1
|
Nakamura Y, Ozaki H, Ueno M, Komatsu Y, Yuki S, Esaki T, Taniguchi H, Sunakawa Y, Yamaguchi K, Kato K, Denda T, Nishina T, Takahashi N, Satoh T, Yasui H, Satake H, Oki E, Kato T, Ohta T, Matsuhashi N, Goto M, Okano N, Ohtsubo K, Yamazaki K, Yamashita R, Iida N, Yuasa M, Bando H, Yoshino T. Targeted therapy guided by circulating tumor DNA analysis in advanced gastrointestinal tumors. Nat Med 2025; 31:165-175. [PMID: 39284955 PMCID: PMC11750700 DOI: 10.1038/s41591-024-03244-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 08/09/2024] [Indexed: 01/23/2025]
Abstract
Although comprehensive genomic profiling has become standard in oncology for advanced solid tumors, the full potential of circulating tumor DNA (ctDNA)-based profiling in capturing tumor heterogeneity and guiding therapy selection remains underexploited, marked by a scarcity of evidence on its clinical impact and the assessment of intratumoral heterogeneity. The GOZILA study, a nationwide, prospective observational ctDNA profiling study, previously demonstrated higher clinical trial enrollment rates using liquid biopsy compared with tissue screening. This updated analysis of 4,037 patients further delineates the clinical utility of ctDNA profiling in advanced solid tumors, showcasing a significant enhancement in patient outcomes with a 24% match rate for targeted therapy. Patients treated with matched targeted therapy based on ctDNA profiling demonstrated significantly improved overall survival compared with those receiving unmatched therapy (hazard ratio, 0.54). Notably, biomarker clonality and adjusted plasma copy number were identified as predictors of therapeutic efficacy, reinforcing the value of ctDNA in reflecting tumor heterogeneity for precise treatment decisions. These new insights into the relationship between ctDNA characteristics and treatment outcomes advance our understanding beyond the initial enrollment benefits. Our findings advocate for the broader adoption of ctDNA-guided treatment, signifying an advancement in precision oncology and improving survival outcomes in advanced solid tumors.
Collapse
Affiliation(s)
- Yoshiaki Nakamura
- International Research Promotion Office, National Cancer Center Hospital East, Kashiwa, Japan
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroshi Ozaki
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Makoto Ueno
- Department of Gastroenterology, Kanagawa Cancer Center, Yokohama, Japan
| | - Yoshito Komatsu
- Department of Cancer Center, Hokkaido University Hospital, Sapporo, Japan
| | - Satoshi Yuki
- Department of Gastroenterology and Hepatology, Hokkaido University Hospital, Sapporo, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Hiroya Taniguchi
- Department of Clinical Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, Cancer Institute Hospital of Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Tadamichi Denda
- Division of Gastroenterology, Chiba Cancer Center, Chiba, Japan
| | - Tomohiro Nishina
- Gastrointestinal Medical Oncology, NHO Shikoku Cancer Center, Matsuyama, Japan
| | - Naoki Takahashi
- Department of Gastroenterology, Saitama Cancer Center, Ina, Japan
| | - Taroh Satoh
- Center for Cancer Genomics and Precision Medicine, Osaka University Hospital, Suita, Japan
| | - Hisateru Yasui
- Department of Medical Oncology, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hironaga Satake
- Cancer Treatment Center, Kansai Medical University Hospital, Hirakata, Japan
- Department of Medical Oncology, Kochi Medical School, Nankoku, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Kato
- Department of Surgery, NHO Osaka National Hospital, Osaka, Japan
| | - Takashi Ohta
- Department of Clinical Oncology, Kansai Rosai Hospital, Amagasaki, Japan
| | - Nobuhisa Matsuhashi
- Department of Gastroenterological Surgery and Pediatric Surgery, Center for One Medicine Innovative Translational Research (COMIT), Gifu University Graduate School of Medicine, Gifu, Japan
| | - Masahiro Goto
- Cancer Chemotherapy Center, Osaka Medical and Pharmaceutical University Hospital, Takatsuki, Japan
| | - Naohiro Okano
- Department of Medical Oncology, Kyorin University Faculty of Medicine, Tokyo, Japan
| | - Koushiro Ohtsubo
- Department of Medical Oncology, Kanazawa University Hospital, Kanazawa, Japan
| | - Kentaro Yamazaki
- Division of Gastrointestinal Oncology, Shizuoka Cancer Center, Nagaizumi, Japan
| | - Riu Yamashita
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, Kashiwa, Japan
| | - Naoko Iida
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Mihoko Yuasa
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hideaki Bando
- Translational Research Support Office, National Cancer Center Hospital East, Kashiwa, Japan
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Takayuki Yoshino
- Department of Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan.
- Kindai University Faculty of Medicine, Osakasayama, Japan.
| |
Collapse
|
2
|
Takeuchi S, Yoshimura A, Sofuni A, Ueda Y, Umezu T, Kuroda M, Sukeda A, Matsubayashi J, Nagao T, Bingo M, Inagaki N, Ohira T, Seike M, Ikeda N. A single-institution retrospective study of comprehensive genomic profiling tests based on C-CAT findings for advanced solid cancers. Jpn J Clin Oncol 2024; 54:1298-1305. [PMID: 39297618 DOI: 10.1093/jjco/hyae128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 08/29/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND In recent years, as the availability of precision therapies expands, there is increasing reliance on genomic profiling assays to help identify the most appropriate treatment options for patients with advanced cancers. We retrospectively investigated the results of comprehensive genomic profiling tests from the time insurance coverage began until recently and examined the status of genetic analysis. METHODS We retrospectively reviewed the analysis results of 300 patients with advanced solid tumors who consented to comprehensive genomic profiling tests from October 2019 to December 2022. RESULTS Of the 300 patients who underwent comprehensive genomic profiling tests, analysis results for 274 patients were obtained, and were reviewed by the Clinical Genome Expert Panel. Six specimens (2%) were discontinued due to patient deaths and deteriorations in general condition. The three most frequently occurring actionable genomic alterations observed were TP53 (47.4%), KRAS (28.1%) and CDKN2A (20.4%). The most common druggable variant was CDKN2A, which was noted in 52 (19%) of 274 patients. The next most common were PIK3CA, BRAF, KRAS and PTEN. The cancer types that showed a greater median number of actionable alterations comprised thyroid cancer, pancreatic cancer and colorectal cancer. CONCLUSIONS In conclusion, comprehensive genomic profiling tests have the potential to be valuable in identifying genomic abnormalities. Even if there is no effective treatment at present, it may lead to a treatment in the future. Comprehensive genomic profiling tests should be considered for any cancer.
Collapse
Affiliation(s)
- Susumu Takeuchi
- Department of Surgery, Tokyo Medical University, Tokyo, Japan
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Akinobu Yoshimura
- Department of Clinical Oncology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Atsushi Sofuni
- Department of Clinical Oncology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Yuri Ueda
- Otorhinolaryngology, Head and Neck Surgery, Tokyo Medical University, Tokyo, Japan
| | - Tomohiro Umezu
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Masahiko Kuroda
- Department of Molecular Pathology, Tokyo Medical University, Tokyo, Japan
| | - Aoi Sukeda
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Jun Matsubayashi
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Toshitaka Nagao
- Department of Anatomic Pathology, Tokyo Medical University, Tokyo, Japan
| | - Masato Bingo
- Clinical Genetics Center, Tokyo Medical University Hospital, Tokyo, Japan
| | - Natsuko Inagaki
- Clinical Genetics Center, Tokyo Medical University Hospital, Tokyo, Japan
| | - Tatsuo Ohira
- Department of Surgery, Tokyo Medical University, Tokyo, Japan
| | - Masahiro Seike
- Department of Pulmonary Medicine and Oncology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Norihiko Ikeda
- Department of Surgery, Tokyo Medical University, Tokyo, Japan
| |
Collapse
|
3
|
Takeshita K, Hijioka S, Nagashio Y, Hara H, Agarie D, Kawasaki Y, Takasaki T, Yagi S, Hagiwara Y, Okamoto K, Yamashige D, Fukuda S, Kuwada M, Komori Y, Okada M, Maruki Y, Morizane C, Ueno H, Yatabe Y, Okusaka T. Study Protocol for a Prospective Self-Controlled Trial on Success in Meeting Comprehensive Genomic Profiling Analysis Criteria for Specimens Obtained by Endoscopic Ultrasound-Guided Tissue Acquisition Using a 19G Needle from Primary and Metastatic Lesions in Pancreatic Cancer with Metastatic Lesions: The PRIMATE Study. Diseases 2024; 12:182. [PMID: 39195181 DOI: 10.3390/diseases12080182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
EUS-TA in unresectable pancreatic cancer requires not only a tissue diagnosis but also tissue collection in anticipation of comprehensive genomic profiling. However, the optimal puncture target remains controversial. Therefore, the Primary and Metastatic Lesions in Pancreatic Cancer (PRIMATE) study was designed to clarify the optimal target by comparing the success rates for meeting OncoGuide NCC Oncopanel (NOP) analysis criteria on pre-check primary and metastatic lesion specimens obtained during the same EUS-TA session in patients with invasive pancreatic ductal adenocarcinoma. In this ongoing prospective study, two specimens, each from primary and metastatic lesions, are obtained by EUS-TA (typically using a 19G fine-needle biopsy needle) in patients with invasive pancreatic ductal adenocarcinoma. The primary endpoint is the proportion of EUS-TA specimens that meet NOP analysis criteria during pre-check (i.e., tumor cellularity of ≥20% and a tissue area of ≥4 mm2), which are then compared between primary and metastatic lesions. This study has been approved by the National Cancer Center Institutional Review Board (Research No. 2022-168). The results of this study will be reported at an international conference and published in an international peer-reviewed journal. The trial registration number is UMIN 000048966.
Collapse
Affiliation(s)
- Kotaro Takeshita
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
- Department of Gastroenterology, Tane General Hospital, Osaka 550-0025, Japan
| | - Susumu Hijioka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yoshikuni Nagashio
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Hidenobu Hara
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Daiki Agarie
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yuki Kawasaki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Tetsuro Takasaki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Shin Yagi
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yuya Hagiwara
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Kohei Okamoto
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Daiki Yamashige
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Soma Fukuda
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Masaru Kuwada
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yasuhiro Komori
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Mao Okada
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yuta Maruki
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Chigusa Morizane
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Takuji Okusaka
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| |
Collapse
|
4
|
Takahashi Y, Morimura R, Tsukamoto K, Gomi S, Yamada A, Mizukami M, Naito Y, Irie S, Nagayama S, Shinozaki E, Yamaguchi K, Fujita N, Kitano S, Katayama R, Matsusaki M. In vitro throughput screening of anticancer drugs using patient-derived cell lines cultured on vascularized three-dimensional stromal tissues. Acta Biomater 2024; 183:111-129. [PMID: 38801868 DOI: 10.1016/j.actbio.2024.05.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 05/06/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
The development of high-throughput anticancer drug screening methods using patient-derived cancer cell (PDC) lines that maintain their original characteristics in an in vitro three-dimensional (3D) culture system poses a significant challenge to achieving personalized cancer medicine. Because stromal tissue plays a critical role in the composition and maintenance of the cancer microenvironment, in vitro 3D-culture using reconstructed stromal tissues has attracted considerable attention. Here, a simple and unique in vitro 3D-culture method using heparin and collagen together with fibroblasts and endothelial cells to fabricate vascularized 3D-stromal tissues for in vitro culture of PDCs is reported. Whereas co-treatment with bevacizumab, a monoclonal antibody against vascular endothelial growth factor, and 5-fluorouracil significantly reduced the survival rate of 3D-cultured PDCs to 30%, separate addition of each drug did not induce comparable strong cytotoxicity, suggesting the possibility of evaluating the combined effect of anticancer drugs and angiogenesis inhibitors. Surprisingly, drug evaluation using eight PDC lines with the 3D-culture method resulted in a drug efficacy concordance rate of 75% with clinical outcomes. The model is expected to be applicable to in vitro throughput drug screening for the development of personalized cancer medicine. STATEMENT OF SIGNIFICANCE: To replicate the cancer microenvironment, we constructed a cancer-stromal tissue model in which cancer cells are placed above and inside stromal tissue with vascular network structures derived from vascular endothelial cells in fibroblast tissue using CAViTs method. Using this method, we were able to reproduce the invasion and metastasis processes of cancer cells observed in vivo. Using patient-derived cancer cells, we assessed the possibility of evaluating the combined effect with an angiogenesis inhibitor. Further, primary cancer cells also grew on the stromal tissues with the normal medium. These data suggest that the model may be useful for new in vitro drug screening and personalized cancer medicine.
Collapse
Affiliation(s)
- Yuki Takahashi
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Division of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Rii Morimura
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Division of Clinical Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Kei Tsukamoto
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan
| | - Sayaka Gomi
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan
| | - Asuka Yamada
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Miki Mizukami
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan
| | - Yasuyuki Naito
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Shinji Irie
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan
| | - Satoshi Nagayama
- Department of Colorectal Surgery, Gastroenterological Cancer Center, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan; Department of Surgery, Uji Tokushukai Medical Center, Kyoto 611-0041, Japan
| | - Eiji Shinozaki
- Department of Gastroenterological Chemotherapy, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Kensei Yamaguchi
- Department of Gastroenterological Chemotherapy, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Naoya Fujita
- Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Shiro Kitano
- Business Development Division, Technical Research Institute, TOPPAN Holdings Inc., Saitama 345-8508, Japan; Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan.
| | - Ryohei Katayama
- Division of Experimental Chemotherapy, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan.
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Osaka 565-0871, Japan; Department of Applied Chemistry Graduate School of Engineering Osaka University, Osaka 565-0871, Japan.
| |
Collapse
|
5
|
Zalis M, Viana Veloso GG, Aguiar Jr. PN, Gimenes N, Reis MX, Matsas S, Ferreira CG. Next-generation sequencing impact on cancer care: applications, challenges, and future directions. Front Genet 2024; 15:1420190. [PMID: 39045325 PMCID: PMC11263191 DOI: 10.3389/fgene.2024.1420190] [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: 04/19/2024] [Accepted: 06/13/2024] [Indexed: 07/25/2024] Open
Abstract
Fundamentally precision oncology illustrates the path in which molecular profiling of tumors can illuminate their biological behavior, diversity, and likely outcomes by identifying distinct genetic mutations, protein levels, and other biomarkers that underpin cancer progression. Next-generation sequencing became an indispensable diagnostic tool for diagnosis and treatment guidance in current clinical practice. Nowadays, tissue analysis benefits from further support through methods like comprehensive genomic profiling and liquid biopsies. However, precision medicine in the field of oncology presents specific hurdles, such as the cost-benefit balance and widespread accessibility, particularly in countries with low- and middle-income. A key issue is how to effectively extend next-generation sequencing to all cancer patients, thus empowering treatment decision-making. Concerns also extend to the quality and preservation of tissue samples, as well as the evaluation of health technologies. Moreover, as technology advances, novel next-generation sequencing assessments are being developed, including the study of Fragmentomics. Therefore, our objective was to delineate the primary uses of next-generation sequencing, discussing its' applications, limitations, and prospective paths forward in Oncology.
Collapse
Affiliation(s)
- Mariano Zalis
- Oncoclínicas&Co/MedSir, Rio de Janeiro, Brazil
- Medical School of the Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gilson Gabriel Viana Veloso
- Oncoclínicas&Co/MedSir, Rio de Janeiro, Brazil
- Santa Casa de Misericórdia de Belo Horizonte, Belo Horizonte, Brazil
| | | | | | | | - Silvio Matsas
- Centro de Estudos e Pesquisas de Hematologia e Oncologia (CEPHO), Sao Paulo, Brazil
| | | |
Collapse
|
6
|
Ishikawa M, Nakamura K, Kawano R, Hayashi H, Ikeda T, Saito M, Niida Y, Sasaki J, Okuda H, Ishihara S, Yamaguchi M, Shimada H, Isobe T, Yuza Y, Yoshimura A, Kuroda H, Yukisawa S, Aoki T, Takeshita K, Ueno S, Nakazawa J, Sunakawa Y, Nohara S, Okada C, Nishimiya K, Tanishima S, Nishihara H. Clinical and Diagnostic Utility of Genomic Profiling for Digestive Cancers: Real-World Evidence from Japan. Cancers (Basel) 2024; 16:1504. [PMID: 38672586 PMCID: PMC11048180 DOI: 10.3390/cancers16081504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/12/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
The usefulness of comprehensive genomic profiling (CGP) in the Japanese healthcare insurance system remains underexplored. Therefore, this large-scale study aimed to determine the usefulness of CGP in diagnosing digestive cancers. Patients with various cancer types recruited between March 2020 and October 2022 underwent the FoundationOne® CDx assay at the Keio PleSSision Group (19 hospitals in Japan). A scoring system was developed to identify potentially actionable genomic alterations of biological significance and actionable genomic alterations. The detection rates for potentially actionable genomic alterations, actionable genomic alterations, and alterations equivalent to companion diagnosis (CDx), as well as the signaling pathways associated with these alterations in each digestive cancer, were analyzed. Among the 1587 patients, 547 had digestive cancer. The detection rates of potentially actionable genomic alterations, actionable genomic alterations, and alterations equivalent to CDx were 99.5%, 62.5%, and 11.5%, respectively. APC, KRAS, and CDKN2A alterations were frequently observed in colorectal, pancreatic, and biliary cancers, respectively. Most digestive cancers, except esophageal cancer, were adenocarcinomas. Thus, the classification flowchart for digestive adenocarcinomas proposed in this study may facilitate precise diagnosis. CGP has clinical and diagnostic utility in digestive cancers.
Collapse
Affiliation(s)
- Marin Ishikawa
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Integrated Medical Research Building 3-S5, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (R.K.); (H.H.); (S.T.); (H.N.)
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Integrated Medical Research Building 3-S5, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (R.K.); (H.H.); (S.T.); (H.N.)
| | - Ryutaro Kawano
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Integrated Medical Research Building 3-S5, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (R.K.); (H.H.); (S.T.); (H.N.)
| | - Hideyuki Hayashi
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Integrated Medical Research Building 3-S5, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (R.K.); (H.H.); (S.T.); (H.N.)
| | - Tatsuru Ikeda
- Department of Cancer Genome Medical Center, Hakodate Goryoukaku Hospital, 38-3, Goryoukakucho, Hakodate-shi 040-8611, Hokkaido, Japan;
| | - Makoto Saito
- Department of Genetic Medicine, Ibaraki Prefectural Center Hospital, 6528, Koibuchi, Kasama-shi 309-1793, Ibaraki, Japan;
| | - Yo Niida
- Center for Clinical Genomics, Kanazawa Medical University Hospital, 1-1, Daigaku, Uchinada 920-0293, Ishikawa, Japan;
| | - Jiichiro Sasaki
- Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, 1-15-1 Kitasato, Minami-ku, Sagamihara-shi 252-0329, Kanagawa, Japan;
| | - Hiroyuki Okuda
- Department of Medical Oncology, Keiyukai Sapporo Hospital, 1-1 Minami, Hondori 9, Chome, Shiroishi-ku, Sapporo 003-0026, Hokkaido, Japan;
| | - Satoshi Ishihara
- Cancer Genome Diagnosis and Treatment Center, Central Japan International Medical Center, 1-1 Kenkonomachi, Minokamo-shi 505-0010, Gifu, Japan;
| | - Masatoshi Yamaguchi
- Division of Clinical Genetics, Faculty of Medicine, University of Miyazaki Hospital, 5200 Kihara, Kiyotake-cho, Miyazaki-shi 889-1692, Miyazaki, Japan;
| | - Hideaki Shimada
- Department of Surgery and Clinical Oncology, Toho University Graduate School of Medicine, 6-11-1 Omori-nishi, Ota-ku, Tokyo 143-8541, Japan;
| | - Takeshi Isobe
- Cancer Genome Medical Center, Shimane University Hospital, 89-1, Enya-cho, Izumo-shi 693-8501, Shimane, Japan;
| | - Yuki Yuza
- Department of Hematology and Oncology, Tokyo Metropolitan Children’s Medical Center, 2-8-29 Musashidai, Fuchu-shi 183-8561, Tokyo, Japan;
| | - Akinobu Yoshimura
- Department of Clinical Oncology Director, Outpatient Chemotherapy Center, Tokyo Medical University Hospital, 6-7-1 Nishishinjuku, Shinjuku-ku, Tokyo 160-0023, Japan;
| | - Hajime Kuroda
- Department of Pathology, Tokyo Women’s Medical University, Adachi Medical Center, 4-33-1 Kohta, Adachi-ku, Tokyo 123-8558, Japan;
| | - Seigo Yukisawa
- Department of Medical Oncology, Saiseikai Utsunomiya Hospital, 911-1, Takebayashi, Utsunomiya-shi 321-0974, Tochigi, Japan;
| | - Takuya Aoki
- Department of Clinical Oncology, Tokyo Medical University Hachioji Medical Center, 1163, Tatemachi, Hachioji-shi 193-0998, Tokyo, Japan;
| | - Kei Takeshita
- Department of Clinical Genetics, Tokai University Hospital, 143, Shimokasuya, Isehara-shi 259-1193, Kanagawa, Japan;
| | - Shinichi Ueno
- Oncology Center, Kagoshima University Hospital, 8-35-1 Sakuragaoka, Kagoshima-shi 890-0075, Kagoshima, Japan;
| | - Junichi Nakazawa
- Department of Medical Oncology, Kagoshima City Hospital, 37-1, Uearatacho, Kagoshima-shi 890-8760, Kagoshima, Japan;
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, 2-16-1 Sugao, Miyamae-ku, Kawasaki 216-8511, Kanagawa, Japan;
| | - Sachio Nohara
- Biomedical Informatics Department, Communication Engineering Center, Mitsubishi Electric Software Corporation, Fuji Techno-Square, 5-4-36 Tsukaguchi-Honmachi, Amagasaki-shi 661-0001, Hyogo, Japan; (S.N.); (C.O.); (K.N.)
| | - Chihiro Okada
- Biomedical Informatics Department, Communication Engineering Center, Mitsubishi Electric Software Corporation, Fuji Techno-Square, 5-4-36 Tsukaguchi-Honmachi, Amagasaki-shi 661-0001, Hyogo, Japan; (S.N.); (C.O.); (K.N.)
| | - Ko Nishimiya
- Biomedical Informatics Department, Communication Engineering Center, Mitsubishi Electric Software Corporation, Fuji Techno-Square, 5-4-36 Tsukaguchi-Honmachi, Amagasaki-shi 661-0001, Hyogo, Japan; (S.N.); (C.O.); (K.N.)
| | - Shigeki Tanishima
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Integrated Medical Research Building 3-S5, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (R.K.); (H.H.); (S.T.); (H.N.)
- Biomedical Informatics Department, Communication Engineering Center, Mitsubishi Electric Software Corporation, Fuji Techno-Square, 5-4-36 Tsukaguchi-Honmachi, Amagasaki-shi 661-0001, Hyogo, Japan; (S.N.); (C.O.); (K.N.)
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Integrated Medical Research Building 3-S5, 35, Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan; (K.N.); (R.K.); (H.H.); (S.T.); (H.N.)
| |
Collapse
|
7
|
Arjuna S, Shah M, Dono A, Nunez-Rubiano L, Pichardo-Rojas PS, Zhu JJ, Riascos RF, Luthra R, Roy-Chowdhuri S, Duose D, Wang DH, Lang FF, Esquenazi Y, Ballester LY. Rapid detection of mutations in CSF-cfTNA with the Genexus Integrated Sequencer. J Neurooncol 2024; 166:39-49. [PMID: 38160230 PMCID: PMC11264431 DOI: 10.1007/s11060-023-04487-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/20/2023] [Indexed: 01/03/2024]
Abstract
PURPOSE Genomic alterations are fundamental for molecular-guided therapy in patients with breast and lung cancer. However, the turn-around time of standard next-generation sequencing assays is a limiting factor in the timely delivery of genomic information for clinical decision-making. METHODS In this study, we evaluated genomic alterations in 54 cerebrospinal fluid samples from 33 patients with metastatic lung cancer and metastatic breast cancer to the brain using the Oncomine Precision Assay on the Genexus sequencer. There were nine patients with samples collected at multiple time points. RESULTS Cell-free total nucleic acids (cfTNA) were extracted from CSF (0.1-11.2 ng/μl). Median base coverage was 31,963× with cfDNA input ranging from 2 to 20 ng. Mutations were detected in 30/54 CSF samples. Nineteen (19/24) samples with no mutations detected had suboptimal DNA input (< 20 ng). The EGFR exon-19 deletion and PIK3CA mutations were detected in two patients with increasing mutant allele fraction over time, highlighting the potential of CSF-cfTNA analysis for monitoring patients. Moreover, the EGFR T790M mutation was detected in one patient with prior EGFR inhibitor treatment. Additionally, ESR1 D538G and ESR1::CCDC170 alterations, associated with endocrine therapy resistance, were detected in 2 mBC patients. The average TAT from cfTNA-to-results was < 24 h. CONCLUSION In summary, our results indicate that CSF-cfTNA analysis with the Genexus-OPA can provide clinically relevant information in patients with brain metastases with short TAT.
Collapse
Affiliation(s)
- Srividya Arjuna
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA
| | - Mauli Shah
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA
| | - Antonio Dono
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School at UT Health, Houston, TX, USA
| | - Luis Nunez-Rubiano
- Department of Diagnostic and Interventional Imaging, McGovern Medical School at UT Health, Houston, TX, USA
| | - Pavel S Pichardo-Rojas
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School at UT Health, Houston, TX, USA
| | - Jay-Jiguang Zhu
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School at UT Health, Houston, TX, USA
- Memorial Hermann Hospital-TMC, Houston, TX, USA
| | - Roy F Riascos
- Department of Diagnostic and Interventional Imaging, McGovern Medical School at UT Health, Houston, TX, USA
| | - Rajyalakshmi Luthra
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center at Houston, Houston, TX, USA
| | - Sinchita Roy-Chowdhuri
- Department of Pathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA
| | - Dzifa Duose
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA
| | - Daniel H Wang
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA
| | - Yoshua Esquenazi
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School at UT Health, Houston, TX, USA.
- Memorial Hermann Hospital-TMC, Houston, TX, USA.
- Center for Precision Health, Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, TX, USA.
| | - Leomar Y Ballester
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA.
- Department of Pathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA.
- Neuropathology and Molecular Genetic Pathology, Department of Pathology, The University of Texas MD Anderson Cancer Center Houston, Houston, TX, USA.
| |
Collapse
|
8
|
Pankiw M, Brezden-Masley C, Charames GS. Comprehensive genomic profiling for oncological advancements by precision medicine. Med Oncol 2023; 41:1. [PMID: 37993657 DOI: 10.1007/s12032-023-02228-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/25/2023] [Indexed: 11/24/2023]
Abstract
Considerable advancements in next generation sequencing (NGS) techniques have sparked the use of comprehensive genomic profiling (CGP) as a guiding tool for precision-centered oncological treatments. The past two decades have seen the completion of the human genome project, and the consequential invention of NGS. High-throughput sequencing technologies support the discovery and commonplace use of individualized cancer treatments, specifically immune-centered checkpoint inhibitor therapies, and oncogene and tumor suppressor gene targeted therapies. Nevertheless, CGP is not commonly used in all clinical settings. This review investigates the clinically relevant applications of CGP. Studies published between the years 2000-2023 have shown substantial evidence of the benefits of integrating CGP into routine care practice, while also making important comparisons to current-standard oncological treatment strategies. Findings of a comprehensive genomic profile includes predictive, prognostic, and diagnostic biomarkers, together with somatic mutation identification which can indicate the efficacy of immunotherapies and molecularly guided therapies. This review highlights the importance of CGP in identifying driver mutations in tumors that subsequently can be effectively targeted with molecular therapeutics and lead to drug discovery, allowing for increased precision in treating tumors selectively based on their specific genetic mutations, thereby improving patient outcomes.
Collapse
Affiliation(s)
- Maya Pankiw
- Department of Medicine, Mount Sinai Hospital, Toronto, ON, Canada
- Department of Pathology and Lab Medicine, Mount Sinai Hospital, Toronto, ON, Canada
| | - Christine Brezden-Masley
- Department of Medicine, Mount Sinai Hospital, Toronto, ON, Canada
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - George S Charames
- Department of Pathology and Lab Medicine, Mount Sinai Hospital, Toronto, ON, Canada.
- Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Toronto, ON, Canada.
- Department of Lab Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.
- Mount Sinai Services, Toronto, ON, Canada.
| |
Collapse
|
9
|
Normanno N, Machado JC, Pescarmona E, Buglioni S, Navarro L, Esposito Abate R, Ferro A, Mensink R, Lambiase M, Lespinet-Fabre V, Calgua B, Jermann PM, Ilié M, Hofman P. European Real-World Assessment of the Clinical Validity of a CE-IVD Panel for Ultra-Fast Next-Generation Sequencing in Solid Tumors. Int J Mol Sci 2023; 24:13788. [PMID: 37762091 PMCID: PMC10531166 DOI: 10.3390/ijms241813788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/02/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Molecular profiling of solid tumors facilitates personalized, targeted therapeutic interventions. The ability to perform next-generation sequencing (NGS), especially from small tissue samples, in a short turnaround time (TAT) is essential to providing results that enable rapid clinical decisions. This multicenter study evaluated the performance of a CE in vitro diagnostic (IVD) assay, the Oncomine Dx Express Test, on the Ion Torrent Genexus System for detecting DNA and RNA variants in solid tumors. Eighty-two archived formalin-fixed paraffin embedded (FFPE) tissue samples from lung, colorectal, central nervous system, melanoma, breast, gastric, thyroid, and soft tissue cancers were used to assess the presence of single nucleotide variants (SNVs), insertions and deletions (indels), copy number variations (CNVs), gene fusions, and splice variants. These clinical samples were previously characterized at the various academic centers using orthogonal methods. The Oncomine Dx Express Test showed high performance with 100% concordance with previous characterization for SNVs, indels, CNVs, gene fusions, and splice variants. SNVs and indels with allele frequencies as low as 5% were correctly identified. The test detected all the expected ALK, RET, NTRK1, and ROS1 fusion isoforms and MET exon 14-skipping splice variants. The average TAT from extracted nucleic acids to the final variant report was 18.3 h. The Oncomine Dx Express Test in combination with the Ion Torrent Genexus System is a CE-IVD-compliant, performant, and multicenter reproducible method for NGS detection of actionable biomarkers from a range of tumor samples, providing results in a short TAT that could support timely decision- making for targeted therapeutic interventions.
Collapse
Affiliation(s)
- Nicola Normanno
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - José Carlos Machado
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
- Department of Pathology, Faculty of Medicine, University of Porto (FMUP), 4200-319 Porto, Portugal
| | - Edoardo Pescarmona
- I.R.C.C.S. Regina Elena National Cancer Institute, 00144 Rome, Italy; (E.P.); (S.B.)
| | - Simonetta Buglioni
- I.R.C.C.S. Regina Elena National Cancer Institute, 00144 Rome, Italy; (E.P.); (S.B.)
| | - Lara Navarro
- Consorcio Hospital General de Valencia, 46014 Valencia, Spain;
| | - Riziero Esposito Abate
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - Anabela Ferro
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Rob Mensink
- Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, 4200-135 Porto, Portugal; (J.C.M.); (A.F.); (R.M.)
- Institute of Molecular Pathology and Immunology, University of Porto (Ipatimup), 4200-135 Porto, Portugal
| | - Matilde Lambiase
- Cell Biology and Biotherapy Unit, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Naples, Italy; (N.N.); (R.E.A.)
| | - Virginie Lespinet-Fabre
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| | - Byron Calgua
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (B.C.); (P.M.J.)
| | - Philip M. Jermann
- Institute of Pathology, University Hospital Basel, 4031 Basel, Switzerland; (B.C.); (P.M.J.)
| | - Marius Ilié
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, Biobank BB-0033-00025, FHU OncoAge, IHU RespirERA, CHU de Nice, Université Côte d’Azur, 06000 Nice, France; (V.L.-F.); (M.I.)
| |
Collapse
|
10
|
Matsuo M, Hashimoto K, Kogo R, Jiromaru R, Hongo T, Manako T, Nakagawa T. Utility of Precision Oncology Using Cancer Genomic Profiling for Head and Neck Malignancies. In Vivo 2023; 37:2147-2154. [PMID: 37652518 PMCID: PMC10500534 DOI: 10.21873/invivo.13312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Revised: 05/21/2023] [Accepted: 06/06/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND/AIM In recent years, individual patient cancer genomic profiling (CGP) has become more accessible, allowing determination of therapeutic strategies using driver gene mutations in cancer therapy. However, this precision oncology approach, tailored to specific patients, remains experimental. In this study, we verified the feasibility and benefit of using CGP to guide treatment of malignant head and neck tumors. We aimed to evaluate the profiling and clinical courses of patients with head and neck malignancies who underwent CGP and determine the extent to which CGP for head and neck malignancies has resulted in beneficial drug administration. PATIENTS AND METHODS We analyzed CGP results, prognosis, and drug administration status in 27 patients. These patients had completed (or were expected to complete) standard therapy or had rare cancers without standard therapy. RESULTS At least one somatic actionable gene alteration was seen in 25 (92.6%) patients, with a median number of actionable alterations per patient of 4 (range=0-11). Drugs in clinical trials were recommended to 22 (81.5%) patients, but none could participate. However, 3 patients (11.1%) could use approved drugs off-label based on CGP results. The most common genetic abnormality was TP53 (66.7%), with TP53 mutations leading to poor prognosis. CONCLUSION CGP is clinically useful and serves as a bridge to increase the number of therapeutic options. However, candidate drugs confirmed using CGP may be ineffective when administered. Therefore, oncologists should not blindly accept CGP therapeutic recommendations but should make recommendations that lead to optimal therapies after proper verification.
Collapse
Affiliation(s)
- Mioko Matsuo
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuki Hashimoto
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryunosuke Kogo
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Rina Jiromaru
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takahiro Hongo
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tomomi Manako
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takashi Nakagawa
- Department of Otorhinolaryngology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
11
|
Matsubara J, Mukai K, Kondo T, Yoshioka M, Kage H, Oda K, Kudo R, Ikeda S, Ebi H, Muro K, Hayashi R, Tokudome N, Yamamoto N, Muto M. First-Line Genomic Profiling in Previously Untreated Advanced Solid Tumors for Identification of Targeted Therapy Opportunities. JAMA Netw Open 2023; 6:e2323336. [PMID: 37459099 DOI: 10.1001/jamanetworkopen.2023.23336] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
IMPORTANCE Precision oncology using comprehensive genomic profiling (CGP) by next-generation sequencing is aimed at companion diagnosis and genomic profiling. The clinical utility of CGP before the standard of care (SOC) is still not resolved, and more evidence is needed. OBJECTIVE To investigate the clinical utility of next-generation CGP (FoundationOne CDx [F1CDx]) in patients with previously untreated metastatic or recurrent solid tumors. DESIGN, Setting, and Participants This multicenter, prospective, observational cohort study enrolled patients with previously untreated advanced solid tumors between May 18, 2021, and February 16, 2022, with follow-up through August 16, 2022. The study was conducted at 6 hospitals in Japan. Eligible patients were aged 20 years or older and had Eastern Cooperative Oncology Group performance status of 0 to 1 with previously untreated metastatic or recurrent cancers in the gastrointestinal or biliary tract; pancreas, lung, breast, uterus, or ovary; and malignant melanoma. EXPOSURE Comprehensive genomic profiling testing before SOC for advanced solid tumors. MAIN OUTCOMES AND MEASURES Proportion of patients with actionable or druggable genomic alterations and molecular-based recommended therapy (MBRT). RESULTS A total of 183 patients met the inclusion criteria and 180 patients (92 men [51.1%]) with a median age of 64 years (range, 23-88 years) subsequently underwent CGP (lung [n = 28], colon/small intestine [n = 27], pancreas [n = 27], breast [n = 25], biliary tract [n = 20], gastric [n = 19], uterus [n = 12], esophagus [n = 10], ovary [n = 6], and skin melanoma [n = 6]). Data from 172 patients were available for end point analyses. Actionable alterations were found in 172 patients (100.0%; 95% CI, 97.9%-100.0%) and druggable alternations were identified in 109 patients (63.4%; 95% CI, 55.7%-70.6%). The molecular tumor board identified MBRT for 105 patients (61.0%; 95% CI, 53.3%-68.4%). Genomic alterations included in the companion diagnostics list of the CGP test were found in 49 patients (28.5%; 95% CI, 21.9%-35.9%) in a tumor-agnostic setting. After a median follow-up of 7.9 months (range, 0.5-13.2 months), 34 patients (19.8%; 95% CI, 14.1%-26.5%) received MBRT. CONCLUSIONS AND RELEVANCE The findings of this study suggest that CGP testing before SOC for patients with advanced solid tumors may be clinically beneficial to guide the subsequent anticancer therapies, including molecularly matched treatments.
Collapse
Affiliation(s)
- Junichi Matsubara
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Kumi Mukai
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Masahiro Yoshioka
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Hidenori Kage
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Katsutoshi Oda
- Department of Clinical Genomics, The University of Tokyo Hospital, Tokyo, Japan
| | - Ryo Kudo
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kei Muro
- Department of Clinical Oncology, Aichi Cancer Center, Nagoya, Japan
| | - Ryuji Hayashi
- Department of Clinical Oncology, Toyama University Hospital, Toyama, Japan
| | - Nahomi Tokudome
- Internal Medicine III, Wakayama Medical University, Wakayama, Japan
| | | | - Manabu Muto
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
12
|
Lazar J, Antal-Szalmas P, Kurucz I, Ferenczi A, Jozsi M, Tornyi I, Muller M, Fekete JT, Lamont J, FitzGerald P, Gall-Debreceni A, Kadas J, Vida A, Tardieu N, Kieffer Y, Jullien A, Guergova-Kuras M, Hempel W, Kovacs A, Kardos T, Bittner N, Csanky E, Szilasi M, Losonczy G, Szondy K, Galffy G, Csada E, Szalontai K, Somfay A, Malka D, Cottu P, Bogos K, Takacs L. Large-Scale Plasma Proteome Epitome Profiling is an Efficient Tool for the Discovery of Cancer Biomarkers. Mol Cell Proteomics 2023; 22:100580. [PMID: 37211046 PMCID: PMC10319867 DOI: 10.1016/j.mcpro.2023.100580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 05/23/2023] Open
Abstract
Current proteomic technologies focus on the quantification of protein levels, while little effort is dedicated to the development of system approaches to simultaneously monitor proteome variability and abundance. Protein variants may display different immunogenic epitopes detectable by monoclonal antibodies. Epitope variability results from alternative splicing, posttranslational modifications, processing, degradation, and complex formation and possesses dynamically changing availability of interacting surface structures that frequently serve as reachable epitopes and often carry different functions. Thus, it is highly likely that the presence of some of the accessible epitopes correlates with function under physiological and pathological conditions. To enable the exploration of the impact of protein variation on the immunogenic epitome first, here, we present a robust and analytically validated PEP technology for characterizing immunogenic epitopes of the plasma. To this end, we prepared mAb libraries directed against the normalized human plasma proteome as a complex natural immunogen. Antibody producing hybridomas were selected and cloned. Monoclonal antibodies react with single epitopes, thus profiling with the libraries is expected to profile many epitopes which we define by the mimotopes, as we present here. Screening blood plasma samples from control subjects (n = 558) and cancer patients (n = 598) for merely 69 native epitopes displayed by 20 abundant plasma proteins resulted in distinct cancer-specific epitope panels that showed high accuracy (AUC 0.826-0.966) and specificity for lung, breast, and colon cancer. Deeper profiling (≈290 epitopes of approximately 100 proteins) showed unexpected granularity of the epitope-level expression data and detected neutral and lung cancer-associated epitopes of individual proteins. Biomarker epitope panels selected from a pool of 21 epitopes of 12 proteins were validated in independent clinical cohorts. The results demonstrate the value of PEP as a rich and thus far unexplored source of protein biomarkers with diagnostic potential.
Collapse
Affiliation(s)
- Jozsef Lazar
- Biosystems International Kft., Debrecen, Hungary; Biosystems Immunolab Zrt., Debrecen, Hungary.
| | - Peter Antal-Szalmas
- Biosystems Immunolab Zrt., Debrecen, Hungary; Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Istvan Kurucz
- Biosystems International Kft., Debrecen, Hungary; Biosystems Immunolab Zrt., Debrecen, Hungary
| | | | - Mihaly Jozsi
- Department of Immunology, ELTE Eötvös Loránd University, Budapest, Hungary; MTA-ELTE Complement Research Group, Eötvös Loránd Research Network (ELKH), Budapest, Hungary
| | - Ilona Tornyi
- Biosystems Immunolab Zrt., Debrecen, Hungary; Department of Human Genetics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | | | - John Lamont
- Randox Laboratories Ltd, Crumlin, United Kingdom
| | | | | | - Janos Kadas
- Biosystems International Kft., Debrecen, Hungary
| | - Andras Vida
- Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | | | | | | | | | | | - Tamas Kardos
- Department of Pulmonology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Nora Bittner
- Department of Pulmonology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Eszter Csanky
- Department of Pulmonology, Miskolc Semmelweis Hospital and University Hospital, Miskolc, Hungary
| | - Maria Szilasi
- Department of Pulmonology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Gyorgy Losonczy
- Department of Pulmonology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Klara Szondy
- Department of Pulmonology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Gabriella Galffy
- Department of Pulmonology, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Edit Csada
- Csongrád County Hospital of Chest Diseases, Deszk, Hungary
| | | | - Attila Somfay
- Department of Pulmonology, Faculty of Medicine, University of Szeged, Deszk, Hungary
| | - David Malka
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Paul Cottu
- Department of Medical Oncology, Institut Curie, Paris, France
| | - Krisztina Bogos
- National Koranyi Institute for Pulmonology, Budapest, Hungary
| | - Laszlo Takacs
- Biosystems International Kft., Debrecen, Hungary; Biosystems Immunolab Zrt., Debrecen, Hungary; Department of Human Genetics, Faculty of Medicine, University of Debrecen, Debrecen, Hungary; Biosystems International SAS, Evry, France.
| |
Collapse
|
13
|
Kunimasa K, Sugimoto N, Kawamura T, Yamasaki T, Honma K, Nagata S, Kukita Y, Fujisawa F, Inoue T, Yamaguchi Y, Kitasaka M, Wakamatsu T, Yamai T, Yamamoto S, Hayashi T, Inoue T, Tamiya M, Imamura F, Nishimura K, Nishino K. Clinical application of comprehensive genomic profiling panel to thoracic malignancies: A single-center retrospective study. Thorac Cancer 2022; 13:2970-2977. [PMID: 36100256 PMCID: PMC9626350 DOI: 10.1111/1759-7714.14643] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND The usefulness of comprehensive genomic profiling (CGP) panels for thoracic malignancies after completion of the standard treatment is unclear. METHODS The results of CGP panels for malignant thoracic diseases performed at our hospital between December 2019 and June 2022 were collected. We examined whether CGP panel results led to new treatment, correlated with the effectiveness of immune checkpoint inhibitors (ICIs), or revealed secondary findings related to hereditary tumors. RESULTS A total of 60 patients were enrolled, of which 52 (86.6%) had lung cancer. In six (10%) patients, the panel results led to treatment with insurance-listed molecular-targeted agents; four patients had EGFR mutations not detected by the real-time polymerase chain reaction assay and two had MET ex.14 skipping mutations. In small-cell lung cancer, the tumor mutation burden was high in 4/6 (66.7%) patients and pembrolizumab was available. Another MET ex.14 skipping mutation was detected in two cases with EGFR-tyrosine kinase inhibitor resistance. ICI efficacy was ≤1 year in patients with STK-11, KEAP1, and NEF2L2 mutations. A BRCA2 mutation with a high probability of germline mutation was detected in one patient. A thymic carcinoma with no detectable oncogenic mutation responded to second-line treatment with Tegafur-Gimeracil-Oteracil Potassium (TS-1) for ≥9 years. CONCLUSIONS CGP panels are useful in thoracic malignancies, especially lung cancer, because they can detect overlooked driver mutations and genetic alterations. We believe that the significance of conducting a CGP panel prior to treatment may also exist, as it may lead to the prediction of ICI treatment efficacy.
Collapse
Affiliation(s)
- Kei Kunimasa
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan,Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Naotoshi Sugimoto
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Department of Medical OncologyOsaka International Cancer InstituteOsakaJapan
| | - Takahisa Kawamura
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan,Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Tomoyuki Yamasaki
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Department of Endocrinology/Metabolism Internal Medicine, Clinical ExaminationOsaka International Cancer InstituteOsakaJapan
| | - Keiichiro Honma
- Department of Diagnostic Pathology and CytologyOsaka International Cancer InstituteOsakaJapan
| | - Shigenori Nagata
- Department of Diagnostic Pathology and CytologyOsaka International Cancer InstituteOsakaJapan
| | - Yoji Kukita
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Laboratory of Genomic PathologyOsaka International Cancer InstituteOsakaJapan
| | - Fumie Fujisawa
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Department of Medical OncologyOsaka International Cancer InstituteOsakaJapan
| | - Tazuko Inoue
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Yuko Yamaguchi
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Mitsuko Kitasaka
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Toru Wakamatsu
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Musculoskeletal Oncology ServiceOsaka International Cancer InstituteOsakaJapan
| | - Takuo Yamai
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Department of Hepatobiliary and Pancreatic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Sachiko Yamamoto
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Department of Gastrointestinal OncologyOsaka International Cancer InstituteOsakaJapan
| | - Takuji Hayashi
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Department of UrologyOsaka International Cancer InstituteOsakaJapan
| | - Takako Inoue
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Motohiro Tamiya
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Fumio Imamura
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan
| | - Kazuo Nishimura
- Department of Genetic OncologyOsaka International Cancer InstituteOsakaJapan,Department of UrologyOsaka International Cancer InstituteOsakaJapan
| | - Kazumi Nishino
- Department of Thoracic OncologyOsaka International Cancer InstituteOsakaJapan
| |
Collapse
|
14
|
Kanai M. Current Clinical Practice of Precision Medicine Using Comprehensive Genomic Profiling Tests in Biliary Tract Cancer in Japan. Curr Oncol 2022; 29:7272-7284. [PMID: 36290850 PMCID: PMC9599999 DOI: 10.3390/curroncol29100573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
With the recent advances of next generation sequencing technologies, comprehensive genomic profiling (CGP) tests, which are designed to measure more than hundreds of cancer-related genes at a time, have now been widely introduced into daily clinical practice. For the patients whose tumor samples are not fit for tissue-based CGP tests, a blood-based CGP test (liquid biopsy) is available as an alternative option. Three CGP tests, "OncoGuide NCC™Oncopanel System (124 genes)", "FoundationOne®CDx (324 genes)", and "Founda-tionOne®CDx Liquid (324 genes)", are now reimbursed by public insurance in 233 hospitals designated for cancer genomic medicine in Japan. In biliary tract cancer, the prevalence of druggable variants is relatively higher compared to other cancer types and the European Society for Medical Oncology recommends routine use of CGP tests for advanced biliary tract cancer to guide treatment options. The latest National Cancer Center Network guideline lists eight druggable markers (NTRK fusion, MSI-H, TMB-H, BRAF V600E, FGFR2 fusions/rearrangement, IDH1 mutations, RET fusion, and HER2 overexpression) and matched therapies. In Japan, matched therapies for four markers (NTRK, MSI-H, TMB-H, and FGFR2) are reimbursed by public insurance (as of September 2022). The progress of genomic profiling technology will contribute to the improvement of the dismal clinical outcomes of this disease in the future.
Collapse
Affiliation(s)
- Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
15
|
Ogiri M, Seishima R, Nakamura K, Aimono E, Matsui S, Shigeta K, Chiyoda T, Tanishima S, Okabayashi K, Nishihara H, Kitagawa Y. Real-world application of next-generation sequencing-based test for surgically resectable colorectal cancer in clinical practice. Future Oncol 2022; 18:2701-2711. [PMID: 35818975 DOI: 10.2217/fon-2022-0122] [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: 11/21/2022] Open
Abstract
Aim: To evaluate the significance of next-generation sequencing-based gene panel testing in surgically resectable colorectal cancer by analyzing real-world data. Materials & methods: A total of 107 colorectal cancer patients who underwent curative surgery were included, and correlations between next-generation sequencing data and clinicopathological findings were evaluated. Results: More combination patterns in gene alteration were identified in advanced-stage tumors than in early-stage tumors. The copy number alteration count was significantly lower in right-sided colon tumors and early-stage tumors. Homologous recombination deficiency was more often identified in advanced-stage tumors, and high homologous recombination deficiency status was useful for identifying high-risk stage II tumors. Conclusion: Homologous recombination deficiency was identified as a useful result of gene panel testing with novel utility in clinical practice.
Collapse
Affiliation(s)
- Masayo Ogiri
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Seishima
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kohei Nakamura
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Eriko Aimono
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Shimpei Matsui
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Kohei Shigeta
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Tatsuyuki Chiyoda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Shigeki Tanishima
- Department of Biomedical Informatics, Kansai Division, Mitsubishi Space Software Co., Ltd., Tokyo, Japan
| | - Koji Okabayashi
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Hiroshi Nishihara
- Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Yuko Kitagawa
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
16
|
HER2 G776S mutation promotes oncogenic potential in colorectal cancer cells when accompanied by loss of APC function. Sci Rep 2022; 12:9213. [PMID: 35654814 PMCID: PMC9163061 DOI: 10.1038/s41598-022-13189-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 05/04/2022] [Indexed: 12/03/2022] Open
Abstract
Clinical cancer genome sequencing detects oncogenic variants that are potential targets for cancer treatment, but it also detects variants of unknown significance. These variants may interact with each other to influence tumor pathophysiology, however, such interactions have not been fully elucidated. Additionally, the effect of target therapy for those variants also unclarified. In this study, we investigated the biological functions of a HER2 mutation (G776S mutation) of unknown pathological significance, which was detected together with APC mutation by cancer genome sequencing of samples from a colorectal cancer (CRC) patient. Transfection of the HER2 G776S mutation alone slightly increased the kinase activity and phosphorylation of HER2 protein, but did not activate HER2 downstream signaling or alter the cell phenotype. On the other hand, the HER2 G776S mutation was shown to have strong oncogenic potential when loss of APC function was accompanied. We revealed that loss of APC function increased Wnt pathway activity but also increased RAS–GTP, which increased ERK phosphorylation triggered by HER2 G776S transfection. In addition, afatinib, a pan-HER tyrosine kinase inhibitor, suppressed tumor growth in xenografts derived from HER2 G776S-transfected CRC cells. These findings suggest that this HER2 mutation in CRC may be a potential therapeutic target.
Collapse
|
17
|
Quy PN, Fukuyama K, Kanai M, Kou T, Kondo T, Yoshioka M, Matsubara J, Sakuma T, Minamiguchi S, Matsumoto S, Muto M. Inter-assay variability of next-generation sequencing-based gene panels. BMC Med Genomics 2022; 15:86. [PMID: 35428255 PMCID: PMC9013031 DOI: 10.1186/s12920-022-01230-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 04/04/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Tumor heterogeneity has been known to cause inter-assay discordance among next-generation sequencing (NGS) results. However, whether preclinical factors such as sample type, sample quality and analytical features of gene panel can affect the concordance between two different assays remains largely unexplored. METHODS Replicate sets of DNA samples extracted from formalin-fixed paraffin-embedded tissues (FFPE) (n = 20) and fresh frozen (FF) tissues (n = 10) were herein analyzed using a tumor-only (TO) and paired tumor-normal (TN) gene panel in laboratories certified by the Clinical Laboratory Improvement Amendment. Reported variants from the TO and TN panels were then compared. Furthermore, additional FFPE samples were sequentially sliced from the same FFPE block and submitted to another TN panel assay. RESULTS Substantial discordance (71.8%) was observed between the results of the two panels despite using identical DNA samples, with the discordance rate being significantly higher for FFPE samples (p < 0.05). Among the 99 variants reported only in the TO panel, 32.3% were consistent with germline variants, which were excluded in the TN panel, while 30.3% had an allele frequency of less than 5%, some of which were highly likely to be artificial calls. The comparison of two independent TN panel assay results from the same FFPE block also showed substantial discordance rate (55.3%). CONCLUSIONS In the context of clinical settings, our comparative analysis revealed that inter-NGS assay discordance commonly occurred due to sample types and the different analytical features of each panel.
Collapse
Affiliation(s)
- Pham Nguyen Quy
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keita Fukuyama
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Real World Data Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Tadayuki Kou
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiro Yoshioka
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junichi Matsubara
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Sakuma
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Sachiko Minamiguchi
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigemi Matsumoto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Department of Real World Data Research and Development, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
18
|
Takeda H, Imoto K, Umemoto K, Doi A, Arai H, Horie Y, Mizukami T, Oguri T, Ogura T, Izawa N, Yamamoto H, Yamano Y, Sunakawa Y. Clinical Utility of Genomic Profiling Tests in Patients with Advanced Gastrointestinal Cancers. Target Oncol 2022; 17:177-185. [PMID: 35366174 DOI: 10.1007/s11523-022-00871-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/02/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Comprehensive analyses of cancer-related genomic alterations are expected to lead to increased availability of targeted therapies. However, in patients with gastrointestinal (GI) cancers, the utility of genomic profiling is unclear because of common non-druggable alterations and rapid disease progression that prevent a sufficient time period to seek targets. OBJECTIVE The aim of this study was to determine the utility of genomic profiling tests in patients with GI cancers. METHODS The subjects of this retrospective study were patients with GI cancers and patients with non-GI cancers who underwent tissue-based genomic profiling at a single institution from April 2017 to October 2020. The profile of gene alterations, frequency of tumor mutational burden-high (≥ 10 Muts/Mb), and accessibility of recommended molecular targeted therapy were compared between patients with GI cancers and patients with non-GI cancers. RESULTS In all, 133 patients with GI cancers and 63 patients with non-GI cancers were included. The genomic profiles of GI cancers showed the highest frequencies of TP53, KRAS, and APC mutations and a significantly lower frequency of PIK3CA mutations than those of non-GI cancers. Tumor mutational burden-high was significantly less prevalent in GI cancers (4% vs 20%, p = 0.008). Twenty-nine patients with GI cancers (40%) and 35 patients with non-GI cancers (56%) were recommended for targeted therapies based on the findings. Among them, seven patients each with GI cancers and non-GI cancers received the recommended therapy on their genomic findings, which showed similar treatment accessibility between the GI and non-GI cancer groups (10% vs 11%, p = 0.791). HER2-targeted and BRAF-targeted therapies were the primary treatments administered to patients with GI cancers. CONCLUSIONS Although their genomic profiles revealed fewer druggable sites, patients with GI cancers accessed targeted therapies similarly to patients with non-GI cancers. The utility of genomic profile testing in patients with GI cancers was highlighted to determine if patients can receive specific treatments, such as HER2-targeted and BRAF-targeted therapies.
Collapse
Affiliation(s)
- Hiroyuki Takeda
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Kiyomi Imoto
- Center for Genomic Medicine, St. Marianna University Hospital, Kawasaki, Japan
| | - Kumiko Umemoto
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Ayako Doi
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hiroyuki Arai
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshiki Horie
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takuro Mizukami
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Tomoyo Oguri
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Takashi Ogura
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Naoki Izawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Hiroyuki Yamamoto
- Department of Bioinformatics, St. Marianna University Graduate School of Medicine, Kawasaki, Japan.,Division of Gastroenterology and Hepatology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yoshihisa Yamano
- Division of Neurology, Department of Internal Medicine, St. Marianna University School of Medicine, Kawasaki, Japan.,Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kawasaki, Japan
| | - Yu Sunakawa
- Department of Clinical Oncology, St. Marianna University School of Medicine, Kawasaki, Japan. .,Center for Genomic Medicine, St. Marianna University Hospital, Kawasaki, Japan.
| |
Collapse
|
19
|
Aoyagi Y, Kano Y, Tohyama K, Matsudera S, Kumaki Y, Takahashi K, Mitsumura T, Harada Y, Sato A, Nakamura H, Sueoka E, Aragane N, Kimura K, Onishi I, Takemoto A, Akahoshi K, Ono H, Ishikawa T, Tokunaga M, Nakagawa T, Oshima N, Nakamura R, Takagi M, Asakage T, Uetake H, Tanabe M, Miyake S, Kinugasa Y, Ikeda S. Clinical utility of comprehensive genomic profiling in Japan: Result of PROFILE-F study. PLoS One 2022; 17:e0266112. [PMID: 35358259 PMCID: PMC8970371 DOI: 10.1371/journal.pone.0266112] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 03/14/2022] [Indexed: 11/18/2022] Open
Abstract
Introduction
Clinical sequencing has provided molecular and therapeutic insights into the field of clinical oncology. However, despite its significance, its clinical utility in Japanese patients remains unknown. Here, we examined the clinical utility of tissue-based clinical sequencing with FoundationOne® CDx and FoundationOne® Heme. Between August 2018 and August 2019, 130 Japanese pretreated patients with advanced solid tumors were tested with FoundationOne® CDx or FoundationOne® Heme.
Results
The median age of 130 patients was 60.5 years (range: 3 to 84 years), and among them, 64 were males and 66 were females. Major cancer types were gastrointestinal cancer (23 cases) and hepatic, biliary, and pancreatic cancer (21 cases). A molecular tumor board had been completed on all 130 cases by October 31, 2019. The median number of gene alterations detected by Foundation testing, excluding variants of unknown significance (VUS) was 4 (ranged 0 to 21) per case. Of the 130 cases, one or more alterations were found in 123 cases (94.6%), and in 114 cases (87.7%), actionable alterations with candidates for therapeutic agents were found. In 29 (22.3%) of them, treatment corresponding to the gene alteration was performed. Regarding secondary findings, 13 cases (10%) had an alteration suspected of a hereditary tumor. Of the 13 cases, only one case received a definite diagnosis of hereditary tumor.
Conclusions
Our study showed that clinical sequencing might be useful for detecting gene alterations in various cancer types and exploring treatment options. However, many issues still need to be improved.
Collapse
Affiliation(s)
- Yasuko Aoyagi
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail: (YA); (SI)
| | - Yoshihito Kano
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Clinical Oncology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kohki Tohyama
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shotaro Matsudera
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
- Department of Specialized Surgeries, Tokyo Medical and Dental University, Tokyo, Japan
- First Department of Surgery, Dokkyo Medical University, Tochigi, Japan
| | - Yuichi Kumaki
- Department of Specialized Surgeries, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kenta Takahashi
- Department of Obstetrics and Gynecology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takahiro Mitsumura
- Department of Respiratory Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yohei Harada
- Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University, Saga, Japan
| | - Akemi Sato
- Department of Transfusion Medicine, Saga University Hospital, Saga, Japan
| | - Hideaki Nakamura
- Department of Transfusion Medicine, Saga University Hospital, Saga, Japan
| | - Eisaburo Sueoka
- Department of Transfusion Medicine, Saga University Hospital, Saga, Japan
| | - Naoko Aragane
- Division of Hematology, Respiratory Medicine and Oncology, Faculty of Medicine, Saga University, Saga, Japan
| | - Koichiro Kimura
- Department of Radiology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Iichiro Onishi
- Department of Pathology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akira Takemoto
- Department of Bioresource Research Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keiichi Akahoshi
- Department of Hepato-Biliary-Pancreatic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroaki Ono
- Department of Hepato-Biliary-Pancreatic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toshiaki Ishikawa
- Department of Specialized Surgeries, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masanori Tokunaga
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Tsuyoshi Nakagawa
- Department of Specialized Surgeries, Tokyo Medical and Dental University, Tokyo, Japan
| | - Noriko Oshima
- Department of Obstetrics and Gynecology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Reiko Nakamura
- Department of Obstetrics and Gynecology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masatoshi Takagi
- Department of Pediatrics, Tokyo Medical and Dental University, Tokyo, Japan
| | - Takahiro Asakage
- Department of Head and Neck Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Hiroyuki Uetake
- Department of Specialized Surgeries, Tokyo Medical and Dental University, Tokyo, Japan
| | - Minoru Tanabe
- Department of Hepato-Biliary-Pancreatic Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Satoshi Miyake
- Department of Clinical Oncology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yusuke Kinugasa
- Department of Gastrointestinal Surgery, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sadakatsu Ikeda
- Department of Precision Cancer Medicine, Center for Innovative Cancer Treatment, Tokyo Medical and Dental University, Tokyo, Japan
- * E-mail: (YA); (SI)
| |
Collapse
|
20
|
Physicians' perceptions of the factors influencing disclosure of secondary findings in tumour genomic profiling in Japan: a qualitative study. Eur J Hum Genet 2022; 30:88-94. [PMID: 34400810 PMCID: PMC8738764 DOI: 10.1038/s41431-021-00944-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/18/2021] [Accepted: 08/05/2021] [Indexed: 01/03/2023] Open
Abstract
Tumour genomic profiling (TGP), conducted in search of therapeutics, sometimes reveals potentially pathogenic germline variants as secondary findings (SFs). Physicians involved in TGP are often specialised in oncology and not in clinical genetics. To better utilise SFs, we explored issues physicians have during disclosure and the potential for collaborations with clinical genetics professionals. Semi-structured interviews were conducted with 14 physicians who had experience in handling outpatient TGP at designated core hospitals for cancer genomic medicine in Japan. The data were analysed thematically. The difficulties physicians experienced during informed consent (IC) included educating patients about SFs, providing detailed information on SFs, and explaining the impact of SFs on patients' family members. When SFs were detected, physicians had reservations regarding the relevance of the disclosure criteria. Confirmatory germline tests were performed using peripheral blood when tumour-only tests detected suspected SFs. Some physicians had reservations about the necessity of confirmatory tests when they did not affect the patients' treatment options. To encourage patients to receive confirmatory tests, improvements are necessary in the healthcare system, such as insurance reimbursements, education for physicians so that they can provide a better explanation to their patients, and genetic literacy of physicians and patients. The physicians offered insights into the challenges they experienced related to IC, disclosure of SFs, and expectations for active collaborations with clinical genetics professionals. Wider healthcare insurance coverage and better genetic literacy of the population may lead to more patients taking confirmatory tests when SFs are suspected.
Collapse
|
21
|
Fukui T, Sakai K, Sasaki J, Kakegawa MI, Igawa S, Mitsufuji H, Takeda M, Takahama T, Nakagawa K, Nishio K, Naoki K. Implementation of clinical sequencing for molecular profiling in patients with advanced cancer. Cancer Biomark 2021; 31:119-126. [PMID: 33896820 DOI: 10.3233/cbm-200781] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND The advancement of cancer genomics has allowed for multiplex gene assays using next-generation sequencing (NGS) to be practically implemented, however, a clinical practice system remains to be established. OBJECTIVE We evaluated the feasibility of clinical sequencing using NGS-based multiplex gene assays between cooperating medical institutions in patients with advanced cancers. METHODS In this observational study, DNA and RNA samples prepared from existing tumor tissues were subjected to comprehensive genomic profiling using targeted sequencing. RESULTS From January 2017 to March 2019, 36 samples from 33 patients were assessed. Of all patients, 27 (82%) had lung cancer, with the median age of 50 years (range 38-83). Multiplex gene panel tests were successfully carried out on 35/36 (97%) samples. Potentially actionable gene alterations were identified in 10/30 (33%) samples (3 HER2, 2 KRAS, 2 ALK, 1 PIK3CA, 1 RET, and 1 CDKN2A). In the 6 samples examined for resistant mechanisms, ALK I1171N mutation and MET copy number gain were detected in 2 patients with ALK rearrangement-positive lung cancer. CONCLUSIONS Clinical sequencing using NGS-based multiplex gene assays between collaborating domestic medical institutions was feasible, with a success rate of > 97%. Overall, clinical sequencing benefits therapeutic decision-making in patients with advanced cancer.
Collapse
Affiliation(s)
- Tomoya Fukui
- Department of Respiratory Medicine, Kitasato University School of Medicine, Kanagawa, Japan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Jiichiro Sasaki
- Research and Development Center for New Medical Frontiers, Kitasato University School of Medicine, Kanagawa, Japan
| | | | - Satoshi Igawa
- Department of Respiratory Medicine, Kitasato University School of Medicine, Kanagawa, Japan
| | - Hisashi Mitsufuji
- Fundamental Nursing, Kitasato University School of Nursing, Kanagawa, Japan
| | - Masayuki Takeda
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Takayuki Takahama
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Katsuhiko Naoki
- Department of Respiratory Medicine, Kitasato University School of Medicine, Kanagawa, Japan
| |
Collapse
|
22
|
Yam C, Ma BBY, Yap TA. Global Implementation of Precision Oncology. JCO Precis Oncol 2021; 5:PO.21.00001. [PMID: 34250385 PMCID: PMC8232074 DOI: 10.1200/po.21.00001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 01/17/2021] [Indexed: 01/12/2023] Open
Affiliation(s)
- Clinton Yam
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Brigette B Y Ma
- Department of Clinical Oncology, Phase I Clinical Trial Center, Kingboard Precision Oncology Program, The Chinese University of Hong Kong, Hong Kong, China
| | - Timothy A Yap
- Department of Investigational Cancer Therapeutics (Phase I Clinical Trials Program), The University of Texas MD Anderson Cancer Center, Houston, TX.,Khalifa Institute of Personalized Cancer Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX.,Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX.,The Institute for Applied Cancer Science, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
23
|
Kikuchi J, Ohhara Y, Takada K, Tanabe H, Hatanaka K, Amano T, C Hatanaka K, Hatanaka Y, Mitamura T, Kato M, Shibata Y, Yabe I, Endoh A, Komatsu Y, Matsuno Y, Sugiyama M, Manabe A, Sakurai A, Takahashi M, Naruse H, Torimoto Y, Dosaka-Akita H, Kinoshita I. Clinical significance of comprehensive genomic profiling tests covered by public insurance in patients with advanced solid cancers in Hokkaido, Japan. Jpn J Clin Oncol 2021; 51:753-761. [PMID: 33532831 DOI: 10.1093/jjco/hyaa277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 12/30/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Comprehensive cancer genomic profiling has been used recently for patients with advanced solid cancers. Two cancer genomic profiling tests for patients with no standard treatment are covered by Japanese public health insurance since June 2019. METHODS We prospectively analyzed data of 189 patients with solid cancers who underwent either of the two-cancer genomic profiling tests at Hokkaido University Hospital and its liaison hospitals and whose results were discussed in molecular tumor board at Hokkaido University Hospital between August 2019 and July 2020. RESULTS All 189 patients had appropriate results. Actionable gene alterations were identified in 93 patients (49%). Frequent mutations included PIK3CA (12%) mutation, BRCA1/2 alteration (7%), ERBB2 amplification (6%) and tumor mutation burden-High (4%). The median turnaround time from sample shipping to acquisition by the expert panel was 26 days. Although 115 patients (61%) were provided with information for genotype-matched therapies, only 21 (11%) received them. Notably, four of eight patients below the age of 20 years were provided information for genotype-matched therapies, and three received them. Their response rates and disease control rates were 29% and 67%, respectively. Most patients who did not undergo the genotype-matched therapies were provided information for only investigational drugs in phases I and II at distant clinical trial sites in central Japan. Twenty-six patients were informed of suspected germline findings, while 11 patients (42%) received genetic counseling. CONCLUSIONS The publicly reimbursed cancer genomic profilings may lead to the modest but favorable therapeutic efficacy of genotype-matched therapy for solid cancer patients with no standard therapy. However, poor access to genotype-matched therapy needs to be resolved.
Collapse
Affiliation(s)
- Junko Kikuchi
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Respiratory Medicine, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| | - Yoshihito Ohhara
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Japan
| | - Kohichi Takada
- Department of Medical Oncology, School of Medicine, Sapporo Medical University, Sapporo, Japan
| | - Hiroki Tanabe
- Genetic Oncology Department, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Kazuteru Hatanaka
- Department of Gastroenterology, Hakodate Municipal Hospital, Hakodate, Japan
| | - Toraji Amano
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Japan
| | - Kanako C Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan.,Clinical Biobank, Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, Japan
| | - Yutaka Hatanaka
- Research Division of Genome Companion Diagnostics, Hokkaido University Hospital, Sapporo, Japan.,Clinical Biobank, Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, Japan
| | - Takashi Mitamura
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Momoko Kato
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Yuka Shibata
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Yabe
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Akira Endoh
- Division of Medical Information Planning, Hokkaido University Hospital, Sapporo, Japan
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital, Cancer Center, Sapporo, Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Minako Sugiyama
- Department of Pediatrics, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Atsushi Manabe
- Department of Pediatrics, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Sakurai
- Department of Medical Genetics and Genomics, Sapporo Medical University, Sapporo, Japan
| | - Masato Takahashi
- Department of Cancer Genome Medical Center, NHO Hokkaido Cancer Center, Sapporo, Japan
| | - Hirohito Naruse
- Department of Gastroenterology, Hakodate Municipal Hospital, Hakodate, Japan
| | - Yoshihiro Torimoto
- Genetic Oncology Department, Asahikawa Medical University Hospital, Asahikawa, Japan
| | - Hirotoshi Dosaka-Akita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Kinoshita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Hospital, Sapporo, Japan
| |
Collapse
|
24
|
Takeda M, Takahama T, Sakai K, Shimizu S, Watanabe S, Kawakami H, Tanaka K, Sato C, Hayashi H, Nonagase Y, Yonesaka K, Takegawa N, Okuno T, Yoshida T, Fumita S, Suzuki S, Haratani K, Saigoh K, Ito A, Mitsudomi T, Handa H, Fukuoka K, Nakagawa K, Nishio K. Clinical Application of the FoundationOne CDx Assay to Therapeutic Decision-Making for Patients with Advanced Solid Tumors. Oncologist 2021; 26:e588-e596. [PMID: 33325566 PMCID: PMC8018334 DOI: 10.1002/onco.13639] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 12/04/2020] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Implementation of personalized medicine requires the accessibility of tumor molecular profiling in order to allow prioritization of appropriate targeted therapies for individual patients. Our aim was to study the role of comprehensive genomic profiling assays that may inform treatment recommendations for patients with solid tumors. MATERIALS AND METHODS We performed a prospective study to evaluate the feasibility of application of the FoundationOne CDx panel-which detects substitutions, insertions and deletions, and copy number alterations in 324 genes, select gene rearrangements, and genomic signatures including microsatellite instability and tumor mutation burden (TMB)-to patients with advanced or recurrent solid tumors before its approval in Japan. RESULTS A total of 181 samples were processed for genomic testing between September 2018 and June 2019, with data being successfully obtained for 175 of these samples, yielding a success rate of 96.7%. The median turnaround time was 41 days (range, 21-126 days). The most common known or likely pathogenic variants were TP53 mutations (n = 113), PIK3CA mutations (n = 33), APC mutations (n = 32), and KRAS mutations (n = 29). Among the 153 patients assessed for TMB, the median TMB was 4 mutations/Mb, and tumors with a high TMB (≥10 mutations/Mb) were more prevalent for lung cancer (11/32) than for other solid tumor types (9/121, Fisher's exact test p < .01). No clear trend toward increased efficacy for immune checkpoint inhibitor (ICI) monotherapy or ICI combination chemotherapy in patients with a high programmed cell death-ligand 1 tumor proportion score or a high TMB was apparent. Among the 174 patients found to harbor known or likely pathogenic actionable alterations, 24 individuals (14%) received matched targeted therapy. CONCLUSION The FoundationOne CDx assay was performed with formalin-fixed, paraffin-embedded tumor specimens with a success rate of >95%. Such testing may inform the matching of patients with cancer with investigational or approved targeted drugs. IMPLICATIONS FOR PRACTICE This prospective cohort study was initiated to investigate the feasibility and utility of clinical application of FoundationOne CDx. A total of 181 samples were processed for genomic testing between September 2018 and June 2019, with data being successfully obtained for 175 of these samples, yielding a success rate of 96.7%, and 24 individuals (14%) received matched targeted therapy.
Collapse
Affiliation(s)
- Masayuki Takeda
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Takayuki Takahama
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Kazuko Sakai
- Department of Genome Biology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Shigeki Shimizu
- Department of Pathology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Satomi Watanabe
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Hisato Kawakami
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Kaoru Tanaka
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Chihiro Sato
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Hidetoshi Hayashi
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Yoshikane Nonagase
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Kimio Yonesaka
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Naoki Takegawa
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Tatsuya Okuno
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Takeshi Yoshida
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Soichi Fumita
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Shinichiro Suzuki
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Koji Haratani
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Kazumasa Saigoh
- Clinical Genetics, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Akihiko Ito
- Department of Pathology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Tetsuya Mitsudomi
- Division of Thoracic Surgery, Department of Surgery, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Hisashi Handa
- Research Institute for Informatics, Kindai UniversityHigashi‐OsakaOsakaJapan
- Research Institute for Science and Technology, Kindai UniversityHigashi‐OsakaOsakaJapan
- Faculty of Science and Engineering, Kindai UniversityHigashi‐OsakaOsakaJapan
| | - Kazuya Fukuoka
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Kazuhiko Nakagawa
- Department of Medical Oncology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of MedicineOsaka‐SayamaOsakaJapan
| |
Collapse
|
25
|
Nishimura S, Sugimoto A, Kushiyama S, Togano S, Kuroda K, Yamamoto Y, Yamauchi M, Sumi T, Kaneda H, Kawaguchi T, Kato M, Tagami M, Oebisu N, Hoshi M, Kimura K, Kubo S, Muguruma K, Takashima T, Ohira M, Yashiro M. Clinical benefit for clinical sequencing using cancer panel testing. PLoS One 2021; 16:e0247090. [PMID: 33635883 PMCID: PMC7909652 DOI: 10.1371/journal.pone.0247090] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/25/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Clinical sequencing using a panel of genes has recently been applied worldwide for patients with refractory solid tumors, but the significance of clinical sequencing using gene panel testing remains uncertain. Here we sought to clarify the feasibility and utility of clinical sequencing in the treatment of refractory tumors at our hospital. METHODS A total of 39 patients with advanced solid tumors treated at our hospital between 2018 and 2020 were enrolled in the clinical sequencing. Among them, we identified 36 patients whose tissue samples were of suitable quality for clinical sequencing, and we analyzed the genomic profiles of these tumors. RESULTS Pathogenic alterations were detected in 28 (78%) of the 36 patients. The most common mutation was TP53 (55%), followed by KRAS (22%), and the highest frequency of gene amplification was ERBB2 (17%). Nine of the 36 patients were identified as candidates for novel molecular-targeted therapy based on their actionable gene alterations, but only one case ended up receiving novel targeted therapy following the genetic tests. CONCLUSIONS Our current results suggested that clinical sequencing might be useful for the detection of pathogenic alterations and the management of additional cancer treatment. However, molecular target based on actionable genomic alteration does not always bridge to subsequent therapy due to clinical deterioration, refusal for unapproved drug, and complexity of clinical trial access. Both improved optimal timing of clinical sequencing and a consensus about its off-label use might help patients receive greater benefit from clinical sequencing.
Collapse
Affiliation(s)
- Sadaaki Nishimura
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
- Department of Hepato-Biliary-Pancreatic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Sugimoto
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shuhei Kushiyama
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shingo Togano
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kenji Kuroda
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Yurie Yamamoto
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Makoto Yamauchi
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Toshiyuki Sumi
- Department of Obstetrics and Gynecology, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Genomic Center, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Hiroyasu Kaneda
- Department of Clinical Oncology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tomoya Kawaguchi
- Department of Respiratory Medicine, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Minoru Kato
- Department of Urology, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Mizuki Tagami
- Department of Ophthalmology and Visual Sciences, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Naoto Oebisu
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Manabu Hoshi
- Department of Orthopedic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kenjiro Kimura
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Department of Hepato-Biliary-Pancreatic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Shoji Kubo
- Department of Hepato-Biliary-Pancreatic Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Kazuya Muguruma
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Tsutomu Takashima
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masaichi Ohira
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Department of Breast and Endocrine Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
| | - Masakazu Yashiro
- Department of Gastroenterological Surgery, Osaka City University Graduate School of Medicine, Osaka, Japan
- Molecular Oncology and Therapeutics, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Center for Translational Research, Osaka City University Graduate School of Medicine, Osaka, Japan
- Cancer Genomic Center, Osaka City University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
26
|
Mukai Y, Ueno H. Establishment and implementation of Cancer Genomic Medicine in Japan. Cancer Sci 2021; 112:970-977. [PMID: 33289217 PMCID: PMC7935799 DOI: 10.1111/cas.14754] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 11/23/2020] [Accepted: 11/27/2020] [Indexed: 12/27/2022] Open
Abstract
Approximately 1 in 2 Japanese people are estimated to be diagnosed with cancer during their lifetime. Cancer still remains the leading cause of death in Japan, therefore the government of Japan has decided to develop a better cancer control policy and launched the Cancer Genomic Medicine (CGM) program. The Ministry of Health, Labour, and Welfare (MHLW) held a consortium at their headquarters with leading academic authorities and the representatives of related organizations to discuss ways to advance CGM in Japan. Based on the report of the consortium, the CGM system under the national health insurance system has gradually been realized. Eleven hospitals were designated in February 2018 as core hospitals for CGM; subsequently, the MHLW built the Center for Cancer Genomics and Advanced Therapeutics (C‐CAT) as an institution to aggregate and manage genomic and clinical information on cancer patients, and support appropriate secondary use of the aggregated information to develop research aimed at medical innovation. As the first step in Japan's CGM in routine practice, in June 2019 the MHLW started reimbursement of 2 types of tumor profiling tests for advanced solid cancer patients using the national insurance system. Japan's CGM has swiftly been spreading nationwide with the collaboration of 167 hospitals and patients. The health and research authorities are expected to embody personalized cancer medicine and promote CGM utilizing state‐of‐the‐art technologies.
Collapse
Affiliation(s)
- Yosuke Mukai
- Department of Surgery, Osaka International Cancer Institute, Osaka, Japan.,Department of Gastroenterological Surgery, Graduate School of Medicine, Osaka University, Osaka, Japan.,Cancer and Disease Control Division, Health Service Bureau, Government of Japan Ministry of Health, Labour, and Welfare, Tokyo, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
27
|
Jha BS, Farnoodian M, Bharti K. Regulatory considerations for developing a phase I investigational new drug application for autologous induced pluripotent stem cells-based therapy product. Stem Cells Transl Med 2021; 10:198-208. [PMID: 32946199 PMCID: PMC7848308 DOI: 10.1002/sctm.20-0242] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2020] [Revised: 08/04/2020] [Accepted: 08/17/2020] [Indexed: 12/11/2022] Open
Abstract
Induced pluripotent stem cells (iPSC)-based therapies have been hailed as the future of regenerative medicine because of their potential to provide treatment options for most degenerative diseases. A key promise of iPSC-based therapies is the possibility of an autologous transplant that may engraft better in the longer-term due to its compatibility with the patient's immune system. Despite over a decade of research, clinical translation of autologous iPSC-based therapies has been slow-partly due to a lacking pre-defined regulatory path. Here, we outline regulatory considerations for developing an autologous iPSC-based product and challenges associated with the clinical manufacturing of autologous iPSCs and their derivatives. These challenges include donor tissue source, reprogramming methods, heterogeneity of differentiated cells, controls for the manufacturing process, and preclinical considerations. A robust manufacturing process with appropriate quality controls and well-informed, prospectively designed preclinical studies provide a path toward successful approval of autologous iPSC-based therapies.
Collapse
Affiliation(s)
- Balendu Shekhar Jha
- Center for Cell Engineering, Department of Transfusion MedicineClinical Center, National Institutes of HealthBethesdaMarylandUSA
| | - Mitra Farnoodian
- Ocular and Stem Cell Translational Research Section, Ophthalmic Genetics and Visual Function BranchNational Eye Institute, National Institutes of HealthBethesdaMarylandUSA
| | - Kapil Bharti
- Ocular and Stem Cell Translational Research Section, Ophthalmic Genetics and Visual Function BranchNational Eye Institute, National Institutes of HealthBethesdaMarylandUSA
| |
Collapse
|
28
|
The initial assessment of expert panel performance in core hospitals for cancer genomic medicine in Japan. Int J Clin Oncol 2021; 26:443-449. [PMID: 33385275 PMCID: PMC7895780 DOI: 10.1007/s10147-020-01844-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/09/2020] [Indexed: 10/27/2022]
Abstract
BACKGROUND Since June 2019, cancer genomic profiling (CGP) tests have been reimbursed by the National Health Insurance system in Japan, with restrictions for government-designated hospitals with a molecular tumor board composed of multidisciplinary specialists, known as an expert panel (EP). The standardization of EPs is a critical challenge for implementing precision oncology in the clinical setting. METHODS Data on consecutive cases who underwent the CGP tests at 11 core hospitals between June 2019 and January 2020 were collected. We evaluated the proportions of cases that received genomically matched treatments, including investigational new drugs (INDs) based on CGP results, and/or for which genetic counseling was recommended. Two simulated cases were annotated by each EP. The annotated reports were then centrally assessed. RESULTS Each EP mainly discussed the applicability to genomically matched treatments and the necessity of performing genetic counseling. A pre-review of the report by key members in each EP reportedly made the EP conference more interactive and efficient, and thereby saved time. A total of 747 cases underwent CGP tests, 28 cases (3.7%) received genomically matched treatment, and 17 cases (2.3%) were referred for genetic counseling. Annotated reports for the simulated cases varied across the EPs, particularly the number of recommended IND trials, which seemed to be associated with the actual number of participants in IND trials. CONCLUSIONS This investigation provides reference data for the application of precision oncology in a clinical setting. Further investigations on the standardization of clinical annotations are warranted.
Collapse
|
29
|
Kawaji H, Kubo M, Yamashita N, Yamamoto H, Kai M, Kajihara A, Yamada M, Kurata K, Kaneshiro K, Harada Y, Hayashi S, Shimazaki A, Mori H, Akiyoshi S, Oki E, Oda Y, Baba E, Mori M, Nakamura M. Comprehensive molecular profiling broadens treatment options for breast cancer patients. Cancer Med 2020; 10:529-539. [PMID: 33274848 PMCID: PMC7877356 DOI: 10.1002/cam4.3619] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/12/2020] [Accepted: 10/31/2020] [Indexed: 12/30/2022] Open
Abstract
Precision oncology with next generation sequencing (NGS) using tumor tissue with or without blood has begun in Japan. Tumor molecular profiling tests are available, including the OncoGuide™ NCC Oncopanel System and FoundationOne® CDx (F1CDx). Our purpose was to identify potentially actionable genetic alterations in breast cancer with this comprehensive tumor profiling test. We enrolled 115 patients with pathologically diagnosed advanced or metastatic breast cancer. Comprehensive tumor genomic profiling, microsatellite instability, and tumor mutational burden (TMB) were determined using F1CDx. Testing was successful in 109/115 cases (94.8%). Clinically actionable alterations were identified in 76% of advanced breast cancer patients. The most frequent short variants were in TP53 (48.6%), PIK3CA (38.5%), GATA3 (11.0%), PTEN (11.0%), and BRCA1 (10.1%), and structural variants were in ERBB2 (24.8%), MYC (21.1%), RAD21 (21.1%), CCND1 (11.9%), FGF19 (10.1%), and PTEN (10.1%). Regarding human epidermal growth factor receptor (HER)2 status, 106/109 samples (97.2%) were concordant between F1CDx and HER2 testing with immunohistochemistry/fluorescence in situ hybridization. However, ERBB2 amplification was newly detected in four samples and ERBB2 mutations were detected in five HER2‐negative breast cancer samples. Oncogenic BRCA mutations were found in three samples with F1CDx among 27 germline testing‐negative samples. The mean TMB in all samples was 6.28 mut/Mb and tended to be higher in luminal B and triple‐negative breast cancer (mean = 8.1 and 5.9 mut/Mb, respectively) compared with other subtypes. In conclusion, we established a system for precision oncology and obtained preliminary data with NGS as the first step. The information in this clinical sequencing panel will help guide the development of new treatments for breast cancer patients.
Collapse
Affiliation(s)
- Hitomi Kawaji
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Makoto Kubo
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Nami Yamashita
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hidetaka Yamamoto
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaya Kai
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsuko Kajihara
- Foundation Medicine Business Department, Foundation Medicine Unit, Chugai Pharmaceutical Co., Ltd., Tokyo, Japan
| | - Mai Yamada
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kanako Kurata
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kazuhisa Kaneshiro
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yurina Harada
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Saori Hayashi
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Akiko Shimazaki
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hitomi Mori
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Sayuri Akiyoshi
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eiji Oki
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masaki Mori
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masafumi Nakamura
- Department of Surgery and Oncology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
30
|
Naito Y, Aburatani H, Amano T, Baba E, Furukawa T, Hayashida T, Hiyama E, Ikeda S, Kanai M, Kato M, Kinoshita I, Kiyota N, Kohno T, Kohsaka S, Komine K, Matsumura I, Miura Y, Nakamura Y, Natsume A, Nishio K, Oda K, Oda N, Okita N, Oseto K, Sunami K, Takahashi H, Takeda M, Tashiro S, Toyooka S, Ueno H, Yachida S, Yoshino T, Tsuchihara K. Clinical practice guidance for next-generation sequencing in cancer diagnosis and treatment (edition 2.1). Int J Clin Oncol 2020; 26:233-283. [PMID: 33249514 PMCID: PMC7819967 DOI: 10.1007/s10147-020-01831-6] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Accepted: 10/30/2020] [Indexed: 12/20/2022]
Abstract
Background To promote precision oncology in clinical practice, the Japanese Society of Medical Oncology, the Japanese Society of Clinical Oncology, and the Japanese Cancer Association, jointly published “Clinical practice guidance for next-generation sequencing in cancer diagnosis and treatment” in 2017. Since new information on cancer genomic medicine has emerged since the 1st edition of the guidance was released, including reimbursement for NGS-based multiplex gene panel tests in 2019, the guidance revision was made. Methods A working group was organized with 33 researchers from cancer genomic medicine designated core hospitals and other academic institutions. For an impartial evaluation of the draft version, eight committee members from each society conducted an external evaluation. Public comments were also made on the draft. The finalized Japanese version was published on the websites of the three societies in March 2020. Results The revised edition consists of two parts: an explanation of the cancer genomic profiling test (General Discussion) and clinical questions (CQs) that are of concern in clinical practice. Particularly, patient selection should be based on the expectation that the patient's post-test general condition and organ function will be able to tolerate drug therapy, and the optimal timing of test should be considered in consideration of subsequent treatment plans, not limited to treatment lines. Conclusion We expect that the revised version will be used by healthcare professionals and will also need to be continually reviewed in line with future developments in cancer genome medicine.
Collapse
Affiliation(s)
- Yoichi Naito
- Department of General Internal Medicine/Breast and Medical Oncology/Experimental Therapeutics, National Cancer Center Hospital East, Kashiwa, Japan
| | - Hiroyuki Aburatani
- Genome Science Division, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo, Japan
| | - Toraji Amano
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, Japan
| | - Eishi Baba
- Department of Oncology and Social Medicine, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Toru Furukawa
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsu Hayashida
- Department of Surgery, Keio University School of Medicine, Tokyo, Japan
| | - Eiso Hiyama
- Natural Science Center for Basic Research and Development, Hiroshima University, Hiroshima, Japan
| | - Sadakatsu Ikeda
- Cancer Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masashi Kanai
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Motohiro Kato
- Children's Cancer Center, National Center for Child Health and Development, Tokyo, Japan
| | - Ichiro Kinoshita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan
| | - Naomi Kiyota
- Kobe University Hospital Cancer Center, Kobe, Japan
| | - Takashi Kohno
- Division of Genome Biology, National Cancer Center Research Institute, Tokyo, Japan
| | - Shinji Kohsaka
- Division of Cellular Signaling, National Cancer Center Research Institute, Tokyo, Japan
| | - Keigo Komine
- Department of Medical Oncology, Tohoku University Hospital, Sendai, Japan
| | - Itaru Matsumura
- Department of Hematology and Rheumatology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Yuji Miura
- Department of Medical Oncology, Toranomon Hospital, Tokyo, Japan
| | - Yoshiaki Nakamura
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Atsushi Natsume
- Department of Neurosurgery, Nagoya University, Nagoya, Japan
| | - Kazuto Nishio
- Department of Genome Biology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Katsutoshi Oda
- Department of Obstetrics and Gynecology, Faculty of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoyuki Oda
- Section of Information Technology Support, Center for Cancer Genomics and Advanced Therapeutics, National Cancer Center, Tokyo, Japan
| | - Natsuko Okita
- Clinical Research Support Office, National Cancer Center Hospital, Tokyo, Japan
| | - Kumiko Oseto
- Department of Clinical Genomics, The University of Tokyo, Tokyo, Japan
- Konica Minolta Precision Medicine Japan, Inc., Tokyo, Japan
| | - Kuniko Sunami
- Department of Laboratory Medicine, National Cancer Center Hospital, Tokyo, Japan
| | - Hideaki Takahashi
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Masayuki Takeda
- Medical Oncology, Kindai University Faculty of Medicine, Osakasayama, Japan
| | - Shimon Tashiro
- Department of Sociology, Graduate School of Arts and Letters, Tohoku University, Sendai, Japan
| | - Shinichi Toyooka
- Department of General Thoracic Surgery and Breast and Endocrine Surgery, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Hideki Ueno
- Department of Hepatobiliary and Pancreatic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Takayuki Yoshino
- Department of Gastroenterology and Gastrointestinal Oncology, National Cancer Center Hospital East, Kashiwa, Japan
| | - Katsuya Tsuchihara
- Division of Translational Informatics, Exploratory Oncology Research and Clinical Trial Center, National Cancer Center, 6-5-1 Kashiwanoha, Kashiwa, Chiba, 277-8577, Japan.
| |
Collapse
|
31
|
Kondo T, Matsubara J, Quy PN, Fukuyama K, Nomura M, Funakoshi T, Doi K, Sakamori Y, Yoshioka M, Yokoyama A, Tamaoki M, Kou T, Hirohashi K, Yamada A, Yamamoto Y, Minamiguchi S, Nishigaki M, Yamada T, Kanai M, Matsumoto S, Muto M. Comprehensive genomic profiling for patients with chemotherapy-naïve advanced cancer. Cancer Sci 2020; 112:296-304. [PMID: 33007138 PMCID: PMC7780032 DOI: 10.1111/cas.14674] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/17/2020] [Accepted: 09/26/2020] [Indexed: 12/30/2022] Open
Abstract
Comprehensive genomic profiling (CGP) testing by next‐generation sequencing has been introduced into clinical practice as part of precision cancer medicine to select effective targeted therapies. However, whether CGP testing at the time of first‐line chemotherapy could be clinically useful is not clear. We conducted this single‐center, prospective, observational study to investigate the feasibility of CGP testing for chemotherapy‐naïve patients with stage III/IV gastrointestinal cancer, rare cancer, and cancer of unknown primary, using the FoundationOne® companion diagnostic (F1CDx) assay. The primary outcome was the detection rate of at least one actionable/druggable cancer genomic alteration. Actionable/druggable cancer genomic alterations were determined by the F1CDx report. An institutional molecular tumor board determined the molecular‐based recommended therapies. A total of 197 patients were enrolled from October 2018 to June 2019. CGP success rate was 76.6% (151 of 197 patients), and median turnaround time was 19 days (range: 10‐329 days). Actionable and druggable cancer genomic alterations were reported in 145 (73.6%) and 124 (62.9%) patients, respectively. The highest detection rate of druggable genomic alterations in gastrointestinal cancers was 80% in colorectal cancer (48 of 60 patients). Molecular‐based recommended therapies were determined in 46 patients (23.4%). CGP testing would be a useful tool for the identification of a potentially effective first‐line chemotherapy.
Collapse
Affiliation(s)
- Tomohiro Kondo
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Junichi Matsubara
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Pham Nguyen Quy
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Keita Fukuyama
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Motoo Nomura
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Taro Funakoshi
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Keitaro Doi
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Yuichi Sakamori
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Masahiro Yoshioka
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Akira Yokoyama
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Masashi Tamaoki
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Tadayuki Kou
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Kenshiro Hirohashi
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Atsushi Yamada
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Yoshihiro Yamamoto
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | | | | | - Takahiro Yamada
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Masashi Kanai
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Shigemi Matsumoto
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| | - Manabu Muto
- Department of Clinical Oncology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
32
|
Prieto-Potin I, Carvajal N, Plaza-Sánchez J, Manso R, Aúz-Alexandre CL, Chamizo C, Zazo S, López-Sánchez A, Rodríguez-Pinilla SM, Camacho L, Longarón R, Bellosillo B, Somoza R, Hernández-Losa J, Fernández-Soria VM, Ramos-Ruiz R, Cristóbal I, García-Foncillas J, Rojo F. Validation and clinical application of a targeted next-generation sequencing gene panel for solid and hematologic malignancies. PeerJ 2020; 8:e10069. [PMID: 33083132 PMCID: PMC7546223 DOI: 10.7717/peerj.10069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/09/2020] [Indexed: 01/29/2023] Open
Abstract
Background Next-generation sequencing (NGS) is a high-throughput technology that has become widely integrated in molecular diagnostics laboratories. Among the large diversity of NGS-based panels, the Trusight Tumor 26 (TsT26) enables the detection of low-frequency variants across 26 genes using the MiSeq platform. Methods We describe the inter-laboratory validation and subsequent clinical application of the panel in 399 patients presenting a range of tumor types, including gastrointestinal (GI, 29%), hematologic (18%), lung (13%), gynecological and breast (8% each), among others. Results The panel is highly accurate with a test sensitivity of 92%, and demonstrated high specificity and positive predictive values (95% and 96%, respectively). Sequencing testing was successful in two-thirds of patients, while the remaining third failed due to unsuccessful quality-control filtering. Most detected variants were observed in the TP53 (28%), KRAS (16%), APC (10%) and PIK3CA (8%) genes. Overall, 372 variants were identified, primarily distributed as missense (81%), stop gain (9%) and frameshift (7%) altered sequences and mostly reported as pathogenic (78%) and variants of uncertain significance (19%). Only 14% of patients received targeted treatment based on the variant determined by the panel. The variants most frequently observed in GI and lung tumors were: KRAS c.35G > A (p.G12D), c.35G > T (p.G12V) and c.34G > T (p.G12C). Conclusions Prior panel validation allowed its use in the laboratory daily practice by providing several relevant and potentially targetable variants across multiple tumors. However, this study is limited by high sample inadequacy rate, raising doubts as to continuity in the clinical setting.
Collapse
Affiliation(s)
- Iván Prieto-Potin
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Nerea Carvajal
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Jenifer Plaza-Sánchez
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Rebeca Manso
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Carmen Laura Aúz-Alexandre
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Cristina Chamizo
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Sandra Zazo
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Almudena López-Sánchez
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | | | - Laura Camacho
- Department of Pathology, Hospital Del Mar Medical Research Institute, Barcelona, Spain
| | - Raquel Longarón
- Department of Pathology, Hospital Del Mar Medical Research Institute, Barcelona, Spain
| | - Beatriz Bellosillo
- Department of Pathology, Hospital Del Mar Medical Research Institute, Barcelona, Spain
| | - Rosa Somoza
- Department of Pathology, Vall d'Hebron University Hospital, Barcelona, Spain
| | | | | | | | - Ion Cristóbal
- Translational Oncology Division, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Jesús García-Foncillas
- Translational Oncology Division, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| | - Federico Rojo
- Department of Pathology, CIBERONC, UAM, Fundación Jiménez Díaz University Hospital Health Research Institute, Madrid, Spain
| |
Collapse
|
33
|
Hayashi H, Tanishima S, Fujii K, Mori R, Okada C, Yanagita E, Shibata Y, Matsuoka R, Amano T, Yamada T, Yabe I, Kinoshita I, Komatsu Y, Dosaka-Akita H, Nishihara H. Clinical impact of a cancer genomic profiling test using an in-house comprehensive targeted sequencing system. Cancer Sci 2020; 111:3926-3937. [PMID: 32772458 PMCID: PMC7540994 DOI: 10.1111/cas.14608] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 07/14/2020] [Accepted: 08/02/2020] [Indexed: 12/11/2022] Open
Abstract
Precision medicine is a promising strategy for cancer treatment. In this study, we developed an in‐house clinical sequencing system to perform a comprehensive cancer genomic profiling test as a clinical examination and analyzed the utility of this system. Genomic DNA was extracted from tumor tissues and peripheral blood cells collected from 161 patients with different stages and types of cancer. A comprehensive targeted amplicon exome sequencing for 160 cancer‐related genes was performed using next‐generation sequencing (NGS). The sequencing data were analyzed using an original bioinformatics pipeline, and multiple cancer‐specific gene alterations were identified. The success rate of our test was 99% (160/161), while re‐biopsy was required for 24% (39/161) of the cases. Potentially actionable and actionable gene alterations were detected in 91% (145/160) and 46% (73/160) of the patients, respectively. The actionable gene alterations were frequently detected in PIK3CA (9%), ERBB2 (8%), and EGFR (4%). High tumor mutation burden (TMB) (≥10 mut/Mb) was observed in 12% (19/160) of the patients. The secondary findings in germline variants considered to be associated with hereditary tumors were detected in 9% (15/160) of the patients. Seventeen patients (11%, 17/160) were treated with genotype‐matched therapeutic agents, and the response rate was 47% (8/17). The median turnaround time for physicians was 20 days, and the median survival time after the initial visit was 8.7 months. The results of the present study prove the feasibility of implementing in‐house clinical sequencing as a promising laboratory examination technique for precision cancer medicine.
Collapse
Affiliation(s)
- Hideyuki Hayashi
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Shigeki Tanishima
- Department of Biomedical Informatics Development, Mitsubishi Space Software Co., Ltd, Tokyo, Japan
| | - Kyoko Fujii
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan
| | - Ryo Mori
- Department of Biomedical Informatics Development, Mitsubishi Space Software Co., Ltd, Tokyo, Japan
| | - Chihiro Okada
- Department of Biomedical Informatics Development, Mitsubishi Space Software Co., Ltd, Tokyo, Japan
| | - Emmy Yanagita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| | - Yuka Shibata
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Ryosuke Matsuoka
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Pathology, International University of Health and Welfare, Narita, Japan
| | - Toraji Amano
- Clinical Research and Medical Innovation Center, Hokkaido University Hospital, Sapporo, Japan
| | - Takahiro Yamada
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan.,Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Ichiro Yabe
- Division of Clinical Genetics, Hokkaido University Hospital, Sapporo, Japan
| | - Ichiro Kinoshita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yoshito Komatsu
- Department of Cancer Chemotherapy, Hokkaido University Hospital Cancer Center, Sapporo, Japan
| | - Hirotoshi Dosaka-Akita
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Department of Medical Oncology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Hiroshi Nishihara
- Division of Clinical Cancer Genomics, Hokkaido University Hospital, Sapporo, Japan.,Genomics Unit, Keio Cancer Center, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
34
|
Ma Y, Li Q, Du Y, Chen W, Zhao G, Liu X, Ye L, Li H, Wang X, Liu J, Shen Z, Ma L, Zhou Y. Tumor Mutational Burden and PD-L1 Expression in Non-Small-Cell Lung Cancer (NSCLC) in Southwestern China. Onco Targets Ther 2020; 13:5191-5198. [PMID: 32606739 PMCID: PMC7292484 DOI: 10.2147/ott.s255947] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/22/2020] [Indexed: 12/12/2022] Open
Abstract
PURPOSE To explore the impact between the tumor mutational burden (TMB) and programmed death ligand-1 (PD-L1) expression on NSCLC in the Yunnan region of southwestern China. PATIENTS AND METHODS Seventy-one NSCLC specimens that were pathologically confirmed were collected at first. The TMB and driver genetic alterations were evaluated accordingly by next-generation sequencing (NGS). Afterwards, clinical parameters and tumor PD-L1 expressions were collected. Finally, the relationship between TMB, PD-L1 expression and clinical outcome was evaluated. RESULTS The median TMB was 5 (0.6-49) mutations/Mb by our NGS panel and the majority of patients (63/71, 88.7%) did not receive immunotherapy. The progression-free survival (PFS) was longer in TMB-low patients versus TMB-high ones (median 18.0 vs. 9.0 months, hazard ratio = 0.34, 95% confidence interval 0.14 to 0.84, p = 0.02) and the cut-off value was 10 mutations/Mb. The overall survival (OS) was longer in TMB-low patients vs. TMB-high ones (median 21.0 vs. 10.0 months, HR = 0.32, 95% CI 0.12 to 0.82, p = 0.02). Notably, our study also found that, excluding the eight patients with immunotherapy, the PFS was longer in patients with TMB-low vs. TMB-high (median 19.0 vs. 8.0 months, HR = 0.11, 95% CI 0.03 to 0.39, p < 0.01) and the OS was longer in TMB-low patients vs. TMB-high (median 21.0 vs 10.0 months, HR = 0.12, 95% CI 0.03 to 0.42, p < 0.01). CONCLUSION TMB was a valid and independent prognostic biomarker for NSCLC patients' clinical outcome and comprehensive screening of TMB based on NGS is recommended for individualized treatment strategies in Yunnan population.
Collapse
Affiliation(s)
- Yuhui Ma
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming650118, People’s Republic of China
| | - Quan Li
- Key Laboratory of Lung Cancer Research of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| | - Yaxi Du
- Key Laboratory of Lung Cancer Research of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| | - Wanlin Chen
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming650118, People’s Republic of China
| | - Guangqiang Zhao
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming650118, People’s Republic of China
| | - Xing Liu
- Key Laboratory of Lung Cancer Research of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| | - Lianhua Ye
- Department of Thoracic Surgery I, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming650118, People’s Republic of China
| | - Hongsheng Li
- International Joint Laboratory on High Altitude Regional Cancer of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| | - Xiaoxiong Wang
- International Joint Laboratory on High Altitude Regional Cancer of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| | - Junxi Liu
- International Joint Laboratory on High Altitude Regional Cancer of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| | - Zhenghai Shen
- Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| | - Luyao Ma
- Key Laboratory of Lung Cancer Research of Yunnan Province, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| | - Yongchun Zhou
- Yunnan Cancer Center, The Third Affiliated Hospital of Kunming Medical University, Kunming650118, People’s Republic of China
| |
Collapse
|
35
|
Abstract
Advances in genomic medicine have enabled the development of precise cancer therapies (precision cancer medicine) through multigene testing. Toward this end, we have developed a novel clinical sequencing system called PleSSision (Pathologist edited, Mitsubishi Space Software supervised clinical sequence system for personalized medicine) that performs amplicon exome sequencing targeting 160 cancer genes. Using this system, we have examined more than 600 cases over 3 years, and have identified druggable gene alteration in approximately 60% of the cases. Performing such clinical sequencing requires management of the sample quality and sequencing by pathologists and laboratory technicians; bioinformatics analysis by biomedical scientists; and patient care by nurses and pharmacists, all based on specific skills and knowledge of genomics. In addition, patients diagnosed with a hereditary cancer syndrome based on clinical sequencing results must receive care from a genetic counselor and a medical doctor with expertise in genetics. Recently, poly(ADP-ribose)polymerase (PARP) inhibitors and immune checkpoint inhibitors have been used in the treatment of patients with hereditary cancer syndromes, so collaboration involving other medical staff, especially genomic pharmacists, is also required. In this session, we provide an overview of cancer genomic medicine and emphasize the role that genomic pharmacists play in cancer precision medicine.
Collapse
Affiliation(s)
- Eriko Aimono
- Division of Clinical Cancer Genomics, Keio University School of Medicine
| | - Hiroshi Nishihara
- Division of Clinical Cancer Genomics, Keio University School of Medicine
| |
Collapse
|
36
|
Lombardo R, Tosi F, Nocerino A, Bencardino K, Gambi V, Ricotta R, Spina F, Siena S, Sartore-Bianchi A. The Quest for Improving Treatment of Cancer of Unknown Primary (CUP) Through Molecularly-Driven Treatments: A Systematic Review. Front Oncol 2020; 10:533. [PMID: 32457826 PMCID: PMC7225282 DOI: 10.3389/fonc.2020.00533] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 03/25/2020] [Indexed: 01/01/2023] Open
Abstract
Background: Carcinomas of unknown primary (CUP) account for 3–5% of all malignancy and, despite a reduction in incidence, the overall survival has not improved over the last decade. Chemotherapy regimens have not provided encouraging results. New diagnostic technologies, such as next generation sequencing (NGS), could represent a chance to identify potentially targetable genomic alterations in order to personalize treatment of CUP and provide insights into tumor biology. Methods: A systematic review of studies of patients with CUP, whose tumor specimen was evaluated through a NGS panel, has been performed on June 10th, 2019 according to PRISMA criteria from PubMed, ASCO meeting library and Clinicaltrial.gov. We have identified potentially targetable alterations for which approved/off-label/in clinical trials drugs are available. Moreover, we have included case reports about CUP patients treated with targeted therapies driven by NGS results in order to explore the clinical role of NGS in this setting. Results: We have evaluated 15 publications of which eleven studies (9 full-text articles and 2 abstracts) have analyzed the genomic profiling of CUPs through NGS technology, with different platforms and with different patients cohorts, ranging from 16 to 1,806 patients. Among all these studies, 85% of patients demonstrated at least one molecular alteration, the most frequent involving TP53 (41.88%), KRAS (18.81%), CDKN2A (8.8%), and PIK3CA (9.3%). A mean of 47.3% of patients harbored a potentially targetable alteration for which approved/off-label/in clinical trials drugs were available. Furthermore, we have identified 4 case reports in order to evaluate the clinical relevance of a specific targeted therapy identified through NGS. Conclusions: NGS may represent a tool to improve diagnosis and treatment of CUP by identifying therapeutically actionable alterations and providing insights into tumor biology.
Collapse
Affiliation(s)
- Roberta Lombardo
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Federica Tosi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Annunziata Nocerino
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Katia Bencardino
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Valentina Gambi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Riccardo Ricotta
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Francesco Spina
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Salvatore Siena
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Andrea Sartore-Bianchi
- Niguarda Cancer Center, Grande Ospedale Metropolitano Niguarda, Milan, Italy.,Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
37
|
Feng F, Cheng Q, Zhang D, Li B, Qin H, Xu C, Han M, Yu Y, Li Z, Li JY, Qiu Z, Xiong L, Liu C, Li F, Yi B, Jiang X. Targeted deep sequencing contributes to guiding personalized targeted therapy for advanced biliary tract cancer patients with non‑radical resection: A real‑world study. Oncol Rep 2020; 43:1089-1102. [PMID: 32323774 PMCID: PMC7057932 DOI: 10.3892/or.2020.7491] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 01/10/2020] [Indexed: 12/30/2022] Open
Abstract
Targeted therapy based on specific genetic alterations has been proven to be an effective treatment for various types of cancer. In the present study, we aimed to explore the efficacy of personalized targeted therapy guided by targeted deep sequencing for patients with advanced biliary tract cancer (BTC) after non-radical resection. Targeted deep sequencing was performed on 49 patients with BTC, to whom biologic agents were recommended. Among 32 patients with stage IV and R2 resection (a non-radical resection), 21 patients underwent conventional chemotherapy (mGEMOX), while the remaining 11 patients received a personalized targeted agent. The genomic landscape of the 49 patients with BTC was determined and the results showed that genetic alterations were enriched in the ERBB family and cell cycle pathway. After a median follow-up of 12 months, the 11 BTC patients with personalized targeted therapy showed a median progression-free survival (PFS) of 4.5 months (2.5–20.5 months), a median overall survival (OS) of 12.9 months (4.7–24.8 months) and a disease control rate (DCR) of 63.6%. In the other 21 BTC patients, who were undergoing conventional chemotherapy, the BTC patients had a median PFS of 1.5 months (0.5–11.6 months), a median OS of 4.1 months (1.3–18.4 months), and a DCR of 33.3%. In addition, 36.4% of the patients in the personalized targeted therapy group experienced grade >2 treatment-related toxicity vs. 19.0% of patients in the conventional chemotherapy group. This real-world study suggests that targeted deep sequencing contributes to the guidance of personalized targeted therapy based on individual actionable mutations, which may benefit advanced BTC patients undergoing non-radical resection.
Collapse
Affiliation(s)
- Feiling Feng
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Qingbao Cheng
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Dadong Zhang
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Bin Li
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Hao Qin
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Chang Xu
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Miao Han
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Yong Yu
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Zhizhen Li
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Jing-Yu Li
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Zhiquan Qiu
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Lei Xiong
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Chen Liu
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Fugen Li
- Research and Development Institute of Precision Medicine, 3D Medicines Inc., Shanghai 201114, P.R. China
| | - Bin Yi
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| | - Xiaoqing Jiang
- Department of Biliary I, Shanghai Eastern Hepatobiliary Surgery Hospital, Navy Military Medical University, Shanghai 200438, P.R. China
| |
Collapse
|
38
|
Ebi H, Bando H. Precision Oncology and the Universal Health Coverage System in Japan. JCO Precis Oncol 2019; 3:1900291. [PMID: 32923862 PMCID: PMC7446489 DOI: 10.1200/po.19.00291] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2019] [Indexed: 12/22/2022] Open
Abstract
Although precision oncology is transforming clinical management of patients with cancer, many hospitals face challenges to effectively implement precision oncology. In addition, the cost and time exerted for genomic profiling needs to be balanced with expectations of benefit for each patient. This article summarizes the effort to implement precision oncology in Japan. The most promising development is that tests to profile the genomes of select cancers are now fully covered by the national health insurance system. In May 2019, two gene panels were approved with reimbursement: FoundationOne CDx Cancer Genomic Profile and OncoGuide NCC Oncopanel System, the latter of which was developed in Japan. To make better use of scarce resources, the reimbursement is restricted to patients with solid tumors that have progressed on standard chemotherapy, rare tumors, or tumors of unknown primary. To centralize Japanese precision oncology, the government designated approximately 170 hospitals and stratified them to three layers on the basis of their roles. In addition, Japan’s National Cancer Center launched a Center for Cancer Genomics and Advanced Therapeutics (C-CAT) that collects genomic information and clinical characteristics of patients who received genomic profiling tests. C-CAT is expected to be the central data repository, to match patients with clinical trials, and to assist translational research. The centralized system under the national health insurance system could be a double-edged sword. Although tight regulation may make it hard to keep up with the rapid development of precision oncology, a federated ecosystem for sharing clinical and genomic data will be a precious asset and allow for shared access to data. Access to unapproved drugs and administrative support from C-CAT will be keys for Japanese precision oncology to meet its full potential.
Collapse
Affiliation(s)
- Hiromichi Ebi
- Division of Molecular Therapeutics, Aichi Cancer Center Research Institute, Nagoya, Japan.,Precision Medicine Center, Aichi Cancer Center, Nagoya, Japan.,Division of Advanced Cancer Therapeutics, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hideaki Bando
- Department of Clinical Oncology, Aichi Cancer Center Hospital, Nagoya, Japan
| |
Collapse
|
39
|
Yamamoto Y, Kanai M, Kou T, Sugiyama A, Nakamura E, Miyake H, Yamada T, Nishigaki M, Kondo T, Murakami H, Torishima M, Matsumoto S, Kosugi S, Muto M. Clinical significance of TP53 variants as possible secondary findings in tumor-only next-generation sequencing. J Hum Genet 2019; 65:125-132. [PMID: 31628423 PMCID: PMC6917569 DOI: 10.1038/s10038-019-0681-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 09/24/2019] [Indexed: 12/16/2022]
Abstract
In tumor-only next-generation sequencing (NGS), identified variants have the potential to be secondary findings (SFs), but they require verification through additional germline testing. In the present study, 194 patients with advanced cancer who underwent tumor-only NGS between April 2015 and March 2018 were enrolled, and the incidences of possible and true SFs were evaluated. Among them, 120 patients (61.9%) harbored at least one possible SF. TP53 was the most frequent gene in which 97 variants were found in 91 patients (49.5%). Nine patients provided informed consent to undergo additional germline testing, and a total of 14 variants (BRCA1, n = 1; BRCA2, n = 2; PTEN, n = 2; RB1, n = 1; SMAD4, n = 1; STK11, n = 1; TP53, n = 6) were analyzed. Three variants (BRCA1, n = 1; BRCA2, n = 2) were confirmed to be SFs, whereas TP53 variants were confirmed to be somatic variants. To confirm the low prevalence of SFs in TP53, we analyzed 24 patients with TP53 variants who underwent a paired tumor–normal NGS assay. As expected, all TP53 variants were confirmed to be somatic variants. A total of 30 patients were tested for germline variants in TP53, but none of them resulted in true SFs, suggesting the low prevalence of SFs in this gene. Therefore, the significance of additional germline testing for TP53 variants appears to be relatively low in daily clinical practice using a tumor-only NGS assay, unless patients have any relevant medical or family history.
Collapse
Affiliation(s)
- Yoshihiro Yamamoto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Kanai
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| | - Tadayuki Kou
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aiko Sugiyama
- DSK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eijiro Nakamura
- DSK Project, Medical Innovation Center, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidehiko Miyake
- Faculty of Core Research, Natural Science Division, Ochanomizu University, Tokyo, Japan.,Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Takahiro Yamada
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan.,Department of Medical Ethics and Medical Genetics, Kyoto University School of Public Health, Kyoto, Japan
| | - Masakazu Nishigaki
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan.,Department of Human Health Sciences, School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Hiromi Murakami
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | | | - Shigemi Matsumoto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinji Kosugi
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan.,Department of Medical Ethics and Medical Genetics, Kyoto University School of Public Health, Kyoto, Japan
| | - Manabu Muto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
40
|
Kiyozumi Y, Matsubayashi H, Horiuchi Y, Higashigawa S, Oishi T, Abe M, Ohnami S, Urakami K, Nagashima T, Kusuhara M, Miyake H, Yamaguchi K. Germline mismatch repair gene variants analyzed by universal sequencing in Japanese cancer patients. Cancer Med 2019; 8:5534-5543. [PMID: 31386297 PMCID: PMC6745857 DOI: 10.1002/cam4.2432] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 06/25/2019] [Accepted: 07/04/2019] [Indexed: 12/16/2022] Open
Abstract
Background Lynch syndrome (LS) is the commonest inherited cancer syndrome caused by pathogenic variants of germline DNA mismatch repair (g.MMR) genes. Genome‐wide sequencing is now increasingly applied for tumor characterization, but not for g.MMR. The aim of this study was to evaluate the incidence and pathogenicity of g.MMR variants in Japanese cancer patients. Methods Four g.MMR genes (MLH1, MSH2, MSH6, and PMS2) were analyzed by next generation sequencing in 1058 cancer patients (614 male, 444 female; mean age 65.6 years) without past diagnosis of LS. The g.MMR variant pathogenicity was classified based on the ClinVar 2015 database. Tumor MMR immunohistochemistry, microsatellite instability (MSI), and BRAF sequencing were also investigated in specific cases. Results Overall, 46 g.MMR variants were detected in 167 (15.8%) patients, 17 likely benign variants in 119 patients, 24 variants of uncertain significance (VUSs) in 68 patients, two likely pathogenic variants in two patients, and three pathogenic variants in three (0.3%) patients. The three pathogenic variants included two colorectal cancers with MLH1 loss and high MSI and one endometrial cancer with MSH6 loss and microsatellite stability. Two likely pathogenic variants were shifted to VUSs by ClinVar (2018). One colon cancer with a likely benign variant demonstrated MLH1 loss and BRAF mutation, but other nonpathogenic variants showed sustained MMR and microsatellite stability. Conclusions Universal sequencing of g.MMR genes demonstrated sundry benign variants, but only a small proportion of cancer patients had pathogenic variants. Pathogenicity evaluation using the ClinVar database agreed with MSI, MMR immunohistochemistry, and BRAF sequencing.
Collapse
Affiliation(s)
- Yoshimi Kiyozumi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan
| | - Hiroyuki Matsubayashi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan.,Division of Endoscopy, Shizuoka Cancer Center, Shizuoka, Japan
| | - Yasue Horiuchi
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan.,Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | - Satomi Higashigawa
- Division of Genetic Medicine Promotion, Shizuoka Cancer Center, Shizuoka, Japan
| | - Takuma Oishi
- Division of Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Masato Abe
- Division of Pathology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Sumiko Ohnami
- Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| | | | - Takeshi Nagashima
- Shizuoka Cancer Center Research Institute, Shizuoka, Japan.,SRL Inc., Tokyo, Japan
| | | | - Hidehiko Miyake
- Department of Genetic Counseling, Graduate School of Ochanomizu University, Tokyo, Japan
| | - Ken Yamaguchi
- Shizuoka Cancer Center Research Institute, Shizuoka, Japan
| |
Collapse
|
41
|
Davis W, Makar G, Mehta P, Zhu GG, Somer R, Morrison J, Kubicek GJ. Next-Generation Sequencing in 305 Consecutive Patients: Clinical Outcomes and Management Changes. J Oncol Pract 2019; 15:e1028-e1034. [PMID: 31373836 DOI: 10.1200/jop.19.00269] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
PURPOSE Next-generation sequencing (NGS) is increasingly used to identify actionable mutations for oncology treatment. We examined the results and use of NGS assays at our institution. PATIENTS AND METHODS We retrospectively reviewed the medical records of 305 consecutive patients who had NGS testing of tumor samples from March 2014 to April 2017. NGS was performed by FoundationOne. RESULTS Of the 305 tissue samples sent to FoundationOne, 189 reports were potentially usable. Of these reports, 76 (40.21%) demonstrated an aberration targetable by on-label therapies and 126 (66.67%) by off-label therapies, and 170 (89.94%) revealed actionable aberrations via all potential avenues, including clinical trials; 21 of these 189 potentially usable reports (11.1%) yielded a change in management, including use of on-label therapies (n = 7), use of off-label therapies (n = 6), enrollment in a clinical trial (n = 6), and discontinuation of a medication with a predicted poor response (n = 3; one report was used twice). For the six patients with off-label use, median duration of treatment was 46 days and discontinued after death (n = 3) or progression (n = 3). CONCLUSION Only a minority of NGS assay results (6.9% percent of all tests ordered and 11.1% of useable tests) resulted in a management change. A small minority of patients started off-label therapy on the basis of NSG assay results and overall had poor responses to off-label treatment. Although in theory NGS assays may improve oncologic outcomes, the results of our initial 305 patients showed low clinical utility.
Collapse
Affiliation(s)
- William Davis
- Cooper Medical School of Rowan University, Camden, NJ
| | - Gabriel Makar
- Cooper Medical School of Rowan University, Camden, NJ
| | | | | | | | | | | |
Collapse
|
42
|
Quy PN, Kanai M, Fukuyama K, Kou T, Kondo T, Yamamoto Y, Matsubara J, Hiroshima A, Mochizuki H, Sakuma T, Kamada M, Nakatsui M, Eso Y, Seno H, Masui T, Takaori K, Minamiguchi S, Matsumoto S, Muto M. Association Between Preanalytical Factors and Tumor Mutational Burden Estimated by Next-Generation Sequencing-Based Multiplex Gene Panel Assay. Oncologist 2019; 24:e1401-e1408. [PMID: 31186376 DOI: 10.1634/theoncologist.2018-0587] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 05/09/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Tumor mutational burden (TMB) measured via next-generation sequencing (NGS)-based gene panel is a promising biomarker for response to immune checkpoint inhibitors (ICIs) in solid tumors. However, little is known about the preanalytical factors that can affect the TMB score. MATERIALS AND METHODS Data of 199 patients with solid tumors who underwent multiplex NGS gene panel (OncoPrime), which was commercially provided by a Clinical Laboratory Improvement Amendments-licensed laboratory and covered 0.78 megabase (Mb) of capture size relevant to the TMB calculation, were reviewed. Associations between the TMB score and preanalytical factors, including sample DNA quality, sample type, sampling site, and storage period, were analyzed. Clinical outcomes of patients with a high TMB score (≥10 mutations per megabase) who received anti-programmed cell death protein 1 antibodies (n = 22) were also analyzed. RESULTS Low DNA library concentration (<5 nM), formalin-fixed paraffin-embedded tissue (FFPE), and the prolonged sample storage period (range, 0.9-58.1 months) correlated with a higher TMB score. After excluding low DNA library samples from the analysis, FFPE samples, but not the sample storage period, exhibited a marked correlation with a high TMB score. Of 22 patients with a high TMB score, we observed the partial response in 2 patients (9.1%). CONCLUSION Our results indicate that the TMB score estimated via NGS-based gene panel could be affected by the DNA library concentration and sample type. These factors could potentially increase the false-positive and/or artifactual variant calls. As each gene panel has its own pipeline for variant calling, it is unknown whether these factors have a significant effect in other platforms. IMPLICATIONS FOR PRACTICE A high tumor mutational burden score, as estimated via next-generation sequencing-based gene panel testing, should be carefully interpreted as it could be affected by the DNA library concentration and sample type.
Collapse
Affiliation(s)
- Pham Nguyen Quy
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Kanai
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Keita Fukuyama
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tadayuki Kou
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Kondo
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihiro Yamamoto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Junichi Matsubara
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akinori Hiroshima
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Hiroaki Mochizuki
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Tomohiro Sakuma
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Mayumi Kamada
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiko Nakatsui
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuji Eso
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshihiko Masui
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kyoichi Takaori
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Sachiko Minamiguchi
- Department of Diagnostic Pathology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigemi Matsumoto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
43
|
Eso Y, Kou T, Nagai H, Kim YH, Kanai M, Matsumoto S, Mishima M, Arasawa S, Iguchi E, Nakamura F, Takeda H, Takai A, Takahashi K, Ueda Y, Muto M, Seno H. Utility of ultrasound-guided liver tumor biopsy for next-generation sequencing-based clinical sequencing. Hepatol Res 2019; 49:579-589. [PMID: 30645782 DOI: 10.1111/hepr.13312] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Revised: 12/19/2018] [Accepted: 01/08/2019] [Indexed: 12/15/2022]
Abstract
AIM Recent advances in next-generation sequencing (NGS) technologies allow for evaluation of genetic alterations in various cancer-related genes in daily clinical practice. Archival formalin-fixed paraffin-embedded (FFPE) tumor tissue is often used for NGS-based clinical sequencing assays; however, the success rate of NGS assays using archival FFPE tumor tissue is reported to be lower than that using fresh tumor tissue. We aimed to evaluate the feasibility and safety of ultrasound (US)-guided liver tumor biopsy for NGS-based multiplex gene assays. METHODS We compared the success rate of NGS assays between archival FFPE tumor tissues and US-guided liver tumor biopsy tissues, and summarized the treatment progress of the patients. RESULTS Next-generation sequencing assays using US-guided liver biopsy samples were successful in all patients (22/22), whereas the success rate with archival FFPE tumor tissue was 84.8% (151/178, P < 0.05). At least one potentially actionable genetic alteration was identified from the US-guided liver biopsy samples in 20 of 22 patients. Among the 18 patients with actionable genetic alterations targetable with drugs approved by the US Food and Drug Administration, eight initiated mutation-driven targeted therapies. Of these eight patients, four achieved partial response or stable disease for at least 4 months, and three were not assessable for response due to short exposure. There were no biopsy-related complications requiring additional treatment. CONCLUSION Our findings suggest that US-guided liver tumor biopsy is a useful and safe method for obtaining high-quality samples for NGS-based clinical sequencing. In cases with metastatic liver tumors, US-guided biopsy should be considered to provide accurate and optimal sequencing results for patients.
Collapse
Affiliation(s)
- Yuji Eso
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tadayuki Kou
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroki Nagai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Young Hak Kim
- Department of Respiratory Medicine, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigemi Matsumoto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masako Mishima
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Soichi Arasawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Eriko Iguchi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Fumiyasu Nakamura
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhiko Takeda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Atsushi Takai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Ken Takahashi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshihide Ueda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
44
|
Sunami K, Ichikawa H, Kubo T, Kato M, Fujiwara Y, Shimomura A, Koyama T, Kakishima H, Kitami M, Matsushita H, Furukawa E, Narushima D, Nagai M, Taniguchi H, Motoi N, Sekine S, Maeshima A, Mori T, Watanabe R, Yoshida M, Yoshida A, Yoshida H, Satomi K, Sukeda A, Hashimoto T, Shimizu T, Iwasa S, Yonemori K, Kato K, Morizane C, Ogawa C, Tanabe N, Sugano K, Hiraoka N, Tamura K, Yoshida T, Fujiwara Y, Ochiai A, Yamamoto N, Kohno T. Feasibility and utility of a panel testing for 114 cancer-associated genes in a clinical setting: A hospital-based study. Cancer Sci 2019; 110:1480-1490. [PMID: 30742731 PMCID: PMC6447843 DOI: 10.1111/cas.13969] [Citation(s) in RCA: 220] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2018] [Revised: 01/21/2019] [Accepted: 02/06/2019] [Indexed: 12/12/2022] Open
Abstract
Next-generation sequencing (NGS) of tumor tissue (ie, clinical sequencing) can guide clinical management by providing information about actionable gene aberrations that have diagnostic and therapeutic significance. Here, we undertook a hospital-based prospective study (TOP-GEAR project, 2nd stage) to investigate the feasibility and utility of NGS-based analysis of 114 cancer-associated genes (the NCC Oncopanel test). We examined 230 cases (comprising more than 30 tumor types) of advanced solid tumors, all of which were matched with nontumor samples. Gene profiling data were obtained for 187 cases (81.3%), 111 (59.4%) of which harbored actionable gene aberrations according to the Clinical Practice Guidelines for Next Generation Sequencing in Cancer Diagnosis and Treatment (Edition 1.0) issued by 3 major Japanese cancer-related societies. Twenty-five (13.3%) cases have since received molecular-targeted therapy according to their gene aberrations. These results indicate the utility of tumor-profiling multiplex gene panel testing in a clinical setting in Japan. This study is registered with UMIN Clinical Trials Registry (UMIN 000011141).
Collapse
|
45
|
Bai Y, Wang G, Wei J, Dai C, Xu X, Cai X, Wu B, Sun W, Xu Q, Jiao S. Using clinical genomic sequencing to guide personalized cancer therapy in China. Per Med 2019; 16:287-299. [PMID: 30895868 DOI: 10.2217/pme-2018-0056] [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] [Indexed: 11/21/2022]
Abstract
Aim: To evaluate whether clinical genomic sequencing may benefit Chinese patients with stage IV cancer. Patients & methods: Chinese patients with cancer and their oncologists were provided with genomic sequencing results and corresponding clinical treatment recommendations based on evidence-based medicine, defined as CWES (clinical whole-exome sequencing) analysis. Chinese patients with stage IV cancer who failed the previous treatment upon receiving the CWES reports were included for analyzing the impact of CWES on clinical outcomes in 1-year follow-ups. Results: A total of 88.6% of 953 Chinese patients with cancer had clinically actionable somatic genomic alterations. Eleven patients followed the CWES reports, and 11 patients did not follow the CWES suggestions. The median progression-free survival of two groups were 12 and 4 months, and 45 and 91% of patients failed this round of therapy, respectively. Conclusion: The current study suggested that CWES has the potential to increase clinical benefits for Chinese patients with stage IV cancer.
Collapse
Affiliation(s)
- Yuxian Bai
- Department of Digestive Internal Medicine & Photodynamic Therapy Center, Harbin Medical University Cancer Hospital
| | - Guan Wang
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Jinwang Wei
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Chun Dai
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Xiaoman Xu
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Xin Cai
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Bing Wu
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Wending Sun
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Qiang Xu
- GenomiCare Biotechnology Co. Ltd, Shanghai, China
| | - Shunchang Jiao
- Department of Medical Oncology, Chinese PLA General Hospital, Beijing
| |
Collapse
|
46
|
Nagahashi M, Shimada Y, Ichikawa H, Kameyama H, Takabe K, Okuda S, Wakai T. Next generation sequencing-based gene panel tests for the management of solid tumors. Cancer Sci 2019; 110:6-15. [PMID: 30338623 PMCID: PMC6317963 DOI: 10.1111/cas.13837] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 10/13/2018] [Accepted: 10/16/2018] [Indexed: 12/16/2022] Open
Abstract
Next generation sequencing (NGS) has been an invaluable tool to put genomic sequencing into clinical practice. The incorporation of clinically relevant target sequences into NGS-based gene panel tests has generated practical diagnostic tools that enable individualized cancer-patient care. The clinical utility of gene panel testing includes investigation of the genetic basis for an individual's response to therapy, such as signaling pathways associated with a response to specific therapies, microsatellite instability and a hypermutated phenotype, and deficiency in the DNA double-strand break repair pathway. In this review, we describe the concept of precision cancer medicine using target sequences in gene panel tests as well as the importance of the control of sample quality in routine NGS-based genomic testing. We describe geographic and ethnic differences in cancer genomes, and discuss issues that need to be addressed in the future based on our experiences in Japan.
Collapse
Affiliation(s)
- Masayuki Nagahashi
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Yoshifumi Shimada
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Hiroshi Ichikawa
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Hitoshi Kameyama
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Kazuaki Takabe
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
- Breast SurgeryRoswell Park Cancer InstituteBuffaloNew York
- Department of SurgeryThe State University of New York Jacobs School of Medicine and Biomedical SciencesUniversity at BuffaloBuffaloNew York
| | - Shujiro Okuda
- Division of BioinformaticsGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| | - Toshifumi Wakai
- Division of Digestive and General SurgeryGraduate School of Medical and Dental SciencesNiigata UniversityNiigataJapan
| |
Collapse
|
47
|
Taniwaki M, Yamasaki M, Kawata K, Kawamoto K, Funaishi K, Matsumoto Y, Matsumoto N, Ohashi N, Hattori N. ROS1-rearranged putative lung adenocarcinoma presenting as carcinoma of unknown primary site: a case report. Oncotarget 2018; 9:35278-35282. [PMID: 30443294 PMCID: PMC6219662 DOI: 10.18632/oncotarget.26233] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 10/06/2018] [Indexed: 11/25/2022] Open
Abstract
Carcinoma of unknown primary site (CUP) is diagnosed only in 2-9% of all cancer cases. Adenocarcinomas account for approximately 60% of CUP, and some of these are putative lung adenocarcinomas. The frequency of driver oncogene positivity in the putative lung adenocarcinomas is unknown, and the efficacy of targeting therapies for the driver oncogene is also unknown. This is the first case report of C-ros oncogene 1 (ROS1)-rearranged putative lung adenocarcinoma presenting as CUP showing a good response to ROS1 inhibitor therapy. A 55-year-old woman presented with neck lymphadenopathy. Computed tomography and [18F]-fluorodeoxyglucose (FDG) positron emission tomography (PET) showed swelling of the bilateral supraclavicular, left accessory, mediastinal, and abdominal lymph nodes. The pathological analysis of the lymph node specimen biopsy indicated adenocarcinoma with cytokeratin 7 and thyroid transcription factor-1 positivity. Thus, this case was identified as ROS1- rearranged putative lung adenocarcinoma presenting as CUP. Oral crizotinib, an ROS1 inhibitor, was administered at a dose of 250 mg twice daily. Four weeks later, several swollen nodes showed marked improvement, and eight weeks later, FDG PET showed almost no uptake. In conclusion, putative lung adenocarcinoma presenting as CUP may involve ROS1 rearrangement, and ROS1 inhibitor therapy may be effective.
Collapse
Affiliation(s)
- Masaya Taniwaki
- Department of Respiratory Disease, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Naka-ku, Hiroshima, Japan
| | - Masahiro Yamasaki
- Department of Respiratory Disease, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Naka-ku, Hiroshima, Japan
| | - Koto Kawata
- Department of Respiratory Disease, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Naka-ku, Hiroshima, Japan
| | - Kazuma Kawamoto
- Department of Respiratory Disease, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Naka-ku, Hiroshima, Japan
| | - Kunihiko Funaishi
- Department of Respiratory Disease, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Naka-ku, Hiroshima, Japan
| | - Yu Matsumoto
- Department of Respiratory Disease, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Naka-ku, Hiroshima, Japan
| | - Naoko Matsumoto
- Department of Respiratory Disease, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Naka-ku, Hiroshima, Japan
| | - Nobuyuki Ohashi
- Department of Respiratory Disease, Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Naka-ku, Hiroshima, Japan.,Ohashi Clinic, Naka-ku, Hiroshima, Japan
| | - Noboru Hattori
- Department of Molecular and Internal Medicine, Institute of Biomedical and Health Sciences, Hiroshima University, Minami-ku, Hiroshima, Japan
| |
Collapse
|
48
|
Kondo T, Kanai M, Kou T, Sakuma T, Mochizuki H, Kamada M, Nakatsui M, Uza N, Kodama Y, Masui T, Takaori K, Matsumoto S, Miyake H, Okuno Y, Muto M. Association between homologous recombination repair gene mutations and response to oxaliplatin in pancreatic cancer. Oncotarget 2018; 9:19817-19825. [PMID: 29731985 PMCID: PMC5929428 DOI: 10.18632/oncotarget.24865] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 03/15/2018] [Indexed: 12/12/2022] Open
Abstract
Objectives We aimed to examine the association between homologous recombination repair (HRR)-related gene mutations and efficacy of oxaliplatin-based chemotherapy in patients with pancreatic ductal adenocarcinoma (PDAC). Results Non-synonymous mutations in HRR-related genes were found in 13 patients and only one patient had a family history of pancreatic cancer. Eight patients with HRR-related gene mutations (group A) and nine without HRR-related gene mutations (group B) received oxaliplatin-based chemotherapy. Median progression-free survival after initiation of oxaliplatin-based chemotherapy was significantly longer in group A than in group B (20.8 months vs 1.7 months, p = 0.049). Interestingly, two patients with inactivating HRR-related gene mutations who received FOLFIRINOX as first-line treatment showed exceptional responses with respect to progression-free survival for > 24 months. Materials and Methods Complete coding exons of 12 HRR-related genes (ATM, ATR, BAP1, BRCA1, BRCA2, BLM, CHEK1, CHEK2, FANCA, MRE11A, PALB2, and RAD51) were sequenced using a Clinical Laboratory Improvement Amendment-certified multiplex next-generation sequencing assay. Thirty consecutive PDAC patients who underwent this assay between April 2015 and July 2017 were included. Conclusions Our results suggest that inactivating HRR-related gene mutations are predictive of response to oxaliplatin-based chemotherapy in patients with PDAC.
Collapse
Affiliation(s)
- Tomohiro Kondo
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masashi Kanai
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tadayuki Kou
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Sakuma
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Hiroaki Mochizuki
- Biomedical Department, Mitsui Knowledge Industry Co., Ltd., Tokyo, Japan
| | - Mayumi Kamada
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masahiko Nakatsui
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Norimitsu Uza
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuzo Kodama
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Toshihiko Masui
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Kyoichi Takaori
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shigemi Matsumoto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hidehiko Miyake
- Clinical Genetics Unit, Kyoto University Hospital, Kyoto, Japan
| | - Yasushi Okuno
- Department of Biomedical Data Intelligence, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Manabu Muto
- Department of Medical Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
49
|
Kohno T. Implementation of "clinical sequencing" in cancer genome medicine in Japan. Cancer Sci 2018; 109:507-512. [PMID: 29285848 PMCID: PMC5834803 DOI: 10.1111/cas.13486] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/24/2017] [Accepted: 12/25/2017] [Indexed: 12/24/2022] Open
Abstract
In oncology, actionable mutations (alterations) in cancer-associated genes are critical in terms of the selection of therapeutic approaches. Next-generation sequencing of tumor sample DNA (ie, clinical sequencing) can guide clinical management by providing diagnostic or prognostic data, and facilitating the identification of potential treatment regimens, such as molecular-targeted and immune checkpoint blockade therapies. In the USA, a variety of tumor-profiling multiplex gene panels have been developed and implemented for this purpose. In Japan, several academic institutions have now carried out detailed investigations of the feasibility and value of clinical sequencing, and cancer societies have issued consensus clinical practice guidance for next-generation sequencing-based gene panel tests. These efforts will facilitate the implementation of cancer genome medicine in Japan.
Collapse
Affiliation(s)
- Takashi Kohno
- Division of Genome BiologyNational Cancer Center Research InstituteTokyoJapan
- Division of Translational GenomicsExploratory Oncology Research and Clinical Trial CenterNational Cancer CenterTokyoJapan
| |
Collapse
|
50
|
Christensen PA, Ni Y, Bao F, Hendrickson HL, Greenwood M, Thomas JS, Long SW, Olsen RJ. Houston Methodist Variant Viewer: An Application to Support Clinical Laboratory Interpretation of Next-generation Sequencing Data for Cancer. J Pathol Inform 2017; 8:44. [PMID: 29226007 PMCID: PMC5719586 DOI: 10.4103/jpi.jpi_48_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 10/12/2017] [Indexed: 01/17/2023] Open
Abstract
Introduction Next-generation-sequencing (NGS) is increasingly used in clinical and research protocols for patients with cancer. NGS assays are routinely used in clinical laboratories to detect mutations bearing on cancer diagnosis, prognosis and personalized therapy. A typical assay may interrogate 50 or more gene targets that encompass many thousands of possible gene variants. Analysis of NGS data in cancer is a labor-intensive process that can become overwhelming to the molecular pathologist or research scientist. Although commercial tools for NGS data analysis and interpretation are available, they are often costly, lack key functionality or cannot be customized by the end user. Methods To facilitate NGS data analysis in our clinical molecular diagnostics laboratory, we created a custom bioinformatics tool termed Houston Methodist Variant Viewer (HMVV). HMVV is a Java-based solution that integrates sequencing instrument output, bioinformatics analysis, storage resources and end user interface. Results Compared to the predicate method used in our clinical laboratory, HMVV markedly simplifies the bioinformatics workflow for the molecular technologist and facilitates the variant review by the molecular pathologist. Importantly, HMVV reduces time spent researching the biological significance of the variants detected, standardizes the online resources used to perform the variant investigation and assists generation of the annotated report for the electronic medical record. HMVV also maintains a searchable variant database, including the variant annotations generated by the pathologist, which is useful for downstream quality improvement and research projects. Conclusions HMVV is a clinical grade, low-cost, feature-rich, highly customizable platform that we have made available for continued development by the pathology informatics community.
Collapse
Affiliation(s)
- Paul A Christensen
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Cornell Medical College of Cornell University, Houston, Texas, USA
| | - Yunyun Ni
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Cornell Medical College of Cornell University, Houston, Texas, USA.,Helix, San Carlos, California 94070, USA
| | - Feifei Bao
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Cornell Medical College of Cornell University, Houston, Texas, USA
| | - Heather L Hendrickson
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Cornell Medical College of Cornell University, Houston, Texas, USA
| | - Michael Greenwood
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Cornell Medical College of Cornell University, Houston, Texas, USA
| | - Jessica S Thomas
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Cornell Medical College of Cornell University, Houston, Texas, USA
| | - S Wesley Long
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Cornell Medical College of Cornell University, Houston, Texas, USA
| | - Randall J Olsen
- Department of Pathology and Genomic Medicine, Houston Methodist Hospital, Weill Cornell Medical College of Cornell University, Houston, Texas, USA
| |
Collapse
|