Prof Ramanuj DasGupta - Cancer Systems Biology and Tumour Evolution
Introduction
Unlocking the Developmental Origins of Cancer
Cancer remains one of the most complex, and devastating diseases of our time, characterized by its ability to evade treatment and adapt to an ever-changing biological landscape with diverse selective pressures. At the forefront of this challenge, our laboratory is pioneering the use of next-generation single-cell and spatial biology tools, paired with functional genomic technologies and relevant murine models, to unravel the developmental origins of cancer. Specifically, we investigate how damage-associated regenerative programs, activated in the context of chronic inflammatory diseases (fatty liver/MASLD, viral hepatis, colitis, IBD) contribute to cancer initiation and progression.
The Origins of Cancer: A Crossroads of Regeneration and Dysfunction
Recent work from our laboratory and others has elucidated remarkable foetal-like, developmental remodelling of the tumour microenvironment (TME) in human hepatocellular carcinoma (HCC) (Sharma et al., 2020, Cell; Nguyen et al., 2022, Nat Commun). Our current research is rooted in understanding how chronic inflammation, a hallmark of diseases such as colitis, chronic hepatitis, or pancreatitis, can activate similar developmental programs to establish a “pro-tumourigenic niche” or a fertile soil for tumourigenesis (Balakrishnan et al., 2024, J Hepatol; Cappellesso et al., 2022, Nat Cancer; Scolaro et al., 2024, Nat Cancer). During these inflammatory processes, regenerative programs are activated to repair damaged tissues. However, when dysregulated, these programs can lead to aberrant cell growth, epithelial damage, endothelial-immune dysfunction, fibrosis and eventually tumour formation. Using advanced single-cell and spatial biology tools (single cell RNA-seq, scATAC-seq, spatial transcriptomics, proteomics and metabolomics, and multi-parametric flow cytometry), we study the cellular and molecular mechanisms underlying this transition from chronic non-healing wounds to cancer. These technologies allow us to map cellular interactions at unprecedented resolution, providing insight into how damaged epithelial cells communicate with their surrounding microenvironment, including immune cells, stromal cells, and endothelial cells. Finally, our understanding of the molecular mechanisms of cross-regulatory interactions between tumour cells and their ecosystem is paving the way for innovative therapeutic approaches that we, in close collaborations with clinicians, are implementing as part of clinical trials aimed at interrogating the efficacy of combining drugs targeting the tumour stroma (anti-angiogenics) along with immune checkpoint inhibitors. Initial data from these clinical studies on nasopharyngeal carcinomas (NPC) show striking improvement in anti-PD1 response when combined with anti-VEGFa therapy, compared to the use of anti-PD1 alone as monotherapy (Chong et al., 2024, Lancet Oncol).
Cells to location: localizing cell types and cell states to their spatial coordinates in NPC
A Focus on Co-Evolutionary Mechanisms in the Tumour Microenvironment
Cancers do not evolve in isolation. It emerges and thrives in the context of its microenvironment, engaging in dynamic cross-regulatory interactions with surrounding cells. Our work centres on uncovering these co-evolutionary mechanisms that shape tumour initiation, tissue remodelling, and progression.
Key areas of focus include:
- Endothelial Dysfunction: Endothelial cells play a critical role in maintaining vascular integrity and regulating tissue homeostasis. In chronic inflammation, persistent endothelial dysfunction can promote abnormal angiogenesis, hypoxia, and endothelial anergy, fostering tumour growth and metastasis.
- Fibrosis: Fibroblasts, activated during tissue repair, can become cancer-associated fibroblasts (CAFs) that contribute to a fibrotic tumour microenvironment. This fibrosis not only supports tumour growth but also serves as a barrier to effective drug delivery, or immune-cell infiltration, making tumours more resistant to treatment.
- Immune Dysregulation: Chronic inflammation disrupts immune homeostasis, leading to immune suppression and the recruitment of tumour-promoting immune cells, such as pro-remodelling macrophages and regulatory T cells (Tregs). By studying how tumours exploit immune-stromal interactions, we aim to identify new therapeutic strategies targeting the tumour ecosystem to restore immune balance.
Bridging Technologies to Decode Tumour Evolution
Our laboratory employs an integrated approach, leveraging cutting-edge single cell and spatial multi-Omics technologies to study mechanisms of damage-associated chronic disease progression to cancer:
- Single-Cell Analysis: Tools like single-cell RNA sequencing (scRNA-seq, ATAC-seq) allow us to dissect cellular heterogeneity of disease-associated cell states (DACs) within tumours and their microenvironments, as well as gene regulatory networks and signalling pathways that specify the transcriptomic signatures of DACs
- Spatial Biology: Advanced imaging technologies enable us to visualize the geo-spatial organization and cellular interactions within tissue architecture, capturing the spatial dynamics of DACs or tumour cells with their ecosystem, including endothelial cells, fibroblasts and immune cells.
- Functional Genomics: CRISPR-based screens and drug libraries in relevant murine and patient-derived cell line/organoid models enable the identification of genes and pathways that drive tumour evolution, offering insights into potential therapeutic vulnerabilities.
Spatial organization of individual cell type within a tumour (left); spatial distribution of distinct neighbourhoods of interacting cells (niches) identified within tumour biopsies
From Discovery to Clinical Translation
Our ultimate goal is to translate these discoveries into tangible clinical benefits. By understanding the developmental origins of cancer and the role of co-evolutionary mechanisms, we aim to:
- Identify early biomarkers that predict tumour initiation and progression.
- Develop preventative therapies targeting the regenerative programs (especially those targeting the tissue/tumour ecosystem) that are hijacked during chronic inflammation.
- Create strategies to disrupt tumour-immune-stromal cross-regulatory interactions in order to prevent tumour progression into treatment-resistant, metastatic disease.
A Vision for the Future
The intersection of inflammation, chronic disease and cancer provides a unique opportunity to address some of the most pressing questions in oncology. By integrating next-generation single-cell and spatial biology tools with functional genomics/validation in preclinical models, we are not only uncovering the fundamental mechanisms driving cancer but also paving the way for innovative therapeutic approaches. Our laboratory remains committed to advancing this field, with the hope that our work will lead to earlier interventions and improved outcomes for patients battling cancer. Stay tuned as we continue to push the boundaries of cancer research and move closer to a future where precision medicine transforms the landscape of oncology.
Biography
Education and qualifications
1996-2002: PhD, Developmental Biology, Cancer Biology, University of Chicago, USA (Mentor: Dr Elaine Fuchs)
1994-1996: BA Genetics, University of Cambridge, UK
1991-1994: BSc (Hons) Chemistry, Delhi University, India
Appointments
2025-present: Professor and Senior Group Leader, Cancer Systems Biology Lab, CRUK Scotland Institute and the School of Cancer Sciences, University of Glasgow, UK
2018-2025: Senior Group Leader, Precision Oncology and Cancer Evolution; Spatial and Single Cell Platform (SSCP), Genome Institute of Singapore, A*STAR, Singapore
2014-2018: Group Leader, Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, A*STAR, Singapore
2014-2017: Research Associate Professor (Adjunct) of Biochemistry and Molecular Pharmacology, New York University School of Medicine/Cancer Institute, NY
2012-2017: Associate Professor of Biochemistry and Molecular Pharmacology, New York University School of Medicine/Cancer Institute, NY
2012-2016: Visiting Scientist at the Institute for Stem Cell Biology and Regenerative Medicine, Bangalore, India
2008-2014: Member, Helen and Martin Kimmel Center for Stem Cell Biology, New York University School of Medicine, NY
2006-2014: Scientific Director-RNAi Screening Facility, New York University School of Medicine, NY
2005-2012: Assistant Professor of Pharmacology, New York University School of Medicine/New York University Cancer Institute, NY
2002-2005: Postdoctoral Training (RNAi-based HTS/HCS, Wnt signalling, cancer genomics and drug discovery), Department of Genetics, Harvard Medical School, Boston, MA (Mentor: Dr Norbert Perrimon)
Other Appointments
2021-present: Founder and Scientific Advisor, NexiOgen Therapeutics, Singapore
2019-present: Adjunct Professor, Dept. of Pharmacology, Faculty of Medicine, Siriraj Hospital, Mahidol University, Bangkok, Thailand
2017-present: Associate Member, National Cancer Center Singapore (NCCS)
2015-present: Associate Member, Cancer Science Institute, National University of Singapore
2014-present: Scientific Advisor, Centre for High-throughput Phenomics (CHiP-GIS)
Honours and Awards
1994 Certificate of Merit from Delhi University for Academic Excellence, Delhi, India.
1995 Wellcome Summer Studentship, Wellcome-CRC Institute, Cambridge, UK.
1996 Summer Research Studentship, King's College, Cambridge, UK.
1994-1996 Cambridge Commonwealth Trust/Cambridge-Nehru Award, Cambridge, UK.
1996-2000 Markey Fellowship on Molecular Medicine. University of Chicago, Chicago IL.
2000-2001 Elaine-Ehrman Award, Student Fellowship, Committee-on-Cancer Biology, University of Chicago, IL.
2001 Harold Weintraub Graduate Student Award, Fred Hutchinson Cancer Center, Seattle, WA
2002 Best graduate thesis award, Division of Biological Sciences, University of Chicago, Chicago IL.
2003-2007 Breast Cancer Research Foundation fellowship from Department of Defense, USA.
2007-2008 Breast Cancer Research Foundation, Concept Award from Department of Defense, USA.
2009-2010 NYSTEM Idea Award from New York Stem Cell Science, New York, USA
2010-2013 March of Dimes Research Grant, USA
2011-2015 American Cancer Society, Research Scholar Grant, USA
2012-2014 NYC BioAccelerate Prize, New York, NY, USA
2019 Chen Award of Excellence by HUGO (Human Genome Organization)
Recent Publications
2024
Gan WL, Ren X, Ng VHE, Ng L, Song Y, Tano V, Han J, An O, Xie J, Ng BYL, Tay DJT, Tang SJ, Shen H, Khare S, Chong KHC, Young DY, Wu B, DasGupta R, Chen L. Hepatocyte-macrophage crosstalk via the PGRN-EGFR axis modulates ADAR1-mediated immunity in the liver. Cell Rep. 2024;43(7):114400.
Narmada BC, Khakpoor A, Shirgaonkar N, Narayanan S, Aw PPK, Singh M, Ong KH, Owino CO, Ng JWT, Yew HC, Binte Mohamed Nasir NS, Au VB, Sng R, Kaliaperumal N, Khine H, di Tocco FC, Masayuki O, Naikar S, Ng HX, Chia SL, Seah CXY, Alnawaz MH, Wai CLY, Tay AYL, Mangat KS, Chew V, Yu W, Connolly JE, Periyasamy G, Plissonnier ML, Levrero M, Lim SG, DasGupta R. Single-cell landscape of functionally cured chronic hepatitis B patients reveals activation of innate and altered CD4-CTL-driven adaptive immunity. J Hepatol. 2024;81(1):42-61.
Scolaro T, Manco M, Pecqueux M, Amorim R, Trotta R, Van Acker HH, Van Haele M, Shirgaonkar N, Naulaerts S, Daniluk J, Prenen F, Varamo C, Ponti D, Doglioni G, Ferreira Campos AM, Fernandez Garcia J, Radenkovic S, Rouhi P, Beatovic A, Wang L, Wang Y, Tzoumpa A, Antoranz A, Sargsian A, Di Matteo M, Berardi E, Goveia J, Ghesquière B, Roskams T, Soenen S, Voets T, Manshian B, Fendt SM, Carmeliet P, Garg AD, DasGupta R, Topal B, Mazzone M. Nucleotide metabolism in cancer cells fuels a UDP-driven macrophage cross-talk, promoting immunosuppression and immunotherapy resistance. Nat Cancer. 2024;5(8):1206-1226.
Shanmugam R, Majee P, Shi W, Ozturk MB, Vaiyapuri TS, Idzham K, Raju A, Shin SH, Fidan K, Low JL, Chua JYH, Kong YC, Qi OY, Tan E, Chok AY, Seow-En I, Wee I, Macalinao DC, Chong DQ, Chang HY, Lee F, Leow WQ, Murata-Hori M, Xiaoqian Z, Shumei C, Tan CSH, Dasgupta R, Tan IB, Tergaonkar V. Iron-(Fe3+)-Dependent Reactivation of Telomerase Drives Colorectal Cancers. Cancer Discov. 2024;14(10):1940-1963.
Singh V, Mondal A, Adhikary S, Mondal P, Shirgaonkar N, DasGupta R, Roy S, Das C. UBR7 E3 Ligase Suppresses Interferon-β Mediated Immune Signaling by Targeting Sp110 in Hepatitis B Virus-Induced Hepatocellular Carcinoma. ACS Infect Dis. 2024;10(11):3775-3796.
Tan DJH, Ng CH, Muthiah M, Yong JN, Chee D, Teng M, Wong ZY, Zeng RW, Chin YH, Wang JW, Danpanichkul P, Rajaram RB, DasGupta R, Suzuki H, Takahashi H, Tamaki N, Dan YY, Lui R, Duseja A, Siddiqui MS, Yeoh KG, Sanyal A, Wijarnpreecha K, Loomba R, Mantzoros CS, Huang DQ. Rising global burden of cancer attributable to high BMI from 2010 to 2019. Metabolism. 2024;152:155744.
Tostado CP, Da Ong LX, Heng JJW, Miccolis C, Chia S, Seow JJW, Toh YC, DasGupta R. An AI-assisted integrated, scalable, single-cell phenomic-transcriptomic platform to elucidate intratumor heterogeneity against immune response. Bioeng Transl Med. 2024;9(2):e10628.
Womersley H, Muliaditan D, DasGupta R, Cheow LF. Single-nucleus CUT&RUN elucidates the function of intrinsic and genomics-driven epigenetic heterogeneity in head and neck cancer progression. Genome Res. 2024.
Lab Members
Bioinformatician
Niranjan Shirgaonkar (single cell and spatial transcriptomics)
Postdoctoral Scientists
Shruti Khare (Bioinformatics; data analytics; single cell and spatial transcriptomics)
Gao Ran (single cell and spatial transcriptomics)
Mengwei Li (Bioinformatics; large scale single cell and spatial data integration)