Metabolon Logo
Metabolon Logo

Guide to Multiomics

Chapter 8 — The Future of Multiomics

This chapter explores the future of multiomics research. We highlight the ways in which multiomics will advance human medicine into the precision medicine era and explore two enabling, emerging technologies: single-cell and spatial omics.

In this guide, we’ve explored the incredible potential of multiomics analysis to deepen our understanding of organisms, ecosystems, and health and disease by providing a holistic view single-omics studies cannot achieve. As technology advances, multiomics research will become increasingly transformative across various domains from basic biological research to personalized and precision medicine.

Although the applications of multiomics research span a broad range from human healthcare to agricultural practices, some of the most transformative applications lie in the future of medicine. In fact, some of the biggest modern buzzwords, such as “precision medicine” and “personalized medicine” depend on multiomics research. In the following sections, we discuss how multiomics can drive the next revolution in human medicine.

Multiomics and the Future of Medicine

Genomics, transcriptomics, metabolomics, and other omics modalities, when applied individually, have revealed the enormous complexity of human health and disease1. A wide array of enabling technologies (Figure 1)2 have introduced this transformation in our understanding of human biology—and have opened the door to detecting and treating disease more accurately and precisely than ever.

Multi-omics of gut microbiome-host interactions in short- and l

Figure 1. Timeline of omics technological developments and milestones, which underpin the future of precision and personalized medicine2.

The biological complexity uncovered by individual omics modalities not only underpins the push for multiomics analyses but is also the underlying concept of precision medicine and personalized medicine.

Personalized and precision medicine

Personalized medicine and precision medicine are closely related and have been used interchangeably3. While both take the approach of deeply characterizing individuals at the molecular level, their applications are slightly different.

Precision medicine aims to predict, prevent, and cure disease more precisely1 and can be used to predict disease risk or the best treatments for specific groups of patients based on omics data collected from groups of individuals with the same biological and clinical characteristics3. Precision medicine drives and enables personalized medicine4, which considers factors specific to an individual, including their environment, when considering how to treat their specific disease3.

Multiomics analyses have made considerable contributions in the precision and personalized medicine spaces already. For example, researchers have found correlations in a cohort of Egyptian people between certain genetic variations and serum vitamin D levels, which can serve as a risk factor for myocardial infarction5. In oncology research, multiomics analyses have been used to identify cancer subgroups with distinct survival rates6, to link gene copy-number variations to post-translational modifications and clinical outcomes7, and many other significant discoveries2.

These are just a few examples of the advanced, precise, and personalized discoveries enabled by multiomics analysis. As technology advances and computational methods for analyzing multiomics data improve (see Chapter 2 of this guide), multiomics analyses will continue to drive the future of precision healthcare.

Disease diagnostics and prognostics

Biomarker detection is one of the most promising applications of multiomics analysis, as discussed in Chapter 1 of this guide. With accurate, strong biomarkers, researchers can develop diagnostic and prognostic approaches8 to identify better therapeutic targets for improved treatment (see next section).

There has been significant research using multiomics approaches to identify diagnostic and prognostic biomarkers in oncology. Researchers have identified diagnostic metabolic biomarkers in pancreatic ductal carcinoma9 and genomic, transcriptomic, proteomic, and metabolic biomarkers for early ovarian cancer diagnosis10.

Diagnostic and prognostic biomarkers for other diseases have also been identified, including stroke11, obesity and its comorbidities12, and even COVID-1913. Additionally, researchers utilized a multiomics approach to diagnose rare diseases and inform ongoing medical care changes in a cohort of over 200 infants14.

Drug discovery and development

The natural follow-up to disease diagnosis is treatment. As with diagnosis and prognosis, drug discovery and development can be informed and aided by multiomics analysis. By revealing the molecular mechanisms behind drug responses, researchers can identify new therapeutic targets, which can be genes, transcription factors, metabolites or metabolic pathways, or other molecular signatures. For example, researchers identified two H3 histone modifications, H3K27ac and H3K9ac, as potential therapeutic targets in Alzheimer’s disease15. In meningiomas, a type of benign brain tumor, the molecular cross-talk between ubiquitin ligase TRAF7 and transcription factor KLF4 was identified as a potential target for the development of novel treatments16. In hypertrophic cardiomyopathy, altered energy metabolism and mitochondrial function suggest that therapies developed to improve metabolic function and reduce mitochondrial injury may help decrease disease17.

Enabling Technologies for the Future of Multiomics

Many of the multiomic discoveries driving precision and personalized medicine, disease diagnosis, and drug discovery are likely to depend on two enabling technologies: single-cell and spatial omics. These approaches consider the cellular and tissue-specific complexities of disease, placing biological discoveries in context at the cellular, tissue, and organismal level, enabling a more precise and accurate understanding of disease.

Single-cell omics

Single-cell analyses18 enable researchers to study cells at the finest resolution possible, enabling the discoveries of rare cell types involved in disease or which cells a certain therapeutic is effective against (or not), for example. Single cell DNA and RNA sequencing were named “2013 Method of the Year” by Nature19 and since then have made important contributions to our understanding of biology and various disease mechanisms20.

Recently, researchers used single-cell analysis to identify a new type of cell called a SWAT cell21, which shares a progenitor with adipocytes—suggesting that toggling the balance between different cellular developmental states could inform precise new therapeutic targets for cardiometabolic disease.

Single-cell analyses are also the foundation for comprehensive maps such as the Human Cell Atlas22 and Tabula Sapiens23, providing foundational reference points of human biology. By comparing new datasets to these baselines, researchers can better understand disease origins and identify biologically and clinically significant alterations (see Figure 2).

Multi-omics of gut microbiome-host interactions in short- and l

Figure 2.Single-cell analyses help build cell atlases used to draw important insights on disease mechanisms and identify new treatments. Multiomics analyses will help address some of the technical challenges facing the broad clinical application of these insights22.

Spatial omics

Just as single omics techniques are unable to provide a complete picture of a biological mechanism, single-cell analyses are necessarily limited. Spatial analyses put cells in the context of one another, providing a more holistic view of health and disease processes. Spatial transcriptomics, for example, has already provided key clues to tumor microenvironment-specific characteristics that affect treatment responses24-25. It has also identified and spatially located a subtype of dopaminergic neurons that are highly susceptible to degeneration and enriched for heritable risk in Parkinson’s disease26.

Concluding Remarks

Multiomics research represents a paradigm shift in our approach to understanding biology and the molecular changes that occur when things go wrong. It offers unprecedented opportunities to not only understand biology at its most basic level, but to more completely understand disease, enabling a future where more effective, safer, and tailored treatments for a wide range of diseases are available.

References

  1. Hasin Y, Seldin M, and Lusis A. Multi-omics approaches to disease. Genome Biol. 2017;18:83.
  2. Babu M and Snyder M. Multi-Omics Profiling for Health. Mol Cell Proteomics. 2023; 22(6):100561. doi: 10.1016/j.mcpro.2023.100561
  3. Delpierre C and Lefèvre T. Precision and personalized medicine: What their current definition says and silences about the model of health they promote. Implication for the development of personalized health. Front Sociol. 2023; 8:1112159. doi: 10.3389/fsoc.2023.1112159
  4. Ahmed Z. Chapter Four – Precision medicine with multi-omics strategies, deep phenotyping, and predictive analysis. In: Teplow DB, ed. Progress in Molecular Biology and Translational Science. Academic Press. 2022;190(1):101-125. 10.1016/bs.pmbts.2022.02.002
  5. Sedky NK, Abdel Rahman MF, Hassanein SI, et al. Genetic variants of CYP2R1 are key regulators of serum vitamin D levels and incidence of myocardial infarction in middle-aged Egyptians. Curr Pharm Biotechnol. 2018;19(3):265–73.
  6. Liu G., Dong C., Liu L. Integrated multiple “-omics” data reveal subtypes of hepatocellular carcinoma. PLoS One. 2016;11
  7. Zhang H., Liu T., Zhang Z., Payne S.H., Zhang B., McDermott J.E., et al. Integrated proteogenomic characterization of human high-grade serous ovarian cancer. Cell. 2016;166:755–765.
  8. Biomarkers Definitions Working Group . Biomarkers and surrogate endpoints: preferred definitions and conceptual framework. Clin Pharmacol Ther. 2001;69(3):89‐95.
  9. Mayerle J, Kalthoff H, Reszka R, et al. Metabolic biomarker signature to differentiate pancreatic ductal adenocarcinoma from chronic pancreatitis. Gut. 2018;67(1):128-137. doi: 10.1136/gutjnl-2016-312432
  10. Xiao Y, Bi M, Guo H, et al. Multi-omics approaches for biomarker discovery in early ovarian cancer diagnosis. BioMedicine. 2022;79:104001. doi: 10.1016/j.ebiom.2022.104001
  11. Montaner J, Ramiro L, Simats A, et al. Multilevel omics for the discovery of biomarkers and therapeutic targets for stroke. Nat Rev Neurol. 2020;16(5):247-264. doi: 10.1038/s41582-020-0350-6
  12. Aleksandrova K, Rodrigues CE, Floegel A, et al. Omics Biomarkers in Obesity: Novel Etiological Insights and Targets for Precision Prevention. Curr Obes Rep. 2020;9(3):219-230. doi: 10.1007/s13679-020-00393-y
  13. Byeon SK, Madugundu AK, Garapati K, et al. Development of a multiomics model for identification of predictive biomarkers for COVID-19 severity: a retrospective cohort study. Lancet Digit Health. 2022; 4(9): e632–e645. doi: 10.1016/S2589-7500(22)00112-1
  14. Lunke S, Bouffler SE, Patel CV, et al. Integrated multi-omics for rapid rare disease diagnosis on a national scale. Nat Med. 2023;29(7):1681-1691. doi: 10.1038/s41591-023-02401-9
  15. Nativio R, Lan Y, Donahue G, et al. An integrated multi-omics approach identifies epigenetic alterations associated with Alzheimer’s disease. Nat Genet. 2020;52(10):1024-1035. doi: 10.1038/s41588-020-0696-0
  16. Najm P, Zhao P, Steklov M, et al. Loss-of-Function Mutations in TRAF7 and KLF4 Cooperatively Activate RAS-Like GTPase Signaling and Promote Meningioma Development. Cancer Res. 2021;81(16):4218-4229. doi: 10.1158/0008-5472.CAN-20-3669
  17. Ranjbarvaziri S, Kookier KB, Ellenberger M, et al. Altered Cardiac Energetics and Mitochondrial Dysfunction in Hypertrophic Cardiomyopathy. Circulation. 2021;144(21):1714-1731. doi: 10.1161/CIRCULATIONAHA.121.053575
  18. Lähnemann D, Köster J, Szczurek E, et al. Eleven grand challenges in single-cell data science. Genome Biol. 2020;21(1):31. doi: 10.1186/s13059-020-1926-6
  19. Method of the Year 2013. Nat Methods. 2014;11(1):1. doi: 10.1038/nmeth.2801
  20. Wang S, Sun S-T, Zhang X-Y, et al. The Evolution of Single-Cell RNA Sequencing Technology and Application: Progress and Perspectives.Int J Mol Sci. 2023;24(3):2943. doi: 10.3390/ijms24032943
  21. Palani NP, Horvath C, Timshel PN, et al. Adipogenic and SWAT cells separate from a common progenitor in human brown and white adipose depots. Nat Metab. 2023;5:996-1013.
  22. Rood JE, Maartens A, Hupalowska A, et al. Impact of the Human Cell Atlas on medicine. Nat Med. 2022;28:2486-2496.
  23. The Tabula Sapiens Consortium. The Tabula Sapiens: A multiple-organ, single-cell transcriptomic atlas of humans. Science. 2022;376(6594):eabl4896. doi: 10.1126/science.abl4896
  24. Larroquette M, Guegan J-P, Besse B, et al. Spatial transcriptomics of macrophage infiltration in non-small cell lung cancer reveals determinants of sensitivity and resistance to anti-PD1/PD-L1 antibodies. J Immunother Cancer. 2022;10(5):e003890. doi: 10.1136/jitc-2021-003890
  25. Peng Z, Ye M, Ding H, et al. Spatial transcriptomics atlas reveals the crosstalk between cancer-associated fibroblasts and tumor microenvironment components in colorectal cancer. J Transl Med. 2022;20(1):302. doi: 10.1186/s12967-022-03510-8
  26. Kamath T, Abdulraouf A, Burris SJ, et al. Single-cell genomic profiling of human dopamine neurons identifies a population that selectively degenerates in Parkinson’s disease. Nat Neurosci. 2022;25(5):588-595. doi: 10.1038/s41593-022-01061-1

See how Metabolon can advance your path to preclinical and clinical insights

Contact Us

Talk with an expert

Request a quote for our services, get more information on sample types and handling procedures, request a letter of support, or submit a question about how metabolomics can advance your research.

Corporate Headquarters

617 Davis Drive, Suite 100
Morrisville, NC 27560

+1 (919) 572-1721