Skip to main content

EDITORIAL article

Front. Immunol., 22 May 2023
Sec. Cancer Immunity and Immunotherapy
This article is part of the Research Topic Translational Phenomics and its Applications in Immunotherapy View all 7 articles

Editorial: Translational phenomics and its applications in immunotherapy

  • 1Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
  • 2Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford, United Kingdom
  • 3Hood-Price Lab, Institute for Systems Biology, Seattle, WA, United States

Immunotherapy has emerged as a promising treatment option for a variety of diseases, including cancer and immune-mediated disorders. Its success depends on identifying the right target, selecting the right patient population, monitoring host immune responses, and managing adverse effects. Coined by Steven Garan (1), the term “phenomics” (2) refers to all measurable micro- and macro-phenotypic manifestations beyond an individual’s genome, including cellular senescence (3), the tumor microenviroment (4), metabolome (5), and gut microbiome (6), to name but a few. Deep phenotyping of the patient immune system and diseased tissues is essential to better understand disease pathogenesis, identify key diagnostic biomarkers, and develop rational therapeutics with favorable clinical outcomes (79). The Frontiers in Immunology Research Topic “Translational Phenomics and its Applications in Immunotherapy” has published cutting-edge articles that use systems thinking and phenomics strategy (8, 10), along with omics and 3D printing technologies, to address significant challenges in immunotherapy. This Editorial summarizes six original articles included in this Research Topic, which can be grouped under three umbrellas, namely, “cellular senescence and cancer immunotherapy”, “rational treatments and diagnostic markers”, and “omics strategies and 3D printing technologies”.

Cellular senescence and cancer immunotherapy

Unlocking the power of cellular senescence-related genes: prognostic and therapeutic significance in clear cell renal cell carcinoma

This study conducted a comprehensive analysis of the significance of cellular senescence-related genes in ccRCC and their impact on patient survival and treatment response (Lu et al.). By leveraging public databases, the authors identified three molecular subtypes of ccRCC with varying clinical manifestations, mutation patterns, and tumor microenvironments. They also developed a risk model based on five cellular senescence-related genes that accurately predicted patient response to immunotherapy and chemotherapy. These findings provide a theoretical foundation for further exploration of the molecular mechanisms underlying cellular senescence-related ccRCC, with the potential of improving the risk stratification of patients. These results have important implications for the development of personalised treatment strategies for ccRCC patients.

A cell senescence-related lncRNA signature predicts prognosis and immune response in colorectal cancer

This study focused on identifying lncRNAs linked to cell senescence and their correlations with prognosis and immune response in CRC (Xu et al.). By analysing data from TCGA and Human Aging Genomic Resources, the authors identified a signature of six cell senescence-related lncRNAs that demonstrated high prognostic value. The risk model was found to be also associated with tumor microenvironments, immune checkpoints, and drug sensitivity, with lower IC50 values observed for AICAR, cisplatin, nilotinib, and bexarotene in the high-risk group. These findings suggest that the identified cell senescence-related lncRNAs may serve as valuable biomarkers for predicting prognosis and response to immune and chemotherapy in CRC patients.

Rational treatments and diagnostic markers

Subgroup analysis predicts recurrence risk and personalises treatment for stage II CRC

This study developed a novel method called “TFunctionalProg” to predict the risk of recurrence and personalised adjuvant drugs for stage II CRC patients (Wang et al.). By analysing data from 222 microsatellite-stable stage II CRC patients, the authors identified two subgroups with high risk of recurrence, each exhibiting distinct phenotypes and mechanisms. Based on these findings, they proposed personalised rational adjuvant drug combinations, including immunotherapy, chemotherapy, and repurposed central nervous system drugs. This approach provides a different utility compared to circulating tumor DNA-based prognostic biomarkers, with the potential of improving the survival rate of stage II CRC patients.

Unraveling the intricate relationship between altered gut microbiota and inflammation markers in patients with Crohn’s disease

This study investigated the correlation between altered gut microbiota and elevated inflammation markers in CD patients, using paired subjects to minimise confounding factors (Hu et al.). By analysing fecal microbiota, the authors found that CD patients had reduced levels of short-chain fatty acid producing bacteria but elevated levels of opportunistic pathogen Escherichia-Shigella. These differential genera had the potential to distinguish between CD patients and healthy controls, and showed correlations with changes in inflammation-related blood biochemical markers. The results suggest that the differential genera could be used as diagnostic markers for CD, and as potential targets for intervention, thereby paving the way for a more precise and effective treatment of the disease.

Omics strategies and 3D printing technologies

Prognostic signature for survival, immune microenvironment, and immunotherapy response prediction using single-cell sequencing data in patients with gastric cancers

In this study, the authors developed an immune-related genetic signature using scRNA-seq data combined with bulk RNA-seq data to predict prognosis, immune status, and response to immunotherapy in GC patients (Hu et al.). Their analysis identified nine cell types and established a signature that accurately predicted overall survival and improved clinical utility. The low-risk group had high tumor mutation burden, increased immune activation, and high microsatellite instability, indicating the suitability for immunotherapy. On the other hand, the high-risk group was associated with microsatellite stability and immunosuppression, suggesting the suitability for targeted therapy. The accuracy of the signature in predicting immunotherapeutic response was validated in an external dataset. These findings provide insight into the immune status of GC patients, prognosis assessment, and the development of efficient immunotherapeutic treatments, as well as personalised treatment strategies in the disease.

3D printed Ti-5Cu alloy induces M2 macrophage polarisation and accelerates osteogenic differentiation of MC3T3-E1 cells

This study investigated the effects of the Ti-5Cu alloy, prepared through 3D printing technologies, on macrophage polarisation and immune-related bone formation (Zhao et al.). The authors found that the alloy had a smoother surface compared to Ti, with no cytotoxicity observed to either RAW264.7 cells or Cu2+ at a concentration of 0.133 mg/L. The results also showed that the alloy was able to modulate macrophage polarisation towards the M2 phenotype, promoting the proliferation and osteogenic differentiation of MC3T3-E1 cells. The gene “Oncostatin M” expressed by RAW264.7 co-cultured with the Ti-5Cu alloy was found to accelerate the osteogenic differentiation of MC3T3-E1 cells through acting on its receptors. These findings highlight the potential of the Ti-5Cu alloy for accelerating osteogenic differentiation and immune-related bone formation.

Author contributions

All authors listed have made a substantial, direct, and intellectual contribution to the work and approved it for publication.

Funding

This work is funded by National Natural Science Foundation of China (32170663 to HF and 82272958 to QT) and Innovative Research Team of High-Level Local Universities in Shanghai.

Acknowledgments

We would like to thank all the authors who have participated in this Research Topic and the reviewers for their insightful comments.

Conflict of interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Jin L. Welcome to the phenomics journal. Phenomics (2021) 1:1–2. doi: 10.1007/s43657-020-00009-4

PubMed Abstract | CrossRef Full Text | Google Scholar

2. Houle D, Govindaraju DR, Omholt S. Phenomics: the next challenge. Nat Rev Genet (2010) 11:855–66. doi: 10.1038/nrg2897

PubMed Abstract | CrossRef Full Text | Google Scholar

3. Kirkland JL. Cellular senescence: the good, the bad and the unknown. Nat Rev Nephrol (2022) 18:611–27. doi: 10.1038/s41581-022-00601-z

PubMed Abstract | CrossRef Full Text | Google Scholar

4. Hu B, Sajid M, Lv R, Liu L, Sun C. A review of spatial profiling technologies for characterizing the tumor microenvironment in immuno-oncology. Front Immunol (2022) 13:996721. doi: 10.3389/fimmu.2022.996721

PubMed Abstract | CrossRef Full Text | Google Scholar

5. Zhang Z, Bao C, Jiang L, Wang S, Wang K, Lu C, et al. When cancer drug resistance meets metabolomics (bulk, single-cell and/or spatial): progress, potential, and perspective. Front Oncol (2023) 12:1054233. doi: 10.3389/fonc.2022.1054233

PubMed Abstract | CrossRef Full Text | Google Scholar

6. Nicholson JK, Holmes E, Wilson ID. Gut microorganisms, mammalian metabolism and personalized health care. Nat Rev Microbiol (2005) 3:431–8. doi: 10.1038/nrmicro1152

PubMed Abstract | CrossRef Full Text | Google Scholar

7. Bycroft C, Freeman C, Petkova D, Band G, Elliott LT, Sharp K, et al. The UK biobank resource with deep phenotyping and genomic data. Nature (2018) 562:203–9. doi: 10.1038/s41586-018-0579-z

PubMed Abstract | CrossRef Full Text | Google Scholar

8. Yurkovich JT, Tian Q, Price ND, Hood L. A systems approach to clinical oncology uses deep phenotyping to deliver personalized care. Nat Rev Clin Oncol (2020) 17:183–94. doi: 10.1038/s41571-019-0273-6

PubMed Abstract | CrossRef Full Text | Google Scholar

9. Fang H, Chen L, Knight JC. From genome-wide association studies to rational drug target prioritisation in inflammatory arthritis. Lancet Rheumatol (2020) 2:e50–62. doi: 10.1016/S2665-9913(19)30134-1

CrossRef Full Text | Google Scholar

10. Hood L, Tian Q. Systems approaches to biology and disease enable translational systems medicine. Genomics Proteomics Bioinforma (2012) 10:181–5. doi: 10.1016/j.gpb.2012.08.004

CrossRef Full Text | Google Scholar

Keywords: phenomics, immunotherapy, systems biology, deep phenotyping, systems immunology, translational medicine

Citation: Fang H, Chen L, Hood L and Tian Q (2023) Editorial: Translational phenomics and its applications in immunotherapy. Front. Immunol. 14:1211704. doi: 10.3389/fimmu.2023.1211704

Received: 25 April 2023; Accepted: 15 May 2023;
Published: 22 May 2023.

Edited and Reviewed by:

Sven Geissler, Center for Regenerative Therapies (BCRT), Germany

Copyright © 2023 Fang, Chen, Hood and Tian. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Hai Fang, fh12355@rjh.com.cn; Leroy Hood, lhood@systemsbiology.org; Qiang Tian, tq12221@rjh.com.cn

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.