated'-Evaluating the Effect of Necrosis on MGMT Promoter Methylation Testing in High-Grade Glioma. Cancers (Basel) 2024; 16:1906. [PMID: 38791984 PMCID: PMC11120496 DOI: 10.3390/cancers16101906] [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: 04/03/2024] [Revised: 05/09/2024] [Accepted: 05/14/2024] [Indexed: 05/26/2024] Open
Abstract
(1) Background: MGMT (O-6-methylguanine-DNA methyltransferase) promoter methylation remains an important predictive biomarker in high-grade gliomas (HGGs). The influence of necrosis on the fidelity of MGMT promoter (MGMTp) hypermethylation testing is currently unknown. Therefore, our study aims to evaluate the effect of varying degrees of necrosis on MGMTp status, as determined by pyrosequencing, in a series of primary and recurrent HGGs; (2) Methods: Within each case, the most viable blocks (assigned as 'true' MGMTp status) and the most necrotic block were determined by histopathology review. MGMTp status was determined by pyrosequencing. Comparisons of MGMTp status were made between the most viable and most necrotic blocks. (3) Results: 163 samples from 64 patients with HGGs were analyzed. MGMTp status was maintained in 84.6% of primary and 78.3% of recurrent HGGs between the most viable and necrotic blocks. A threshold of ≥60% tumor cellularity was established at which MGMTp status was unaltered, irrespective of the degree of necrosis. (4) Conclusions: MGMTp methylation status, as determined by pyrosequencing, does not appear to be influenced by necrosis in the majority of cases at a cellularity of at least 60%. Further investigation into the role of intratumoral heterogeneity on MGMTp status will increase our understanding of this predictive marker.
Collapse
Affiliation(s)
- Laveniya Satgunaseelan
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Maggie Lee
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Sebastian Iannuzzi
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Susannah Hallal
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
| | - Kristine Deang
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
| | - Kristian Stanceski
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Heng Wei
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Sofia Mason
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (S.M.); (H.-W.S.)
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Brindha Shivalingam
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
- Department of Neurosurgery, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Hao-Wen Sim
- Department of Medical Oncology, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia; (S.M.); (H.-W.S.)
- Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
- Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW 2050, Australia
- Department of Medical Oncology, The Kinghorn Cancer Centre, Darlinghurst, NSW 2010, Australia
| | - Michael E. Buckland
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
| | - Kimberley L. Alexander
- Department of Neuropathology, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia; (M.L.); (S.I.); (S.H.); (K.S.); (H.W.); (M.E.B.); (K.L.A.)
- Faculty of Medicine and Health, School of Medicine, University of Sydney, Camperdown Campus, Sydney, NSW 2000, Australia; (K.D.); (B.S.)
- Department of Neurosurgery, Chris O’Brien Lifehouse, Camperdown, NSW 2050, Australia
| |
Collapse
|
2
|
Xiang Y, Li S, Song M, Wang H, Hu K, Wang F, Wang Z, Niu Z, Liu J, Cai Y, Li Y, Zhu X, Geng J, Zhang Y, Teng H, Wang W. KRAS status predicted by pretreatment MRI radiomics was associated with lung metastasis in locally advanced rectal cancer patients. BMC Med Imaging 2023; 23:210. [PMID: 38087207 PMCID: PMC10717608 DOI: 10.1186/s12880-023-01173-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/05/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Mutated KRAS may indicate an invasive nature and predict prognosis in locally advanced rectal cancer (LARC). We aimed to establish a radiomic model using pretreatment T2W MRIs to predict KRAS status and explore the association between the KRAS status or model predictions and lung metastasis. METHODS In this retrospective multicentre study, LARC patients from two institutions between January 2012 and January 2019 were randomly divided into training and testing cohorts. Least absolute shrinkage and selection operator (LASSO) regression and the support vector machine (SVM) classifier were utilized to select significant radiomic features and establish a prediction model, which was validated by radiomic score distribution and decision curve analysis. The association between the model stratification and lung metastasis was investigated by Cox regression and Kaplan‒Meier survival analysis; the results were compared by the log-rank test. RESULTS Overall, 103 patients were enrolled (73 and 30 in the training and testing cohorts, respectively). The median follow-up was 38.1 months (interquartile range: 26.9, 49.4). The radiomic model had an area under the curve (AUC) of 0.983 in the training cohort and 0.814 in the testing cohort. Using a cut-off of 0.679 defined by the receiver operating characteristic (ROC) curve, patients with a high radiomic score (RS) had a higher risk for lung metastasis (HR 3.565, 95% CI 1.337, 9.505, p = 0.011), showing similar predictive performances for the mutant and wild-type KRAS groups (HR 3.225, 95% CI 1.249, 8.323, p = 0.016, IDI: 1.08%, p = 0.687; NRI 2.23%, p = 0.766). CONCLUSIONS We established and validated a radiomic model for predicting KRAS status in LARC. Patients with high RS experienced more lung metastases. The model could noninvasively detect KRAS status and may help individualize clinical decision-making.
Collapse
Affiliation(s)
- Yirong Xiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Shuai Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Maxiaowei Song
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Hongzhi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Ke Hu
- Department of Radiation Oncology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Fengwei Wang
- Department of Oncology, Tianjin Union Medical Center, Tianjin, China
| | - Zhi Wang
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China
| | - Zhiyong Niu
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China
| | - Jin Liu
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China
| | - Yong Cai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yongheng Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Xianggao Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Jianhao Geng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yangzi Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Huajing Teng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Weihu Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
3
|
Dandapath I, Chakraborty R, Kaur K, Mahajan S, Singh J, Sharma MC, Sarkar C, Suri V. Molecular alterations of low-grade gliomas in young patients: Strategies and platforms for routine evaluation. Neurooncol Pract 2021; 8:652-661. [PMID: 34777834 PMCID: PMC8579091 DOI: 10.1093/nop/npab053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
In recent years, it has been established that molecular biology of pediatric low-grade gliomas (PLGGs) is entirely distinct from adults. The majority of the circumscribed pediatric gliomas are driven by mitogen-activated protein kinase (MAPK) pathway, which has yielded important diagnostic, prognostic, and therapeutic biomarkers. Further, the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy (cIMPACT) Steering Committee in their fourth meeting, suggested including a panel of molecular markers for integrated diagnosis in "pediatric-type" diffuse gliomas. However, a designated set of platforms for the evaluation of these alterations has yet not been mentioned for easier implementation in routine molecular diagnostics. Herein, we have reviewed the relevance of analyzing these markers and discussed the strategies and platforms best apposite for clinical laboratories.
Collapse
Affiliation(s)
- Iman Dandapath
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Kavneet Kaur
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Swati Mahajan
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Jyotsna Singh
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Mehar C Sharma
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Chitra Sarkar
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| | - Vaishali Suri
- Department of Pathology, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
4
|
Al Zoughbi W, Kim D, Alperstein SA, Ohara K, Manohar J, Greco N, Khani F, Robinson BD, Rao RA, Elemento O, Mosquera JM, Siddiqui MT. Incorporating cytologic adequacy assessment into precision oncology workflow using telepathology: An institutional experience. Cancer Cytopathol 2021; 129:874-883. [PMID: 33929788 DOI: 10.1002/cncy.22441] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/25/2021] [Accepted: 04/05/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Tumor sample quality and quantity determine the success of somatic mutation analysis. Thus, a rapid on-site evaluation (ROSE) tumor cytology adequacy assessment was incorporated into the workflow of precision oncology at Weill Cornell Medicine in New York City. Optimal samples were obtained from 68 patients with metastatic cancer. METHODS Cytopathologists performed ROSE on fine-needle aspirate samples via telepathology, and subsequently core-needle biopsies were obtained. In a retrospective manner, the concordance between adequacy assessment and the success rate of the procedure was evaluated to obtain sufficient tumor tissue for next-generation sequencing (NGS). RESULTS Out of the 68 procedures, 43 were documented as adequate and 25 were documented as inadequate. The diagnostic yield of adequate procedures was 100%. Adequacy evaluation predicted the success rate of molecular profiling in 40 of 43 procedures (93%; 95% CI, 80.9-98.5 procedures). The success rate of molecular testing was significantly higher in the adequate group: 93% compared with 32% in the inadequate group (P < .0005). Seven procedures that failed to provide quality material for mutational analysis and pathological diagnosis were evaluated as inadequate. Cell block provided sufficient DNA for NGS in 6 cases. In 2 cases, a core biopsy could not be performed; hence, the fine-needle aspirate material confirmed the diagnosis and was used for NGS testing. CONCLUSION These results support the incorporation of ROSE into the workflow of precision oncology to obtain high-quality tissue samples from metastatic lesions. In addition, NGS testing of concurrent cytology specimens with adequate cellularity can be a surrogate for NGS testing of biopsy specimens.
Collapse
Affiliation(s)
- Wael Al Zoughbi
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
| | - David Kim
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Susan Ann Alperstein
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- Papanicolaou Cytology Laboratory, Weill Cornell Medicine, New York, New York
| | - Kentaro Ohara
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
| | - Jyothi Manohar
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
| | - Noah Greco
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
| | - Francesca Khani
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
| | - Brian D Robinson
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
| | - Rema A Rao
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
| | - Olivier Elemento
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York, New York
| | - Juan Miguel Mosquera
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
| | - Momin T Siddiqui
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, New York
- The Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine and New York-Presbyterian Hospital, New York, New York
- Papanicolaou Cytology Laboratory, Weill Cornell Medicine, New York, New York
| |
Collapse
|
5
|
Koboldt DC. Best practices for variant calling in clinical sequencing. Genome Med 2020; 12:91. [PMID: 33106175 PMCID: PMC7586657 DOI: 10.1186/s13073-020-00791-w] [Citation(s) in RCA: 184] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 10/08/2020] [Indexed: 02/08/2023] Open
Abstract
Next-generation sequencing technologies have enabled a dramatic expansion of clinical genetic testing both for inherited conditions and diseases such as cancer. Accurate variant calling in NGS data is a critical step upon which virtually all downstream analysis and interpretation processes rely. Just as NGS technologies have evolved considerably over the past 10 years, so too have the software tools and approaches for detecting sequence variants in clinical samples. In this review, I discuss the current best practices for variant calling in clinical sequencing studies, with a particular emphasis on trio sequencing for inherited disorders and somatic mutation detection in cancer patients. I describe the relative strengths and weaknesses of panel, exome, and whole-genome sequencing for variant detection. Recommended tools and strategies for calling variants of different classes are also provided, along with guidance on variant review, validation, and benchmarking to ensure optimal performance. Although NGS technologies are continually evolving, and new capabilities (such as long-read single-molecule sequencing) are emerging, the “best practice” principles in this review should be relevant to clinical variant calling in the long term.
Collapse
Affiliation(s)
- Daniel C Koboldt
- Steve and Cindy Rasmussen Institute for Genomic Medicine at Nationwide Children's Hospital, Columbus, OH, USA. .,Department of Pediatrics, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
6
|
Jang YO, Noh GS, Liu H, Koo B, Qiao Z, Shin Y. Dimethyl 3,3'-dithiobispropionimidate-functionalized diatomaceous earth particles for efficient biomolecule separation. Sci Rep 2020; 10:15592. [PMID: 32973156 PMCID: PMC7519118 DOI: 10.1038/s41598-020-72913-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/01/2020] [Indexed: 11/09/2022] Open
Abstract
The early diagnosis and monitoring of cancers are key factors in effective cancer treatment. Particularly, the separation of biomolecules is an essential step for both diagnostic and analytical purposes. However, the current techniques used to isolate biomolecules are intensive, laborious, and require multiple instruments as well as repeated sample preparations to separate each biomolecule. Thus, an efficient separation system that can simultaneously separate biomolecules from scarce samples is highly desirable. Hence, in this study, we developed a biosilica-based syringe filtration system for the efficient separation of biomolecules from cancer samples using amine-modified diatomaceous earth (AD) with dimethyl 3,3′-dithiobispropionimidate (DTBP). The syringe filter can be an efficient and rapid tool for use in various procedures without complex instruments. The DTBP-based AD system was combined with the syringe filter system for nucleic acid and protein separation from various cancer cells. We demonstrated the efficacy of the DTBP-based AD in a single-filter system for the efficient separation of DNA and proteins within 40 min. This DTBP-based AD syringe filter system showed good rapidity, efficiency, and affordability in the separation of biomolecules from single samples for the early diagnosis and clinical analysis of cancers.
Collapse
Affiliation(s)
- Yoon Ok Jang
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, Biomedical Engineering Research Center, Asan Institute of Life Sciences, Asan Medical Center, 05505, Seoul, Republic of Korea
| | - Geun Su Noh
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, Biomedical Engineering Research Center, Asan Institute of Life Sciences, Asan Medical Center, 05505, Seoul, Republic of Korea
| | - Huifang Liu
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, Biomedical Engineering Research Center, Asan Institute of Life Sciences, Asan Medical Center, 05505, Seoul, Republic of Korea
| | - Bonhan Koo
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, Biomedical Engineering Research Center, Asan Institute of Life Sciences, Asan Medical Center, 05505, Seoul, Republic of Korea
| | - Zhen Qiao
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, Biomedical Engineering Research Center, Asan Institute of Life Sciences, Asan Medical Center, 05505, Seoul, Republic of Korea
| | - Yong Shin
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology (AMIST), University of Ulsan College of Medicine, Biomedical Engineering Research Center, Asan Institute of Life Sciences, Asan Medical Center, 05505, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Patel NM, Jo H, Eberhard DA, Yin X, Hayward MC, Stein MK, Hayes DN, Grilley-Olson JE. Improved Tumor Purity Metrics in Next-generation Sequencing for Clinical Practice: The Integrated Interpretation of Neoplastic Cellularity and Sequencing Results (IINCaSe) Approach. Appl Immunohistochem Mol Morphol 2020; 27:764-772. [PMID: 30102605 PMCID: PMC6887630 DOI: 10.1097/pai.0000000000000684] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Accepted: 05/08/2018] [Indexed: 12/18/2022]
Abstract
Neoplastic cellularity contributes to the analytic sensitivity of most present technologies for mutation detection, such that they underperform when stroma and inflammatory cells dilute a cancer specimen's variant fraction. Thus, tumor purity assessment by light microscopy is used to determine sample adequacy before sequencing and to interpret the significance of negative results and mutant allele fraction afterwards. However, pathologist estimates of tumor purity are imprecise and have limited reproducibility. With the advent of massively parallel sequencing, large amounts of molecular data can be analyzed by computational purity algorithms. We retrospectively compared tumor purity of 3 computational algorithms with neoplastic cellularity using hematoxylin and eosin light microscopy to determine which was best for clinical evaluation of molecular profiling. Data were analyzed from 881 cancer patients from a clinical trial cohort, LCCC1108 (UNCseq), whose tumors had targeted massively parallel sequencing. Concordance among algorithms was poor, and the specimens analyzed had high rates of algorithm failure partially due to variable tumor purity. Computational tumor purity estimates did not add value beyond the pathologist's estimate of neoplastic cellularity microscopy. To improve present methods, we propose a semiquantitative, clinically applicable strategy based on mutant allele fraction and copy number changes present within a given specimen, which when combined with the morphologic tumor purity estimate, guide the interpretation of next-generation sequencing results in cancer patients.
Collapse
Affiliation(s)
- Nirali M. Patel
- Department of Pathology and Laboratory Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Heejoon Jo
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - David A. Eberhard
- Department of Pathology and Laboratory Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Xiaoying Yin
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Michele C. Hayward
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| | - Matthew K. Stein
- Department of Internal Medicine, Division of Hematology and Oncology
- West Cancer Center, University of Tennessee Health Science Center, Memphis, TN
| | - David Neil Hayes
- Department of Internal Medicine, Division of Hematology and Oncology
- West Cancer Center, University of Tennessee Health Science Center, Memphis, TN
| | - Juneko E. Grilley-Olson
- Department of Internal Medicine, Division of Hematology and Oncology, University of North Carolina School of Medicine
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC
| |
Collapse
|
8
|
Voyten J, Holtzman MP, Pantanowitz L, Dhir R, Beasley HS, Cuda J, Monaco SE. Lessons learned from clinical trial queries on small biopsy collections: importance of rapid on-site evaluation. J Am Soc Cytopathol 2020; 9:461-468. [PMID: 32499137 DOI: 10.1016/j.jasc.2020.04.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/15/2020] [Accepted: 04/15/2020] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Small biopsies and cytology specimens have become increasingly important for clinical trials and biomarker testing. Thus, institutions must ensure that adequate lesional material meeting the specifications for a multitude of different protocols is available. This can be achieved using rapid on-site evaluation (ROSE). The aim of the present study was to determine the recent clinical trial biopsy characteristics and study the feedback on these collections at our institution. MATERIALS AND METHODS Clinical trial biopsies performed at our institution and trial feedback (including "queries") were analyzed from the 2017 to 2019. The query data were reviewed in detail, in addition to any protocol modifications related to biopsy requirements and study protocol changes. RESULTS A total of 698 biopsy collections were performed for clinical trial purposes for 95 trials, with most requiring biopsies at >1 time point (63.2%), for phase I or II trials (92.6%), and for specific tumor types (67.4%). Only 18 of the trials (18.9%) requiring fresh tissue biopsies provided feedback. The feedback included data from 90 cases (12.9%), of which 27 (30.0%) had queries regarding insufficient (n = 10; 37.0%) or borderline (n = 17; 63.0%) tumor tissue. Only 1 (3.7%) of these had had ROSE by cytology. ROSE was performed in accordance with institutional guidelines (45.3%), as required by the study (1.1%), or because of trial modification (5.3%). CONCLUSIONS The present investigation has shown the high volume of clinical trial biopsies managed at our academic cancer center. Feedback from the trials was low at 18.9% and frequently involved suboptimal cases without ROSE used at acquisition. This has led to more widespread adoption of ROSE to mitigate insufficient biopsy specimens and repeat procedures. The high volume of clinical trial biopsies and variability in trial needs necessitates a collaborative multidisciplinary network, including cytology services, to facilitate these important biopsies for patients with cancer.
Collapse
Affiliation(s)
- Jamie Voyten
- Clinical Research Services, UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Matthew P Holtzman
- Division of Surgical Oncology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Liron Pantanowitz
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Rajiv Dhir
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - H Scott Beasley
- Department of Radiology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Jackie Cuda
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania
| | - Sara E Monaco
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania.
| |
Collapse
|
9
|
Tseng LH, De Marchi F, Pallavajjalla A, Rodriguez E, Xian R, Belchis D, Gocke CD, Eshleman JR, Illei P, Lin MT. Clinical Validation of Discordant Trunk Driver Mutations in Paired Primary and Metastatic Lung Cancer Specimens. Am J Clin Pathol 2019; 152:570-581. [PMID: 31264684 DOI: 10.1093/ajcp/aqz077] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES To propose an operating procedure for validation of discordant trunk driver mutations. METHODS Concordance of trunk drivers was examined by next-generation sequencing in 15 patients with two to three metastatic lung cancers and 32 paired primary and metastatic lung cancers. RESULTS Tissue identity was confirmed by genotyping 17 single-nucleotide polymorphisms within the panel. All except three pairs showed concordant trunk drivers. Quality assessment conducted in three primary and metastatic pairs with discordant trunk drivers indicates metastasis from a synchronous or remote lung primary in two patients. Review of literature revealed high discordant rates of EGFR and KRAS mutations, especially when Sanger sequencing was applied to examine primary and lymph node metastatic tumors. CONCLUSIONS Trunk driver mutations are highly concordant in primary and metastatic tumors. Discordance of trunk drivers, once confirmed, may suggest a second primary cancer. Guidelines are recommended to establish standard operating procedures for validation of discordant trunk drivers.
Collapse
Affiliation(s)
- Li-Hui Tseng
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Medical Genetics, National Taiwan University Hospital, Taipei
| | - Federico De Marchi
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Aparna Pallavajjalla
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Erika Rodriguez
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Rena Xian
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Deborah Belchis
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Christopher D Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Peter Illei
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
10
|
McHugh KE, Dermawan JK, Cheng YW, Cruise M, Sohal DPS, Reynolds JP. Molecular testing in metastatic colorectal adenocarcinoma cytology cell pellets. Diagn Cytopathol 2019; 47:1132-1137. [PMID: 31290252 DOI: 10.1002/dc.24275] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/05/2019] [Accepted: 06/25/2019] [Indexed: 01/22/2023]
Abstract
BACKGROUND Mutational status for KRAS, NRAS, and BRAF genes should be performed on all colorectal carcinoma (CRC) specimens in order to guide targeted therapy selection for metastatic disease. Mutations are typically assessed via polymerase chain reaction and/or next generation sequencing (NGS) on formalin-fixed paraffin-embedded tissues. With minimally invasive diagnostic methodologies, the cytology cell pellet obtained by fine-needle aspiration (FNA) can serve as an alternative source of tumor deoxyribonucleic acid. METHODS An electronic record review of the cytopathology files (CoPathPlus, Cerner Corp., North Kansas City, Missouri) from September 1, 2015 through December 31, 2018 was conducted. All cytology specimens obtained via FNA and diagnosed as metastatic CRC on which NGS was performed were included. NGS for KRAS, NRAS, and BRAF mutations using the AmpliSeq Cancer Hotspot Panel v2.0 kit (Thermo Fisher Scientific, Waltham, Massachusetts) was performed on cytology cell pellets. RESULTS Forty-eight cases were identified. Forty-six of 48 specimens (96%) were adequate for molecular testing. Of those adequate specimens, proportion of malignant cells in the sample ranged from 5% to 95% (mean 46%). Twenty-seven of 48 cases (56%) were positive for clinically relevant mutations. Twenty-four of 27 cases (89%) were positive for KRAS mutations, with exon 2 most frequently involved (22/24 cases, 92%). Two of 27 cases (7%) were positive for NRAS mutations and one case (1/27, 4%) was positive for a BRAF mutation involving codon 594. CONCLUSION Mutational analysis performed on cytology cell pellets serves as a useful means of gathering clinically actionable information on tumor mutation status in metastatic CRC.
Collapse
Affiliation(s)
- Kelsey E McHugh
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Josephine K Dermawan
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Yu-Wei Cheng
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Michael Cruise
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| | - Davendra P S Sohal
- Department of Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jordan P Reynolds
- Robert J. Tomsich Pathology and Laboratory Medicine Institute, Cleveland Clinic, Cleveland, Ohio
| |
Collapse
|
11
|
Jang YO, Jin CE, Choi EH, Shin JH, Kweon J, Koo B, Lim SB, Lee SW, Shin Y. A homobifunctional imidoester-based microfluidic system for simultaneous DNA and protein isolation from solid or liquid biopsy samples. LAB ON A CHIP 2019; 19:2256-2264. [PMID: 31173022 DOI: 10.1039/c9lc00367c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The isolation of bio-molecules such as proteins and nucleic acids is a necessary step for both diagnostic and analytical processes in the broad fields of research and clinical applications. Although a myriad of isolation technologies have been developed, a method for simultaneous protein and nucleic acid isolation has not been explored for clinical use. Obtaining samples from certain cancers or rare diseases can be difficult. In addition, the heterogeneity of cancer tissues typically leads to inconsistent results when analyzing biomolecules. We here describe a homobifunctional imidoester (HI)-based microfluidic system for simultaneous DNA and protein isolation from either a solid or liquid single biopsy sample. An efficient and cost effective microfluidic design with less air bubbles was identified among several candidates using simulation and experimental results from the streamlining of isolation processing. HI groups were used as capture reagents for the simultaneous isolation of bio-molecules from a single specimen in a single microfluidic system. The clinical utility of this system for the simultaneous isolation of DNA and proteins within 40 min was validated in cancer cell lines and 23 tissue biopsies from colorectal cancer patients. The quantity of isolated protein and DNA was high using this system compared to the spin-column method. This HI-based microfluidic system shows good rapidity, affordability, and portability in the isolation of bio-molecules from limited samples for subsequent clinical analysis.
Collapse
Affiliation(s)
- Yoon Ok Jang
- Department of Convergence Medicine, Asan Medical Institute of Convergence Science and Technology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
De Marchi F, Haley L, Fryer H, Ibrahim J, Beierl K, Zheng G, Gocke CD, Eshleman JR, Belchis D, Illei P, Lin MT. Clinical Validation of Coexisting Activating Mutations Within EGFR, Mitogen-Activated Protein Kinase, and Phosphatidylinositol 3-Kinase Pathways in Lung Cancers. Arch Pathol Lab Med 2018; 143:174-182. [PMID: 30485130 DOI: 10.5858/arpa.2017-0495-oa] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Mutations within the same signature transduction pathway are redundant and, therefore, most are mutually exclusive. Laboratory errors, however, may introduce unexpected coexisting mutations. OBJECTIVE.— To validate coexisting mutations within epidermal growth factor receptor (EGFR), mitogen-activated protein kinase, and phosphatidylinositol 3-kinase pathways. DESIGN.— In this retrospective study for quality assessment of next-generation sequencing in a clinical diagnostics setting, coexisting mutations within EGFR, KRAS, NRAS, BRAF, AKT1, and PIK3CA genes were examined in 1208 non-small cell lung cancers. RESULTS.— EGFR mutations did not coexist with BRAF mutations, neither kinase-activated nor kinase-impaired mutations. There was a low but similar incidence (3.3%-5.1%) of PIK3CA mutations in BRAF-, EGFR-, and KRAS-mutated lung cancers and a rare incidence of coexisting KRAS and EGFR mutations detected in 1 of 1208 lung cancers (0.08%) or 1 of 226 EGFR-mutated lung cancers (0.4%). Coexisting BRAF p.V600E mutation was observed in 3 of 4 AKT1 p.E17K-mutated lung cancers. Mutational profiling of DNA reisolated from subareas with the same or different histomorphology, using an alternative assay, confirmed that coexisting mutations might present within the same (whole or subclonal) population or different populations and clarified that the so-called coexisting activating KRAS and BRAF mutations originally reported in a specimen were indeed present in separate lung nodules submitted in the same block. CONCLUSIONS.— The results supported that EGFR and BRAF mutations are early driver mutations in lung cancers. Guidelines from official organizations to establish standard operating procedures are warranted to validate unexpected coexisting mutations and, if clinically indicated, to determine their presence in the same or different tumor populations.
Collapse
Affiliation(s)
- Federico De Marchi
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Lisa Haley
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Henderson Fryer
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Junaid Ibrahim
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Katie Beierl
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Gang Zheng
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Christopher D Gocke
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - James R Eshleman
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Deborah Belchis
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Peter Illei
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| | - Ming-Tseh Lin
- From the Departments of Pathology (Drs De Marchi, Ibrahim, Zheng, Gocke, Eshleman, Belchis, Illei, and Lin, Ms Haley, Mr Fryer, and Ms Beierl) and Oncology (Drs Gocke and Eshleman), Johns Hopkins University School of Medicine, Baltimore, Maryland; and the Division of Hematology and Bone Marrow Transplantation, Hospital-University of Udine, Udine, Italy (Dr De Marchi)
| |
Collapse
|
13
|
Thierry AR, El Messaoudi S, Mollevi C, Raoul JL, Guimbaud R, Pezet D, Artru P, Assenat E, Borg C, Mathonnet M, De La Fouchardière C, Bouché O, Gavoille C, Fiess C, Auzemery B, Meddeb R, Lopez-Crapez E, Sanchez C, Pastor B, Ychou M. Clinical utility of circulating DNA analysis for rapid detection of actionable mutations to select metastatic colorectal patients for anti-EGFR treatment. Ann Oncol 2018; 28:2149-2159. [PMID: 28911069 DOI: 10.1093/annonc/mdx330] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Background While tumor-tissue remains the 'gold standard' for genetic analysis in cancer patients, it is challenged with the advent of circulating cell-free tumor DNA (ctDNA) analysis from blood samples. Here, we broaden our previous study on the clinical validation of plasma DNA in metastatic colorectal cancer patients, by evaluating its clinical utility under standard management care. Patients and methods Concordance and data turnaround-time of ctDNA when compared with tumor-tissue analysis were studied in a real-time blinded prospective multicenter clinical study (n = 140 metastatic colorectal patients). Results are presented according to STARD criteria and were discussed in regard with clinical outcomes of patients. Results Much more mutations were found by ctDNA analysis: 59%, 11.8% and 14.4% of the patients were found KRAS, NRAS and BRAF mutant by ctDNA analysis instead of 44%, 8.8% and 7.2% by tumor-tissue analysis. Median tumor-tissue data turnaround-time was 16 days while 2 days for ctDNA analysis. Discordant samples analysis revealed that use of biopsy, long delay between tumor-tissue and blood collection and resection of the tumor at time of blood draw, tumor site, or type of tissue analyzed seem to affect concordance. Altogether, the clinical data with respect to the anti-epidermal growth factor receptor response (RAS status) and the prognosis (BRAF status) of those discordant patients do not appear contradictory to the mutational status as determined by plasma analysis. Lastly, we present the first distribution profile of the RAS and BRAF hotspot mutations as determined by ctDNA analysis (n = 119), revealing a high proportion of patients with multiple mutations (45% of the population and up to 5 mutations) and only 24% of WT scored patients for both genes. Mutation profile as determined from ctDNA analysis with using various detection thresholds highlights the importance of the test sensitivity. Conclusion Our study showed that ctDNA could replace tumor-tissue analysis, and also clinical utility of ctDNA analysis by considerably reducing data turnaround time.
Collapse
Affiliation(s)
- A R Thierry
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier
| | - S El Messaoudi
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier
| | - C Mollevi
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier.,Biometry Unit, Regional Institute of Cancer of Montpellier, Montpellier
| | - J L Raoul
- Department of Medical Oncology, Paoli Calmettes Institute, Marseille
| | - R Guimbaud
- Department of Oncology, University Hospital Center of Toulouse - Hospital Rangueil-Purpan, Toulouse
| | - D Pezet
- Digestive Oncology Unit, Department of Digestive Surgery, University Hospital Center of Clermont-Ferrand, UMR Inserm/Auvergne University U1071, Clermont-Ferrand
| | - P Artru
- Jean-Mermoz Private Hospital, Lyon
| | - E Assenat
- CHRU Montpellier, St. Eloi Hospital, Montpellier
| | - C Borg
- CHRU Jean MINJOZ, Besançon
| | - M Mathonnet
- Digestive Surgery Department, Clinical Investigation Centre, University Hospital Center of Limoges, INSERM 0801, Limoges
| | | | | | - C Gavoille
- Alexis Vautrin Oncology Institute of Lorraine, Nancy
| | - C Fiess
- Digestive Oncology Department, Regional Institute of Cancer of Montpellier, Montpellier, France
| | - B Auzemery
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier
| | - R Meddeb
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier
| | - E Lopez-Crapez
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier
| | - C Sanchez
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier
| | - B Pastor
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier
| | - M Ychou
- IRCM, Institute of Research in Oncology of Montpellier, Montpellier.,INSERM, U1194, Montpellier.,Department of Oncology, Montpellier University, Montpellier.,Regional Institute of Cancer of Montpellier, Montpellier.,Digestive Oncology Department, Regional Institute of Cancer of Montpellier, Montpellier, France
| |
Collapse
|
14
|
Heterogeneity of resistance mutations detectable by nextgeneration sequencing in TKI-treated lung adenocarcinoma. Oncotarget 2018; 7:45237-45248. [PMID: 27304188 PMCID: PMC5216719 DOI: 10.18632/oncotarget.9931] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 05/19/2016] [Indexed: 12/12/2022] Open
Abstract
EGFR-mutated lung adenocarcinomas routinely develop resistance to tyrosine kinase inhibitors (TKI). To better characterize the relative frequencies of the resistance mechanisms, we analyzed 48 EGFR-mutated TKI-resistant specimens from 41 patients. Next-generation sequencing of post-treatment specimens detected EGFR p.T790M in 31 (79%) of 39 patients, PIK3CA mutations in 10 (26%), EGFR p.S768_V769delinsIL in one, and KRAS p.G12C in one. Five PIK3CA mutations were outside of codons 542, 545, and 1047. Three of four pre-treatment specimens did not carry the PIK3CA mutation found in the post-treatment sample. Small cell carcinoma transformation was identified in four patients; none had p.T790M, including two where p.T790M was identified in the co-existing adenocarcinoma. In p.T790M-mutated specimens, the allele frequency was less than 5% in 24% of cases. p.T790M allele frequency was usually lower than that of the sensitizing mutation indicating that the resistance mutation was present either in a subset of cells or, if the sensitizing mutation was amplified, in a subset of the sensitizing alleles of a dominant clone. Eight patients had multiple resistance mutations, suggesting either multiple separate resistant clones or a single clone harboring multiple resistance mechanisms. PIK3CA mutations appear to be a more significant resistance mechanism than previously recognized.
Collapse
|
15
|
Moorcraft SY, Gonzalez de Castro D, Cunningham D, Jones T, Walker BA, Peckitt C, Yuan LC, Frampton M, Begum R, Eltahir Z, Wotherspoon A, Teixeira Mendes LS, Hulkki Wilson S, Gillbanks A, Baratelli C, Fotiadis N, Patel A, Braconi C, Valeri N, Gerlinger M, Rao S, Watkins D, Chau I, Starling N. Investigating the feasibility of tumour molecular profiling in gastrointestinal malignancies in routine clinical practice. Ann Oncol 2018; 29:230-236. [PMID: 29361134 DOI: 10.1093/annonc/mdx631] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Background Targeted capture sequencing can potentially facilitate precision medicine, but the feasibility of this approach in gastrointestinal (GI) malignancies is unknown. Patients and methods The FOrMAT (Feasibility of a Molecular Characterisation Approach to Treatment) study was a feasibility study enrolling patients with advanced GI malignancies from February 2014 to November 2015. Targeted capture sequencing (mainly using archival formalin-fixed paraffin-embedded diagnostic/resection samples) was carried out to detect mutations, copy number variations and translocations in up to 46 genes which had prognostic/predictive significance or were targets in current/upcoming clinical trials. Results Of the 222 patients recruited, 215 patients (96.8%) had available tissue samples, 125 patients (56.3%) had ≥16 genes successfully sequenced and 136 patients (61.2%) had ≥1 genes successfully sequenced. Sample characteristics influenced the proportion of successfully sequenced samples, e.g. tumour type (colorectal 70.9%, biliary 52.6%, oesophagogastric 50.7%, pancreas 27.3%, P = 0.002), tumour cellularity (high versus low: 78.3% versus 13.3%, P ≤ 0.001), tumour content (high versus low: 78.6% versus 27.3%, P = 0.001) and type of sample (resection versus biopsy: 82.4% versus 47.6%, P ≤ 0.001). Currently, actionable alterations were detected in 90 (40.5%) of the 222 patients recruited (66% of the 136 patients sequenced) and 2 patients subsequently received a targeted therapy. The most frequently detected currently actionable alterations were mutations in KRAS, BRAF, TP53 and PIK3CA. For the 205 patients with archival samples, the median time to obtain sequencing results was 18.9 weeks, including a median of 4.9 weeks for sample retrieval and 5.1 weeks for sequencing. Conclusions Targeted sequencing detected actionable alterations in formalin-fixed paraffin-embedded samples, but tissue characteristics are of critical importance in determining sequencing success. Routine molecular profiling of GI tumours outside of clinical trials is not an effective use of healthcare resources unless more targeted drugs become available. ClinicalTrials.gov identifier NCT02112357.
Collapse
Affiliation(s)
- S Y Moorcraft
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - D Gonzalez de Castro
- Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - D Cunningham
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - T Jones
- Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - B A Walker
- Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - C Peckitt
- Department of Statistics, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - L C Yuan
- Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - M Frampton
- Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - R Begum
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - Z Eltahir
- Department of Pathology, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - A Wotherspoon
- Department of Pathology, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - L S Teixeira Mendes
- Department of Pathology, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - S Hulkki Wilson
- Centre for Molecular Pathology, The Royal Marsden NHS Foundation Trust, Sutton, UK
| | - A Gillbanks
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - C Baratelli
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - N Fotiadis
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - A Patel
- Department of Radiology, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - C Braconi
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
- The Institute of Cancer Research, London and Sutton, UK
| | - N Valeri
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
- The Institute of Cancer Research, London and Sutton, UK
| | - M Gerlinger
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
- The Institute of Cancer Research, London and Sutton, UK
| | - S Rao
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - D Watkins
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - I Chau
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| | - N Starling
- Gastrointestinal and Lymphoma Unit, The Royal Marsden NHS Foundation Trust, London and Sutton, UK
| |
Collapse
|
16
|
Li W, Qiu T, Guo L, Ying J. Major challenges related to tumor biological characteristics in accurate mutation detection of colorectal cancer by next-generation sequencing. Cancer Lett 2017; 410:92-99. [PMID: 28942013 DOI: 10.1016/j.canlet.2017.09.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 02/07/2023]
Abstract
Next-generation sequencing (NGS) has been used in mutation detection of colorectal cancer (CRC). We here interrogated 747 CRC samples to detect mutations in 22 cancer-related genes by using NGS, and to explore some key challenges related to tumor biology. RAS mutations (KRAS or NRAS mutations), RAS/BRAF/PIK3CA mutations (mutations in KRAS, NRAS, BRAF or PIK3CA) and mutation burden (mutations in any of the 22 detected genes) were observed in 53.0% (396/747), 57.1% (431/747) and 84.2% (629/747) of specimens, respectively. Higher mutation frequencies were observed in biopsy specimens with ≥20% tumor cellularity than those with <20% tumor cellularity, but these differences were not observed in resection samples. Intratumor mutational heterogeneity was estimated by mutant allele frequency and tumor cellularity, and more likely to occur in PIK3CA mutant tumors. No significant differences of mutation frequencies were detected between primary and metastatic tumors. Additionally, specimens after chemotherapy showed lower mutation frequencies compared with specimens without chemotherapy. Together, our findings demonstrate that poor tumor cellularity, tumor heterogeneity and adjuvant therapy may confound the molecular diagnosis of CRC, and should be highlighted with prospective quality assessment during tissue process.
Collapse
Affiliation(s)
- Weihua Li
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Tian Qiu
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Lei Guo
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Jianming Ying
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China.
| |
Collapse
|
17
|
Illei PB, Belchis D, Tseng LH, Nguyen D, De Marchi F, Haley L, Riel S, Beierl K, Zheng G, Brahmer JR, Askin FB, Gocke CD, Eshleman JR, Forde PM, Lin MT. Clinical mutational profiling of 1006 lung cancers by next generation sequencing. Oncotarget 2017; 8:96684-96696. [PMID: 29228562 PMCID: PMC5722514 DOI: 10.18632/oncotarget.18042] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Accepted: 05/10/2017] [Indexed: 12/15/2022] Open
Abstract
Analysis of lung adenocarcinomas for actionable mutations has become standard of care. Here, we report our experience using next generation sequencing (NGS) to examine AKT1, BRAF, EGFR, ERBB2, KRAS, NRAS, and PIK3CA genes in 1006 non-small cell lung cancers in a clinical diagnostic setting. NGS demonstrated high sensitivity. Among 760 mutations detected, the variant allele frequency (VAF) was 2-5% in 33 (4.3%) mutations and 2-10% in 101 (13%) mutations. A single bioinformatics pipeline using Torrent Variant Caller, however, missed a variety of EGFR mutations. Mutations were detected in KRAS (36% of tumors), EGFR (19%) including 8 (0.8%) within the extracellular domain (4 at codons 108 and 4 at codon 289), BRAF (6.3%), and PIK3CA (3.7%). With a broader reportable range, exon 19 deletion and p.L858R accounted for only 36% and 26% of EGFR mutations and p.V600E accounted for only 24% of BRAF mutations. NGS provided accurate sequencing of complex mutations seen in 19% of EGFR exon 19 deletion mutations. Doublet (compound) EGFR mutations were observed in 29 (16%) of 187 EGFR-mutated tumors, including 69% with two non-p.L858R missense mutations and 24% with p.L858 and non-p.L858R missense mutations. Concordant VAFs suggests doublet EGFR mutations were present in a dominant clone and cooperated in oncogenesis. Mutants with predicted impaired kinase, observed in 25% of BRAF-mutated tumors, were associated with a higher incidence of concomitant activating KRAS mutations. NGS demonstrates high analytic sensitivity, broad reportable range, quantitative VAF measurement, single molecule sequencing to resolve complex deletion mutations, and simultaneous detection of concomitant mutations.
Collapse
Affiliation(s)
- Peter B. Illei
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Deborah Belchis
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Li-Hui Tseng
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Doreen Nguyen
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Federico De Marchi
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Division of Hematology and Bone Marrow Transplantation, University of Udine Hospital, Udine, Italy
| | - Lisa Haley
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Stacy Riel
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Katie Beierl
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Gang Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Julie R. Brahmer
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Frederic B. Askin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Christopher D. Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - James R. Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Patrick M. Forde
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, Maryland, USA
| |
Collapse
|
18
|
Sutton BC, Birse RT, Maggert K, Ray T, Hobbs J, Ezenekwe A, Kazmierczak J, Mosko M, Kish J, Bullock A, Shi Z, Stack MS, Irwin D. Assessment of common somatic mutations of EGFR, KRAS, BRAF, NRAS in pulmonary non-small cell carcinoma using iPLEX® HS, a new highly sensitive assay for the MassARRAY® System. PLoS One 2017; 12:e0183715. [PMID: 28926605 PMCID: PMC5604939 DOI: 10.1371/journal.pone.0183715] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/09/2017] [Indexed: 11/19/2022] Open
Abstract
Increased early detection and personalized therapy for lung cancer have coincided with greater use of minimally invasive sampling techniques such as endobronchial ultrasound-guided biopsy (EBUS), endoscopic ultrasound-guided biopsy (EUS), and navigational biopsy, as well as thin needle core biopsies. As many lung cancer patients have late stage disease and other comorbidities that make open surgical procedures hazardous, the least invasive biopsy technique with the highest potential specimen yield is now the preferred first diagnostic study. However, use of these less invasive procedures generates significant analytical challenges for the laboratory, such as a requirement for robust detection of low level somatic mutations, particularly when the starting sample is very small or demonstrates few intact tumor cells. In this study, we assessed 179 clinical cases of non-small cell lung carcinoma (NSCLC) that had been previously tested for EGFR, KRAS, NRAS, and BRAF mutations using a novel multiplexed analytic approach that reduces wild-type signal and allows for detection of low mutation load approaching 1%, iPLEX® HS panel for the MassARRAY® System (Agena Bioscience, San Diego, CA). This highly sensitive system identified approximately 10% more KRAS, NRAS, EGFR and BRAF mutations than were detected by the original test platform, which had a sensitivity range of 5-10% variant allele frequency (VAF).
Collapse
Affiliation(s)
- Bobbie C. Sutton
- Pathology Department, South Bend Medical Foundation, South Bend, IN, United States of America
| | - Ryan T. Birse
- Agena Bioscience, San Diego, CA, United States of America
| | - Kevin Maggert
- Pathology Department, South Bend Medical Foundation, South Bend, IN, United States of America
| | - Tammy Ray
- Pathology Department, South Bend Medical Foundation, South Bend, IN, United States of America
| | - Jessica Hobbs
- Pathology Department, South Bend Medical Foundation, South Bend, IN, United States of America
| | - Amobi Ezenekwe
- Pathology Department, South Bend Medical Foundation, South Bend, IN, United States of America
| | | | - Michael Mosko
- Agena Bioscience, San Diego, CA, United States of America
| | - Joan Kish
- Pathology Department, South Bend Medical Foundation, South Bend, IN, United States of America
| | - Andrew Bullock
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, United States of America
| | - Zonggao Shi
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, United States of America
| | - M. Sharon Stack
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN, United States of America
| | - Darryl Irwin
- Agena Bioscience, San Diego, CA, United States of America
| |
Collapse
|
19
|
Influence of mucinous and necrotic tissue in colorectal cancer samples on KRAS mutation analysis. Pathol Res Pract 2017; 213:606-611. [DOI: 10.1016/j.prp.2017.04.028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 04/28/2017] [Accepted: 04/28/2017] [Indexed: 12/21/2022]
|
20
|
Sharma A, Zhang G, Aslam S, Yu K, Chee M, Palma JF. Novel Approach for Clinical Validation of the cobas KRAS Mutation Test in Advanced Colorectal Cancer. Mol Diagn Ther 2017; 20:231-40. [PMID: 26984642 PMCID: PMC4879158 DOI: 10.1007/s40291-016-0193-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Aim Our objective was to assess the performance of the cobas test versus comparators for KRAS mutation status and predicting clinical response to anti-epidermal growth factor receptor (EGFR) therapy in patients with metastatic colorectal cancer (mCRC). Methods mCRC samples from 398 patients from Roche study NO16968 (XELOXA) and 82 supplemental samples were tested with the cobas® KRAS mutation test (cobas test), the therascreen® KRAS RGQ PCR kit test (therascreen test), and Sanger sequencing as the reference method for detecting mutations in codons 12/13. Results For 461 eligible samples, the cobas test, therascreen test, and sequencing had invalid results for 5.2, 10.8, and 2.6 % of specimens, respectively. Valid cobas and therascreen test results had similar KRAS mutation-positive rates (37.3 vs. 36.3 %, respectively); sequencing was 28.5 %. Positive and negative percent agreement (PPA/NPA) between the cobas test and sequencing was 96.9 % (95 % confidence interval [CI] 92.2–98.8), and 88.7 % (95 % CI 84.7–91.8), respectively. PPA/NPA between the cobas and therascreen tests was 93.3 % (95 % CI 88.1–96.3) and 96.5 % (95 % CI 93.5–98.1), respectively. Bridging analysis from NCIC-CO.17 and NCT00113763 using the cobas test yielded modeled hazard ratios for overall survival and progression-free survival (PFS) of 0.558 (95 % CI 0.422–0.752) and 0.413 (95 % CI 0.304–0.550), respectively, for cetuximab and 0.989 (95 % CI 0.778–1.299) and 0.471 (95 % CI 0.360–0.626), respectively, for panitumumab, demonstrating significant efficacy in the KRAS-negative population for PFS. Conclusion The cobas test showed similar accuracy to the therascreen test for detecting KRAS mutations and could appropriately identify mCRC patients ineligible for anti-EGFR therapy as demonstrated by bridging analysis results. Electronic supplementary material The online version of this article (doi:10.1007/s40291-016-0193-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Abha Sharma
- Medical Affairs Department, Roche Molecular Systems, 4300 Hacienda Drive, Pleasanton, CA, 94588, USA
| | - Guili Zhang
- Medical Affairs Department, Roche Molecular Systems, 4300 Hacienda Drive, Pleasanton, CA, 94588, USA
| | - Shagufta Aslam
- Medical Affairs Department, Roche Molecular Systems, 4300 Hacienda Drive, Pleasanton, CA, 94588, USA
| | - Karen Yu
- Medical Affairs Department, Roche Molecular Systems, 4300 Hacienda Drive, Pleasanton, CA, 94588, USA
| | - Melody Chee
- Medical Affairs Department, Roche Molecular Systems, 4300 Hacienda Drive, Pleasanton, CA, 94588, USA
| | - John F Palma
- Medical Affairs Department, Roche Molecular Systems, 4300 Hacienda Drive, Pleasanton, CA, 94588, USA.
| |
Collapse
|
21
|
Prognostic factors of successful on-purpose tumor biopsies in metastatic cancer patients included in the SHIVA prospective clinical trial. Oncotarget 2017; 8:1760-1773. [PMID: 27655703 PMCID: PMC5352095 DOI: 10.18632/oncotarget.12051] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Accepted: 09/07/2016] [Indexed: 11/25/2022] Open
Abstract
PURPOSE To identify patient/tumor characteristics associated with success of biopsy in patients who received multiple lines of chemotherapy. METHODS Patients with refractory cancer from our center, who were included in a prospective randomized phase II trial comparing targeted therapies based on molecular profile of tumors versus conventional chemotherapy, were retrospectively included in this IRB-approved study. All patients had a biopsy of a tumor lesion performed during surgery, or using CT/palpation/endoscopic guidance. A biopsy was considered successful if the neoplastic cellularity was greater than 30%. Primary lesion, size and location of biopsied lesion, on-going chemotherapy and the differential attenuation between non-enhanced and venous phase (HU) for CT-guided biopsied lesions were recorded. RESULTS 228 patients (age=59±15yo; M/F=1.9) were included. One hundred and sixty biopsies (72%) of the 221 biopsies performed were successful. Prognostic factors of biopsy success were: no ongoing chemotherapy, surgical or palpation-guided biopsy, lymph nodes/soft tissue location(P <0.01). Among the 221 performed biopsies, 122 (55%) were performed using CT guidance and 82 (67%) were successful. In this subgroup, biopsied lesions located in lymph nodes/soft tissue were associated with a higher success rate while lung location was associated with failure (P <0.01). The mean differential attenuation was significantly higher in lesions with a successful biopsy (P <0.001). CONCLUSION Success of biopsy was less frequent with CT guidance than with surgical or palpation-guided biopsy and was higher in soft tissues and lymph nodes than that in visceral metastasis. Ongoing chemotherapy decreased tumor cell content and consequently the success of the biopsy samples for molecular profiling.
Collapse
|
22
|
Lin EI, Tseng LH, Gocke CD, Reil S, Le DT, Azad NS, Eshleman JR. Mutational profiling of colorectal cancers with microsatellite instability. Oncotarget 2016; 6:42334-44. [PMID: 26517354 PMCID: PMC4747229 DOI: 10.18632/oncotarget.5997] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2015] [Accepted: 10/05/2015] [Indexed: 01/11/2023] Open
Abstract
Microsatellite instability (MSI) is caused by defective mismatch repair in 15–20% of colorectal cancers (CRCs). Higher mutation loads in tumors with mismatch repair deficiency can predict response to pembrolizumab, an anti-programmed death 1 (PD-1) immune checkpoint inhibitor. We analyzed the mutations in 113 CRCs without MSI (MSS) and 29 CRCs with MSI-High (MSI-H) using the 50-gene AmpliSeq cancer panel. Overall, MSI-H CRCs showed significantly higher mutations than MSS CRCs, including insertion/deletion mutations at repeat regions. MSI-H CRCs showed higher incidences of mutations in the BRAF, PIK3CA, and PTEN genes as well as mutations in the receptor tyrosine kinase families. While the increased mutations in BRAF and PTEN in MSI-H CRCs are well accepted, we also support findings of mutations in the mTOR pathway and receptor tyrosine kinase family genes. MSS CRCs showed higher incidences of mutations in the APC, KRAS and TP53 genes, confirming previous findings. NGS assays may be designed to detect driver mutations for targeted therapeutics and to identify tumors with high mutation loads for potential treatment with immune checkpoint blockade therapies. Further studies may be warranted to elucidate potential targeted therapeutics against mutations in the mTOR pathway and the receptor tyrosine kinase family in MSI-H CRCs as well as the benefit of anti-PD-1 immunotherapy in hypermutated MSS CRCs or other cancers.
Collapse
Affiliation(s)
- Elaine I Lin
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Li-Hui Tseng
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Christopher D Gocke
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Stacy Reil
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Dung T Le
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Nilofer S Azad
- Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - James R Eshleman
- Department of Pathology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.,Department of Oncology and The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
23
|
Zhao X, Chang CC, Chuang TL, Lin CW. Detection of KRAS mutations of colorectal cancer with peptide-nucleic-acid-mediated real-time PCR clamping. BIOTECHNOL BIOTEC EQ 2016. [DOI: 10.1080/13102818.2016.1228479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Affiliation(s)
- Xihong Zhao
- Key Laboratory for Green Chemical Process of Ministry of Education, Hubei Key Laboratory of Novel Reactor and Green Chemical Technology, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology , Wuhan, China
- Institute of Biomedical Engineering, National Taiwan University , Taipei, Taiwan
| | - Chia-Chen Chang
- Institute of Biomedical Engineering, National Taiwan University , Taipei, Taiwan
| | - Tsung-Liang Chuang
- Institute of Biomedical Engineering, National Taiwan University , Taipei, Taiwan
| | - Chii-Wann Lin
- Institute of Biomedical Engineering, National Taiwan University , Taipei, Taiwan
| |
Collapse
|
24
|
Molecular Diagnostics for Precision Medicine in Colorectal Cancer: Current Status and Future Perspective. BIOMED RESEARCH INTERNATIONAL 2016; 2016:9850690. [PMID: 27699178 PMCID: PMC5028795 DOI: 10.1155/2016/9850690] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Accepted: 08/10/2016] [Indexed: 02/08/2023]
Abstract
Precision medicine, a concept that has recently emerged and has been widely discussed, emphasizes tailoring medical care to individuals largely based on information acquired from molecular diagnostic testing. As a vital aspect of precision cancer medicine, targeted therapy has been proven to be efficacious and less toxic for cancer treatment. Colorectal cancer (CRC) is one of the most common cancers and among the leading causes for cancer related deaths in the United States and worldwide. By far, CRC has been one of the most successful examples in the field of precision cancer medicine, applying molecular tests to guide targeted therapy. In this review, we summarize the current guidelines for anti-EGFR therapy, revisit the roles of pathologists in an era of precision cancer medicine, demonstrate the transition from traditional “one test-one drug” assays to multiplex assays, especially by using next-generation sequencing platforms in the clinical diagnostic laboratories, and discuss the future perspectives of tumor heterogeneity associated with anti-EGFR resistance and immune checkpoint blockage therapy in CRC.
Collapse
|
25
|
Lin MT, Tseng LH, Dudley JC, Riel S, Tsai H, Zheng G, Pratz KW, Levis MJ, Gocke CD. A Novel Tandem Duplication Assay to Detect Minimal Residual Disease in FLT3/ITD AML. Mol Diagn Ther 2016; 19:409-17. [PMID: 26446915 DOI: 10.1007/s40291-015-0170-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND Internal tandem duplication (ITD) of the fms-related tyrosine kinase 3 (FLT3) gene is associated with a poor prognosis in acute myeloid leukemia (AML) patients with a normal karyotype. The current standard polymerase chain reaction (PCR) assay for FLT3/ITD detection is not sufficiently sensitive to monitor minimal residual disease (MRD). Clone-specific assays may have sufficient sensitivity but are not practical to implement, since each clone-specific primer/probe requires clinical validation. OBJECTIVE To develop an assay for clinical molecular diagnostics laboratories to monitor MRD in FLT3/ITD AMLs. METHODS We designed a simple novel assay, tandem duplication PCR (TD-PCR), and tested its sensitivity, specificity, and clinical utility in FLT3/ITD AML patients. RESULTS TD-PCR was capable of detecting a single ITD molecule and was applicable to 75 % of ITD mutants tested. TD-PCR detected MRD in bone marrow prior to patient relapse. TD-PCR also identified low-level ITD mutants not only in FLT3/ITD AMLs but also in initial diagnostic specimens that were reportedly negative by the standard assay in patients who progressed with the same ITDs detected by the TD-PCR assay. CONCLUSION Detection of MRD by TD-PCR may guide patient selection for early clinical intervention. In contrast to clone-specific approaches, the TD-PCR assay can be more easily validated for MRD detection in clinical laboratories because it uses standardized primers and a universal positive control. In addition, our findings on multi-clonality and low-level ITDs suggest that further studies are warranted to elucidate their clinical/biological significance.
Collapse
Affiliation(s)
- Ming-Tseh Lin
- Division of Molecular Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Park SB202, 600 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Li-Hui Tseng
- Division of Molecular Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Park SB202, 600 North Wolfe Street, Baltimore, MD, 21287, USA
- Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Jonathan C Dudley
- Division of Molecular Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Park SB202, 600 North Wolfe Street, Baltimore, MD, 21287, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Stacey Riel
- Division of Molecular Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Park SB202, 600 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Harrison Tsai
- Division of Molecular Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Park SB202, 600 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Gang Zheng
- Division of Molecular Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Park SB202, 600 North Wolfe Street, Baltimore, MD, 21287, USA
| | - Keith W Pratz
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mark J Levis
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Christopher D Gocke
- Division of Molecular Pathology, Department of Pathology, Johns Hopkins University School of Medicine, Park SB202, 600 North Wolfe Street, Baltimore, MD, 21287, USA.
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
26
|
Zheng G, Tsai H, Tseng LH, Illei P, Gocke CD, Eshleman JR, Netto G, Lin MT. Test Feasibility of Next-Generation Sequencing Assays in Clinical Mutation Detection of Small Biopsy and Fine Needle Aspiration Specimens. Am J Clin Pathol 2016; 145:696-702. [PMID: 27247373 DOI: 10.1093/ajcp/aqw043] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVES To evaluate preanalytic factors contributing to failure of next-generation sequencing (NGS) assays. METHODS AmpliSeq Cancer Hotspot Panel was conducted in 1,121 of 1,152 formalin-fixed paraffin-embedded tissues submitted to a clinical laboratory, including 493 small biopsy or fine needle aspiration (FNA) specimens (44%) and 25 metastatic bone specimens (2.2%). RESULTS Single nucleotide mutations and/or insertion/deletion mutations were detected in 702 specimens. Thirty-eight specimens (3.4%) were reported as "no results" due to NGS assay failure. Higher failure rates were observed in specimens submitted for lung cancer panel and melanoma panel (3.1% and 3.7% vs 1.0% colorectal cancer panel), metastatic bone specimens (36% vs 2.6% nonbone specimens), referred specimens (5.0% vs 1.8% in-house specimens), and small biopsy and FNA specimens (5.8% and 3.1% vs 0.7% resection/excision specimens). Test feasibility was higher in in-house specimens than referred specimens (99.1% vs 96.9% in resection specimens, 94.4% vs 87.3% in small biopsy specimens, and 94.3% vs 58.8% in FNA specimens). CONCLUSIONS NGS assays demonstrated clinical utility in solid tumor specimens, including those taken by biopsy or FNA. Preanalytic factors identified by this study that may contribute to NGS assay failure highlight the need for pathologists to revisit tissue processing protocols in order to better optimize cancer mutational profiling.
Collapse
Affiliation(s)
| | | | - Li-Hui Tseng
- From the Departments of Pathology and Department of Medical Genetics, National Taiwan University Hospital, Taipei
| | | | - Christopher D Gocke
- From the Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - James R Eshleman
- From the Departments of Pathology and Oncology, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | | |
Collapse
|
27
|
Olmedillas López S, García-Olmo DC, García-Arranz M, Guadalajara H, Pastor C, García-Olmo D. KRAS G12V Mutation Detection by Droplet Digital PCR in Circulating Cell-Free DNA of Colorectal Cancer Patients. Int J Mol Sci 2016; 17:484. [PMID: 27043547 PMCID: PMC4848940 DOI: 10.3390/ijms17040484] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 03/21/2016] [Accepted: 03/24/2016] [Indexed: 12/14/2022] Open
Abstract
KRAS mutations are responsible for resistance to anti-epidermal growth factor receptor (EGFR) therapy in colorectal cancer patients. These mutations sometimes appear once treatment has started. Detection of KRAS mutations in circulating cell-free DNA in plasma (“liquid biopsy”) by droplet digital PCR (ddPCR) has emerged as a very sensitive and promising alternative to serial biopsies for disease monitoring. In this study, KRAS G12V mutation was analyzed by ddPCR in plasma DNA from 10 colorectal cancer patients and compared to six healthy donors. The percentage of KRAS G12V mutation relative to wild-type sequences in tumor-derived DNA was also determined. KRAS G12V mutation circulating in plasma was detected in 9 of 10 colorectal cancer patients whose tumors were also mutated. Colorectal cancer patients had 35.62 copies of mutated KRAS/mL plasma, whereas in healthy controls only residual copies were found (0.62 copies/mL, p = 0.0066). Interestingly, patients with metastatic disease showed a significantly higher number of mutant copies than M0 patients (126.25 versus 9.37 copies/mL, p = 0.0286). Wild-type KRAS was also significantly elevated in colorectal cancer patients compared to healthy controls (7718.8 versus 481.25 copies/mL, p = 0.0002). In conclusion, KRAS G12V mutation is detectable in plasma of colorectal cancer patients by ddPCR and could be used as a non-invasive biomarker.
Collapse
Affiliation(s)
- Susana Olmedillas López
- Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid 28040, Spain.
| | - Dolores C García-Olmo
- Experimental Research Unit, General University Hospital of Albacete, Albacete 02006, Spain.
| | - Mariano García-Arranz
- Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid 28040, Spain.
- Department of Surgery, School of Medicine, Autónoma University of Madrid, Madrid 28029, Spain.
| | - Héctor Guadalajara
- Department of Surgery, School of Medicine, Autónoma University of Madrid, Madrid 28029, Spain.
- Department of General Surgery, General Hospital of Villalba, Madrid 28400, Spain.
| | - Carlos Pastor
- Department of Surgery, Fundación Jiménez Díaz University Hospital, Madrid 28040, Spain.
| | - Damián García-Olmo
- Health Research Institute-Fundación Jiménez Díaz University Hospital (IIS-FJD), Madrid 28040, Spain.
- Department of Surgery, School of Medicine, Autónoma University of Madrid, Madrid 28029, Spain.
- Department of Surgery, Fundación Jiménez Díaz University Hospital, Madrid 28040, Spain.
| |
Collapse
|
28
|
Allison DB, Lilo MT, Geddes S, Pallavajjalla A, Askin F, Gabrielson E, Zheng G, Li QK. Detection of PIK3CA mutations, including a novel mutation of V344G in exon 4, in metastatic lung adenocarcinomas: A retrospective study of 115 FNA cases. Cancer Cytopathol 2016; 124:485-92. [PMID: 27007084 DOI: 10.1002/cncy.21714] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations and amplification are detected in 1% of primary lung adenocarcinomas (ADCs) and in 38% of primary lung squamous cell carcinomas. Alterations of PIK3CA in metastatic non-small cell lung carcinoma (NSCLC), however, are still not fully understood. This study investigated PIK3CA alterations in metastatic ADCs and correlated the findings with those for other commonly tested molecular abnormalities via fine-needle aspiration (FNA) and small-core biopsy materials. METHODS This study identified 115 FNA cases of metastatic lung ADC with standard lung cancer panel analysis by targeted next-generation sequencing and fluorescence in situ hybridization at the Johns Hopkins Medical Institute over a 12-month period. The panel included mutational analysis of PIK3CA, AKT, BRAF, EGFR, ERBB2, KRAS, and NRAS genes and tests of rearrangements for ALK and ROS1 genes. RESULTS A PIK3CA mutation was detected in 7 of 115 cases of metastatic ADC (6.1%). The majority of the mutations were located in exon 9 or exon 20; however, a mutation in exon 1 was seen in 1 case. Furthermore, p.V344G in exon 4 was detected in 2 cases. Among cases with PIK3CA mutations, 4 had coexisting EGFR mutations, whereas 2 had a coexisting BRAF or KRAS mutation. CONCLUSIONS Several common mutations as well as a novel mutation in the PIK3CA gene were observed in metastatic NSCLC (particularly ADC). The unique role, however, of PIK3CA mutations in metastatic NSCLC and the clinical implications need to be further investigated. Cancer Cytopathol 2016;124:485-92. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Derek B Allison
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Mohammed T Lilo
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Susan Geddes
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Aparna Pallavajjalla
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Frederic Askin
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Edward Gabrielson
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Gang Zheng
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| | - Qing Kay Li
- Department of Pathology, Johns Hopkins Medical Institutions, Baltimore, Maryland
| |
Collapse
|
29
|
Crumley SM, Pepper KL, Phan AT, Olsen RJ, Schwartz MR, Portier BP. Next-Generation Sequencing of Matched Primary and Metastatic Rectal Adenocarcinomas Demonstrates Minimal Mutation Gain and Concordance to Colonic Adenocarcinomas. Arch Pathol Lab Med 2015; 140:529-35. [PMID: 26536055 DOI: 10.5858/arpa.2015-0261-sa] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
CONTEXT -Colorectal carcinoma is the third most common cause of cancer death in males and females in the United States. Rectal adenocarcinoma can have distinct therapeutic and surgical management from colonic adenocarcinoma owing to its location and anatomic considerations. OBJECTIVE -To determine the oncologic driver mutations and better understand the molecular pathogenesis of rectal adenocarcinoma in relation to colon adenocarcinoma. DESIGN -Next-generation sequencing was performed on 20 cases of primary rectal adenocarcinoma with a paired lymph node or solid organ metastasis by using an amplicon-based assay of more than 2800 Catalogue of Somatic Mutations in Cancer (COSMIC)-identified somatic mutations. RESULTS -Next-generation sequencing data were obtained on both the primary tumor and metastasis from 16 patients. Most rectal adenocarcinoma cases demonstrated identical mutations in the primary tumor and metastasis (13 of 16, 81%). The mutations identified, listed in order of frequency, included TP53, KRAS, APC, FBXW7, GNAS, FGFR3, BRAF, NRAS, PIK3CA, and SMAD4. CONCLUSIONS -The somatic mutations identified in our rectal adenocarcinoma cohort showed a strong correlation to those previously characterized in colonic adenocarcinoma. In addition, most rectal adenocarcinomas harbored identical somatic mutations in both the primary tumor and metastasis. These findings demonstrate evidence that rectal adenocarcinoma follows a similar molecular pathogenesis as colonic adenocarcinoma and that sampling either the primary or metastatic lesion is valid for initial evaluation of somatic mutations and selection of possible targeted therapy.
Collapse
Affiliation(s)
- Suzanne M Crumley
- From the Departments of Pathology and Genomic Medicine (Drs Crumley, Olsen, Schwartz, and Portier and Ms Pepper) and Oncology (Dr Phan), Houston Methodist Hospital, Houston, Texas
| | - Kristi L Pepper
- From the Departments of Pathology and Genomic Medicine (Drs Crumley, Olsen, Schwartz, and Portier and Ms Pepper) and Oncology (Dr Phan), Houston Methodist Hospital, Houston, Texas
| | - Alexandria T Phan
- From the Departments of Pathology and Genomic Medicine (Drs Crumley, Olsen, Schwartz, and Portier and Ms Pepper) and Oncology (Dr Phan), Houston Methodist Hospital, Houston, Texas
| | - Randall J Olsen
- From the Departments of Pathology and Genomic Medicine (Drs Crumley, Olsen, Schwartz, and Portier and Ms Pepper) and Oncology (Dr Phan), Houston Methodist Hospital, Houston, Texas
| | - Mary R Schwartz
- From the Departments of Pathology and Genomic Medicine (Drs Crumley, Olsen, Schwartz, and Portier and Ms Pepper) and Oncology (Dr Phan), Houston Methodist Hospital, Houston, Texas
| | - Bryce P Portier
- From the Departments of Pathology and Genomic Medicine (Drs Crumley, Olsen, Schwartz, and Portier and Ms Pepper) and Oncology (Dr Phan), Houston Methodist Hospital, Houston, Texas
| |
Collapse
|
30
|
Haley L, Tseng LH, Zheng G, Dudley J, Anderson DA, Azad NS, Gocke CD, Eshleman JR, Lin MT. Performance characteristics of next-generation sequencing in clinical mutation detection of colorectal cancers. Mod Pathol 2015; 28:1390-9. [PMID: 26226847 PMCID: PMC4618462 DOI: 10.1038/modpathol.2015.86] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 06/02/2015] [Accepted: 06/05/2015] [Indexed: 02/07/2023]
Abstract
Activating mutations in downstream genes of the epidermal growth factor receptor (EGFR) pathway may cause anti-EGFR resistance in patients with colorectal cancers. We present performance characteristics of a next-generation sequencing assay designed to detect such mutations. In this retrospective quality assessment study, we analyzed mutation detected in the KRAS, NRAS, BRAF, and PIK3CA genes by a clinically validated next-generation sequencing assay in 310 colorectal cancer specimens. Tumor cellularity and mutant allele frequency were analyzed to identify tumor heterogeneity and mutant allele-specific imbalance. Next-generation sequencing showed precise measurement of mutant allele frequencies and detected 23% of mutations with 2-20% mutant allele frequencies. Of the KRAS mutations detected, 17% were outside of codons 12 and 13. Among PIK3CA mutations, 48% were outside of codons 542, 545, and 1047. The percentage of tumors with predicted resistance to anti-EGFR therapy increased from 40% when testing for only mutations in KRAS exon 2 to 47% when testing for KRAS exons 2-4, 48% when testing for KRAS and NRAS exons 2-4, 58% when including BRAF codon 600 mutations, and 59% when adding PIK3CA exon 20 mutations. Right-sided colorectal cancers carried a higher risk of predicted anti-EGFR resistance. A concomitant KRAS mutation was detected in 51% of PIK3CA, 23% of NRAS, and 33% of kinase-impaired BRAF-mutated tumors. Lower than expected mutant allele frequency indicated tumor heterogeneity, while higher than expected mutant allele frequency indicated mutant allele-specific imbalance. Two paired neuroendocrine carcinomas and adjacent adenomas showed identical KRAS mutations, but only PIK3CA mutations in neuroendocrine carcinomas. Next-generation sequencing is a robust tool for mutation detection in clinical laboratories. It demonstrates high analytic sensitivity and broad reportable range, and it provides simultaneous detection of concomitant mutations and a quantitative measurement of mutant allele frequencies to predict tumor heterogeneity and mutant allele-specific imbalance.
Collapse
Affiliation(s)
- Lisa Haley
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Li-Hui Tseng
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA,Department of Medical Genetics, National Taiwan University Hospital, Taipei, Taiwan
| | - Gang Zheng
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Jonathan Dudley
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA,Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Derek A Anderson
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Nilofer S Azad
- Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Christopher D Gocke
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA,Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - James R Eshleman
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA,Department of Oncology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Ming-Tseh Lin
- Department of Pathology, Johns Hopkins University School of Medicine, Johns Hopkins Hospital, Baltimore, MD, USA
| |
Collapse
|
31
|
Carter J, Tseng LH, Zheng G, Dudley J, Illei P, Gocke CD, Eshleman JR, Lin MT. Non-p.V600E BRAF Mutations Are Common Using a More Sensitive and Broad Detection Tool. Am J Clin Pathol 2015; 144:620-8. [PMID: 26386083 DOI: 10.1309/ajcp85atmjozoudj] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVES To assess the performance of a next-generation sequencing (NGS) platform for the clinical detection of BRAF mutations. METHODS In this retrospective quality assessment of an NGS assay, we analyzed BRAF mutations within parts of exons 11 and 15 in 835 neoplastic tissues submitted to our molecular diagnostics laboratory. RESULTS The NGS assays detected a BRAF mutation in 5.9% of lung adenocarcinomas, 13% of colorectal cancers, and 44% of melanomas. Mutant allele frequencies were less than 20% in 28% of 88 BRAF-mutated specimens. Two lymph node specimens with subcapsular or infiltrative metastasis showed 1% to 2% mutant alleles. There were 26 unique BRAF mutations in exons 11 and 15, including three novel mutations. Mutations were located outside codon 600 in 39% of BRAF-mutated tumors. Lung adenocarcinomas showed significantly higher non-p.V600E mutations (86%) than did colorectal cancers (23%) and melanomas (34%). The three most common BRAF mutations in lung cancers accounted for only 41% of the observed BRAF mutations (p.D594G [18%], p.V600E [14%], and p.G469A [9%]). CONCLUSIONS The NGS assay demonstrated a high analytic sensitivity and a broad reportable range for clinical detection of BRAF mutations. Elucidating the spectrum of non-p. V600E BRAF mutations in different malignancies is a first step toward understanding their clinical significance.
Collapse
Affiliation(s)
- Jamal Carter
- Departments of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Li-Hui Tseng
- Departments of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Medical Genetics, National Taiwan University Hospital, Taipei
| | - Gang Zheng
- Departments of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Jonathan Dudley
- Departments of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Pathology, Massachusetts General Hospital, Boston
| | - Peter Illei
- Departments of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Christopher D. Gocke
- Departments of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
- Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - James R. Eshleman
- Departments of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
- Oncology, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Ming-Tseh Lin
- Departments of Pathology, The Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
32
|
Al-Kateb H, Nguyen TT, Steger-May K, Pfeifer JD. Identification of major factors associated with failed clinical molecular oncology testing performed by next generation sequencing (NGS). Mol Oncol 2015; 9:1737-43. [PMID: 26071350 DOI: 10.1016/j.molonc.2015.05.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Revised: 05/05/2015] [Accepted: 05/07/2015] [Indexed: 12/21/2022] Open
Abstract
PURPOSE DNA analysis by NGS has become important to direct the clinical care of cancer patients. However, NGS is not successful in all cases, and the factors responsible for test failures have not been systematically evaluated. MATERIALS AND METHODS A series of 1528 solid and hematolymphoid tumor specimens was tested by an NGS comprehensive cancer panel during 2012-2014. DNA was extracted and 2×101 bp paired-end sequence reads were generated on cancer-related genes utilizing Illumina HiSeq and MiSeq platforms. RESULTS Testing was unsuccessful in 343 (22.5%) specimens. The failure was due to insufficient tissue (INST) in 223/343 (65%) cases, insufficient DNA (INS-DNA) in 99/343 (28.9%) cases, and failed library (FL) in 21/343 (6.1%) cases. 87/99 (88%) of the INS-DNA cases had below 10 ng DNA available for testing. Factors associated with INST and INS-DNA failures were site of biopsy (SOB) and type of biopsy (TOB) (both p < 0.0001), and clinical setting of biopsy (CSB, initial diagnosis or recurrence) (p < 0.0001). Factors common to INST and FL were age of specimen (p ≤ 0.006) and tumor viability (p ≤ 0.05). Factors common to INS-DNA and FL were DNA purity and DNA degradation (all p ≤ 0.005). In multivariate analysis, common predictors for INST and INS-DNA included CSB (p = 0.048 and p < 0.0001) and TOB (both p ≤ 0.003), respectively. SOB (p = 0.004) and number of cores (p = 0.001) were specific for INS-DNA, whereas TOB and DNA degradation were associated with FL (p = 0.04 and 0.02, respectively). CONCLUSIONS Pre-analytical causes (INST and INS-DNA) accounted for about 90% of all failed cases; independent of test design. Clinical setting; site and type of biopsy; and number of cores used for testing all correlated with failure. Accounting for these factors at the time of tissue biopsy acquisition could improve the analytic success rate.
Collapse
Affiliation(s)
- Hussam Al-Kateb
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA.
| | - TuDung T Nguyen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Karen Steger-May
- Division of Biostatistics, Washington University School of Medicine, St. Louis, USA
| | - John D Pfeifer
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| |
Collapse
|
33
|
VanderLaan PA. Molecular markers: Implications for cytopathology and specimen collection. Cancer Cytopathol 2015; 123:454-60. [DOI: 10.1002/cncy.21560] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Revised: 04/09/2015] [Accepted: 04/27/2015] [Indexed: 12/11/2022]
Affiliation(s)
- Paul A. VanderLaan
- Department of Pathology; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston Massachusetts
| |
Collapse
|