Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2024 Nov 1;14(11):2243-2261.
doi: 10.1158/2159-8290.CD-24-0206.

The SCRUM-MONSTAR Cancer-Omics Ecosystem: Striving for a Quantum Leap in Precision Medicine

Affiliations

The SCRUM-MONSTAR Cancer-Omics Ecosystem: Striving for a Quantum Leap in Precision Medicine

Tadayoshi Hashimoto et al. Cancer Discov. .

Abstract

The SCRUM-Japan MONSTAR-SCREEN consortium is a nationwide molecular profiling project employing artificial intelligence-driven multiomics analyses for patients with advanced malignancies, aiming to develop novel therapeutics and diagnostics and deliver effective drugs to patients. Concurrently, studies assessing molecular residual disease-based precision medicine for resectable solid tumors, including CIRCULATE-Japan, are ongoing. The substantial data generated by these platforms are stored within a state-of-the-art supercomputing infrastructure, VAPOR CONE. Since 2015, our project has registered over 24,000 patients as of December 2023. Among 16,144 patients with advanced solid tumors enrolled in MONSTAR-SCREEN projects, 5.0% have participated in matched clinical trials, demonstrating a 29.2% objective response rate and 14.8-month median survival (95% CI, 13.4-16.3) for patients treated in the matched clinical trials. Notably, patients who received matched therapy demonstrated significantly prolonged overall survival compared with those who did not (hazard ratio 0.77; 95% confidence interval, 0.71-0.83). Significance: Our nationwide molecular profiling initiative played pivotal roles in facilitating the enrollment of patients with advanced solid tumors into matched clinical trials and highlighted the substantial survival benefits of patients treated with matched therapy. We aim to facilitate an industry-academia data-sharing infrastructure ecosystem, fostering new drug discovery paradigms and precision medicine.

PubMed Disclaimer

Conflict of interest statement

Y. Nakamura reports personal fees from Guardant Health Pte, Ltd., Natera, Inc., Roche, Ltd., Premo Partners, Inc., Takeda Pharmaceutical, Exact Sciences Corporation, Gilead Sciences, Inc., MSD, Eisai Co., Ltd., Zeria Pharmaceutical Co., Ltd., Miyarisan Pharmaceutical Co., Ltd., Merck Biopharma Co., Ltd., CareNet, Inc., Hisamitsu Pharmaceutical Co., Inc., Taiho Pharmaceutical Co., Ltd., Becton, Dickinson and Company, and Guardant Health Japan Corp.; grants and personal fees from Seagen, Inc., Daiichi Sankyo Co., Ltd., and Chugai Pharmaceutical Co., Ltd.; as well as grants from Genomedia Inc., Guardant Health AMEA, Inc., Guardant Health, Inc., Tempus Labs, Inc., and Roche Diagnostics K.K. outside the submitted work. T. Fujisawa reports personal fees from Amelieff outside the submitted work. M. Imai reports personal fees from Caris Life Sciences, Guardant Health, Inc., and Sumitomo Corp outside the submitted work. N. Nonomura reports personal fees from Takeda Pharmaceutical, Janssen Pharmaceuticals, Pfizer, Astellas Pharma, Ono Pharmaceutical, Novartis, MSD, AstraZeneca, Merck Biopharma Co., Ltd., Bayer, and Bristol-Myers Squibb K.K. during the conduct of the study; as well as a patent for Shionogi issued. C. Morizane reports personal fees from Guardant Health, Chugai Pharmaceutical Co. Ltd., Myriad Genetics, and Servier; grants and personal fees from Boehringer Ingelheim; as well as grants from Ono Pharmaceutical, J-Pharma, Merck Biopharma Co., Ltd., and Daiichi Sankyo RD Novare outside the submitted work. H. Iwata reports personal fees from MSD, Daiichi Sankyo, Chugai Pharmaceutical Co. Ltd., AstraZeneca, Pfizer, and Eli Lilly Japan K. K. outside the submitted work. S. Okano reports personal fees from Merck Biopharma Co. Ltd., MSD, Bristol-Myers Squibb K. K., and Ono Pharmaceutical during the conduct of the study. S. Kadowaki reports grants and personal fees from Ono Pharmaceutical, Taiho Pharmaceutical, MSD, Bayer, Eli Lilly Japan K. K., Chugai Pharmaceutical Co. Ltd., and Daiichi Sankyo; grants from Nobel Pharmaceuticals, Janssen Pharmaceuticals, and AstraZeneca; as well as personal fees from Merck KGaA, Bristol-Myers Squibb K. K., and Eisai Co., Ltd., outside the submitted work. H. Taniguchi reports personal fees from Chugai Pharmaceutical Co. Ltd., Ono Pharmaceutical, Takeda, Eli Lilly Japan K. K., and Merck Biopharma Co. Ltd., as well as grants from Daiichi Sankyo outside the submitted work. M. Ueno reports grants and personal fees from Taiho Pharmaceutical Co., Ltd., AstraZeneca, MSD, Nihon Servier Co., Ltd., Ono Pharmaceutical, Incyte Biosciences Japan GK, Chugai Pharmaceutical Co., Ltd., Boehringer Ingelheim GmbH, J-Pharma Co., Ltd., Eisai Co., Ltd., and Novartis Pharma K.K; personal fees from Yakult Honsha Co., Ltd., and Takeda Pharmaceutical; as well as grants from Daiichi Sankyo Co., Ltd., Astellas Pharma, Delta Fly Pharma, Inc., Novocure GmbH, and Chiome Bioscience, Inc. outside the submitted work. E. Oki reports grants from Guardant Health, Inc., as well as personal fees from Chugai Pharmaceutical Co. Ltd., Eli Lilly Japan K. K., Takeda Pharmaceutical, Bristol-Myers Squibb K. K., and Ono Pharmaceutical outside the submitted work. Y. Komatsu reports grants and personal fees from Eli Lilly Japan K. K., Taiho Pharmaceutical, Chugai Pharmaceutical Co. Ltd., Takeda Pharmaceutical, Bristol-Myers Squibb K. K., Sanofi, Merck Biopharma Co. Ltd., Ono Pharmaceutical, Nipro, Daiichi Sankyo, and Yakult Honsha; personal fees from Bayer; as well as grants from Eisai Co. Ltd., Sysmex, and Astellas Pharma outside the submitted work. S. Yuki reports personal fees from Eli Lilly Japan K.K., Chugai Pharmaceutical Co., Ltd., Taiho Pharmaceutical Co., Ltd., Bayer Yakuhin Ltd., Bristol-Myers Squibb K.K., Takeda Pharmaceutical, Merck Biopharma Co., Ltd., MSD, Ono Pharmaceutical, and Miyarisan Pharmaceutical Co., Ltd., outside the submitted work. A. Makiyama reports personal fees from Eli Lilly Japan K.K., Taiho Pharmaceutical Co., Ltd., Ono Pharmaceutical, Bristol-Myers Squibb K. K., and Daiichi Sankyo Co., Ltd., outside the submitted work. H. Hara reports grants from AstraZeneca, Merck Biopharma Co. Ltd., MSD, Ono Pharmaceutical, Taiho Pharmaceutical, Boehringer Ingelheim, Daiichi Sankyo, BeiGene, Astellas Pharma, Bayer, Amgen, Chugai Pharmaceutical Co. Ltd., Janssen Oncology, ALX Oncology, Bristol-Myers Squibb K. K., Jazz Pharmaceuticals, and Oncolys Biopharma outside the submitted work. N. Okano reports personal fees from Taiho Pharmaceutical, Eli Lilly Japan K. K., Eisai Co. Ltd., Bayer Yakuhin, Chugai Pharmaceutical Co. Ltd., Ono Pharmaceutical, Daiichi Sankyo, AstraZeneca, MSD, Incyte, and Nihon Servier outside the submitted work. T. Nishina reports grants and personal fees from Bristol-Myers Squibb K. K., Ono Pharmaceutical, Taiho Pharmaceutical, Daiichi-Sankyo, MSD, and Astellas Pharma, as well as personal fees from Eli Lilly Japan K. K. outside the submitted work. M. Nagamine reports personal fees from SRL, Inc., GxD, Inc., Chugai Pharmaceutical Co., Ltd., and Bunkodo Co., Ltd., outside the submitted work. H. Bando reports personal fees from Ono Pharmaceutical, Taiho Pharmaceutical, and Eli Lilly Japan K. K. outside the submitted work. T. Kuwata reports personal fees from MSD, Daiichi Sankyo, Roche Diagnostics, Bayer, and FALCO Biosystems; personal fees and nonfinancial support from Astellas Pharma; as well as grants from Takeda Pharmaceutical outside the submitted work. W.-Y. Park reports personal fees from GxD, Inc., during the conduct of the study. A. Ohtsu reports grants and personal fees from Chugai Pharmaceutical Co. Ltd., and Taiho Pharmaceutical; grants from Takeda Pharmaceutical, Ono Pharmaceutical, Daiichi-Sankyo Pharmaceutical, Amgen, Inc., Eisai Co., Ltd., Kyowa-Kirin Co., Ltd., Novartis Pharmaceutical, Pfizer, Inc., Eli Lilly Japan K. K., MSD, AstraZeneca, Nippon Boehringer Ingelheim Co., Ltd., Merck Seronoi Co., Ltd., Bristol-Meyers Squibb K. K., Astellas Pharma, Medical & Biological Laboratories Co., Ltd., Sumitomo Pharma Co., Ltd., Janssen Pharmaceutical, Bayer Yakuhin, Ltd., Merus, AbbVie GK, and Flatiron Health, Inc. during the conduct of the study; as well as personal fees from A2 Healthcare outside the submitted work. T. Yoshino reports personal fees from Chugai Pharmaceutical Co. Ltd., Takeda Pharmaceutical, Merck Biopharma Co. Ltd., Bayer Yakuhin, Ono Pharmaceutical, MSD, and Sumitomo Corp, as well as grants from Amgen, Chugai Pharmaceutical, Daiichi Sankyo, Eisai Co. Ltd., FALCO Biosystems, Genomedia, Molecular Health, MSD, Nippon Boehringer Ingelheim Co., Ltd., Ono Pharmaceutical, Pfizer, Roche Diagnostics, Sanofi, Sysmex, and Taiho Pharmaceutical outside the submitted work. No disclosures were reported by the other authors.

Figures

Figure 1.
Figure 1.
Overview of the SCRUM-MONSTAR project. The SCRUM-Japan MONSTAR-SCREEN project, a nationwide molecular profiling project, uses AI-driven multiomics analyses for patients with advanced malignancies. The project encompasses various sub-studies for advanced solid tumors, including GI-SCREEN, GOZILA, MONSTAR-SCREEN, and MONSTAR-SCREEN-2. Concurrently, the CIRCULATE-Japan and COSMOS platforms manage MRD for resectable solid tumors. The BRANCH project explores the relevance of germline mutations in patients with solid tumor and their families. The project comprises several specialized committees, each diligently administered and operated by its respective group. These committees cover 12 areas: AI, ancillary research, database management, ethical, legal, and social implications (ELSI), genetic information, germline board, meta-information management, pathology, patient and public involvement (PPI), quality assurance/quality control (QA/QC), regulatory affairs, and trial management.
Figure 2.
Figure 2.
The MRD project for resectable solid tumors. A, Annual patient enrollment in the CIRCULATE-Japan and COSMOS project, exceeding 7,000 participants (light green: CIRCULATE-Japan; blue: COSMOS). B, The demographics of the enrolled patients according to cancer types in the CIRCULATE-Japan and COSMOS project. C, The schema of CIRCULATE-Japan related trials and COSMOS projects. CIRCULATE-Japan is composed of GALAXY, VEGA and ALTAIR studies. The GALAXY study is a prospective large-scale registry designed to monitor ctDNA in patients with CRC. The COSMOS projects are composed of six cancer specific studies: CRC, GC, PC, HCC, MM, and BTC to evaluate the utility of early postoperative MRD analysis and early detection of various cancer types. NEXUS, VOLTAGE II, and Nir-Bev trials are linked to the CIRCULATE-Japan platform. D, Comparison of the predictive performance of various biomarkers for cancer recurrence. The HR with 95% CIs for recurrence, recurrence-free survival, or disease-free survival were previously reported for several biomarkers, including tumor budding status, lymphovascular invasion status, tumor differentiation, tumor sidedness, serum carcinoembryonic antigen level, tumor-infiltrating lymphocytes, Immunoscore, CpG island methylator phenotype, microsatellite Instability status, gene expression signature (Recurrence Score and ColoPrint), gene mutational status (BRAF, KRAS, TP53, and PIK3CA), and molecular expression status (MUC2, CDX2, and HER2), and ctDNA status in the GALAXY study. Among these biomarkers, ctDNA positivity after surgery in the GALAXY study demonstrated a substantially higher HR for recurrence compared with the other biomarkers. ACT, adjuvant chemotherapy; CAPOX, capecitabine plus oxaliplatin; CDX2, caudal-type homeobox transcription factor 2; CEA, carcinoembryonic antigen; CIMP, CpG island methylator phenotype; cy, cycles; FTD/TPI, trifluridine/tipiracil; GIST, gastrointestinal stromal tumor; HCC, hepatocellular carcinoma; HER2, human epidermal growth factor receptor 2; LVI, lymphovascular invasion; MM, malignant melanoma; MSI, microsatellite instability; MUC2, mucin 2; R, randomization.
Figure 3.
Figure 3.
Consortium of the clinical trial platform and SCRUM-Japan registry. A, Annual participant enrollment across different projects, exceeding 16,000 participants (light green: GI-SCREEN, blue: GOZILA, light red: MONSTAR-SCREEN, light purple: MONSTAR-SCREEN-2, light orange: BRANCH, red line: total enrollment). B, The demographics of the enrolled participants according to cancer types in the SCRUM-Japan MONSTAR-SCREEN project. C, The schema of SCRUM-MONSTAR–related trials and SCRUM-Japan registry projects. Patients with specific genomic alterations are incorporated into pertinent clinical trials, with several tumor-specific and tumor-agnostic trials on the basis of MONSTAR-SCREEN platform. D, Nine trials completed enrollment. Of these trials, four (TRIUMPH, HERB, TiFFANY, and HERALD) met the primary endpoint, whereas two did not. Three trials are in the process of analyzing data. E, The structure of the SCRUM-Japan registry, which collects prospective data on patients with rare genetic alterations, informing future drug development and serving as an external control group. Currently, more than 500 patients with rare molecular alterations have been pooled, and their data will be used as the external control data for approval. BRCA, breast cancer; CervC, cervical cancer; EC, esophageal cancer; EDC, electronic data capture; EnC, endometrial cancer; GIST, gastrointestinal stromal tumor; HCC, hepatocellular carcinoma; HNC, head and neck cancer; IIT, investigator-initiated trial; MM, malignant melanoma; NEN, neuroendocrine neoplasms; OVC, ovarian cancer; PRAD, prostate adenocarcinoma; RCC, renal cell carcinoma; SIC, small intestinal cancer; UroC, urothelial cancer.
Figure 4.
Figure 4.
The overview of target biomarkers in clinical trials related to SCRUM-MONSTAR-SCREEN. A, Background characteristics of patients participating in clinical trials. B, Left, Treatment lines examined in clinical trials; Right, class of drugs examined in clinical trials. C, Targeted biomarkers in clinical trials. D, Overall drug efficacy in matched clinical trials. CR, complete response; PR, partial response; SD, stable disease; PD, progressive disease. E, Objective response rate by each targeted drug. FGFR, fibroblast growth factor receptor; HER2, human epidermal growth factor receptor 2; MEK, MAPK/ERK; MET, mesenchymal-epithelial transition; mTKI, multitargeted tyrosine kinase inhibitors; PARP1, poly(ADP-ribose) polymerase 1.
Figure 5.
Figure 5.
Long-term outcomes of patients matched to clinical trials and comparison of matched vs. nonmatched therapy. A, PFS in patients treated with biomarker-matched therapy in clinical trials. B, OS in patients treated with biomarker-matched therapy in clinical trials. C, OS of patients treated with matched therapy vs. nonmatched therapy. m, months; MST, median survival time.
Figure 6.
Figure 6.
Structure of SCRUM-Japan MONSTAR-SCREEN-3 and SCRUM-MONSTAR cyber space (VAPOR CONE). MONSTAR-SCREEN-3 consists of three cohorts: the Advanced (N = 1,700), Definitive (N = 1,100), and Hematology (N = 400) cohorts. Comprehensive multiomics analyses are designated per cohort, including various sequencing methods and evaluations for tissue and blood samples; WES, WTS, and spatial ST sequencing for tissue samples; WES, WTS, germline sequencing, and proteome analysis for blood samples; and microbiome analysis for fecal samples. Quality of life (QoL) evaluation using patent-reported outcomes from electric records, digital image collection, and digital pathology collection are also conducted. Sequential tissue–informed WGS-based MRD analysis is performed in the Definitive and Hematology cohorts, supplemented by IgH/TCR-based MRD in the Hematology cohort. VAPOR CONE represents a virtual space dedicated to the generation of quality-assured real-world evidence (RWE) within a secure HPC environment. This platform facilitates the development of RWE extraction, data analysis technologies, and various AI models, propelling forward the fields of cancer genomics and translational research across basic, applied, and clinical domains. Using generative AI technology, existing models within VAPOR CONE undergo continuous refinement, aiming to establish an advanced social infrastructure for cancer genome medicine, achieving decentralized clinical trial (DCT). Data within VAPOR CONE are merged with the collective expertise of a nationwide network of academic professionals. This synergy enables the reorganization, enrichment, and standardization of the VAPOR CONE database, aligning it with international standards in real time. The database serves as a robust, accurate data source, laying the groundwork for sophisticated applications in healthcare and pharmaceutical research. Continuous learning, facilitated by generative AI technology, enhances the machine intelligence of VAPOR CONE’s existing AI models, improving auxiliary diagnostic support, interpretation, report generation, and predictive capabilities for research outcomes. By bolstering the speed, efficiency, and cost-effectiveness of these processes, we aim to pioneer an unparalleled social infrastructure for advanced cancer genome medicine in Japan. ctNA, circulating tumor nucleic acid; CTx, chemotherapy; DCT, decentralized clinical trial; ePRO, electronic patient reported outcome; IgH/TCR, immunoglobulin receptor/T-cell receptor.

References

    1. Nakamura Y, Fujisawa T, Taniguchi H, Bando H, Okamoto W, Tsuchihara K, et al. . SCRUM-Japan GI-SCREEN and MONSTAR-SCREEN: path to the realization of biomarker-guided precision oncology in advanced solid tumors. Cancer Sci 2021;112:4425–32. - PMC - PubMed
    1. Taniguchi H, Nakamura Y, Kotani D, Yukami H, Mishima S, Sawada K, et al. . CIRCULATE-Japan: circulating tumor DNA-guided adaptive platform trials to refine adjuvant therapy for colorectal cancer. Cancer Sci 2021;112:2915–20. - PMC - PubMed
    1. Kotani D, Oki E, Nakamura Y, Yukami H, Mishima S, Bando H, et al. . Molecular residual disease and efficacy of adjuvant chemotherapy in patients with colorectal cancer. Nat Med 2023;29:127–34. - PMC - PubMed
    1. Sato S, Nakamura Y, Oki E, Yoshino T. Molecular residual disease-guided adjuvant treatment in resected colorectal cancer: focus on CIRCULATE-Japan. Clin Colorectal Cancer 2023;22:53–8. - PubMed
    1. Miyamoto Y, Hiyoshi Y, Sawayama H, Tokunaga R, Baba H. Precision medicine for adjuvant chemotherapy of resected colorectal cancer. Ann Gastroenterol Surg 2020;4:635–45. - PMC - PubMed