Blog - Industry Trends ¬

Less is More: The Case for Low-Pass Long-Read Sequencing in Modern Genomics

By spanning large, complex genomic regions and preserving native epigenetic information, LRS enables accurate detection of structural variants and methylation patterns, even at coverage levels below 10x.

Precision at a Price: The Hidden Costs of High-Coverage Short Reads

For genomic and epigenomic research to scale, sequencing methods must balance accuracy, cost, and throughput. Short-read approaches that rely on high genomic coverage (≥30x) often achieve precision but at the cost of throughput and accessibility, limiting their utility in clinical diagnostics, large-scale studies, and real-time disease monitoring.

Low-pass long-read sequencing (LRS) offers a powerful alternative to traditional high-coverage short-read methods. By spanning large, complex genomic regions and preserving native epigenetic information, LRS enables accurate detection of structural variants and methylation patterns, even at coverage levels below 10x. This capability is accelerating insights in oncology, biomarker discovery, and personalized medicine, while significantly reducing sequencing costs and complexity. In this article, we explore how low-pass LRS delivers high-impact results with fewer reads, unlocking broader utility across both research and clinical applications.

The Scalability Problem with High-Coverage Short Reads

Short-read sequencing (SRS) depends on fragmenting, amplifying, and reassembling small DNA fragments (typically 150–300 bp), which introduces bias and limits resolution in complex genomic regions1,2. High GC content, repeat elements, and structural complexity reduce mapping accuracy, while the amplification process itself can distort true sequence composition3,4. As a result, high (≥30x) sequencing depth is often required to generate accurate consensus sequences with short reads, representing an average signal across many reads to correct for noise, errors, and artifacts. However, this level of depth comes at a cost: increased reagent use, longer turnaround times, and greater computational demands.

Adding methylation profiling to short-read workflows introduces even more complexity. Capturing methylation typically requires chemical conversion (e.g., bisulfite treatment) or enrichment steps, as well as separate library preparation and sequencing protocols5. These additions not only raise the cost and time requirements but also limit the ability to scale across larger cohorts or integrate epigenetic data into routine workflows. In sum, SRS struggles to deliver unbiased, cost-effective insights when applied to structurally complex or methylation-rich regions, especially when scalability is a core requirement.

LRS Preserves Native Sequence and Epigenetic Information

By sequencing long, unamplified strands of DNA or RNA, LRS avoids the pitfalls of PCR amplification6,7,8. This allows accurate resolution of structural variants and base modifications, including all major types of DNA methylation, directly from the native molecule9. In structurally complex genomic regions, this results in superior accuracy and deeper insights.LRS has been shown to identify large deletions, translocations, and inversions using hundreds, not millions, of reads10. In one WBL case study, nanopore-based LRS at 5x coverage identified over 6,000 differentially methylated CpG islands across two tissue types—capturing 2.4x more than traditional arrays¹¹. These results demonstrate that low-pass LRS can provide technical advantages and biological breadth and efficiency even at reduced coverage.

Lower Read Depth Enables Economic and Scalable Applications

Low-pass LRS reduces reagent usage and cycle counts, directly lowering costs while preserving high data quality12. Genome skimming, very low-pass sequencing, with nanopore sequencing at coverage levels below 0.05x has shown effective for global methylation profiling across vertebrate and plant species 13,14,15. These studies confirmed reproducible detection of conserved methylation patterns with minimal coverage, validating the approach as a fast and economic tool for epigenomic screening. In these studies, low-pass LRS produced comparable methylation results to more complex bisulfite-based methods, with a fraction of the passes required. This efficiency opens the door to larger cohort studies, rapid diagnostic workflows, and resource-conscious applications.

Oncology Applications: Tumor Monitoring and Molecular Profiling

Low-pass LRS is particularly well suited for oncology. In high-grade ovarian and prostate cancers, patient-specific large structural variants have been identified with just 1.5–5x genomic coverage15. These breakpoints were validated with PCR and used to track disease recurrence via circulating tumor DNA (ctDNA). In some cases, ctDNA monitoring with LRS detected tumor progression earlier than traditional markers like PSA (prostate-specific antigen), a commonly used blood-based biomarker16

Low-coverage LRS is also proving valuable in transcriptomic and epigenetic tumor profiling. In a recent study comparing shallow nanopore RNA-seq to traditional short-read transcriptome profiling across multiple tumor types, LRS achieved strong concordance despite using 36-fold fewer reads. It accurately recapitulated key features, including signaling pathway activity, kinase expression, tumor microenvironment composition, and gene fusions, that are often difficult to resolve with short-read approaches20.

.Additionally, low-pass LRS enabled epigenomic profiling of 189 tumors and 41 matched normal samples, revealing hypermethylation of the BRCA1 and RAD51C gene promoters, tumor suppressors and key regulators of DNA repair, in cancer types where this had not been previously observed22,23,24. These findings expand the potential use of PARP inhibitors and platinum-based therapies beyond their traditional indications. More broadly, they illustrate how low-pass LRS can uncover previously unrecognized molecular features that inform treatment decisions, reveal new therapeutic opportunities, and support more personalized cancer care. By surfacing clinically actionable insights with fewer inputs, lower cost, and faster turnaround, low-pass LRS is opening new doors in translational oncology.

Conclusion

Low-pass long-read sequencing offers a practical and scalable approach to genetic and epigenetic analysis. By delivering high-resolution insights with fewer sequencing reads, it enables efficient detection of structural variants and methylation patterns, even in complex regions of the genome.

As the technology matures, low-pass LRS is poised to play an increasingly important role in translational research, diagnostics, and precision oncology—unlocking clinically meaningful insights in oncology, diagnostics, and population-scale studies at a fraction of the cost and complexity of traditional methods

  1. de Koning APJ, Gu W, Castoe TA, Batzer MA, Pollock DD. Repetitive elements may comprise over Two-Thirds of the human genome. PLoS Genet. 2011;7(12). doi:10.1371/journal.pgen.1002384
  2. Treangen TJ, Salzberg SL. Repetitive DNA and next-generation sequencing: Computational challenges and solutions. Nat Rev Genet. 2012;13(1):36-46. doi:10.1038/nrg3117
  3. Laszlo AH, Derrington IM, Brinkerhoff H, et al. Detection and mapping of 5-methylcytosine and 5-hydroxymethylcytosine with nanopore MspA. Proc Natl Acad Sci U S A. 2013;110(47):18904-18909. doi:10.1073/pnas.1310240110
  4. Schadt EE, Turner S, Kasarskis A. A window into third-generation sequencing. Hum Mol Genet. 2010;19(R2). doi:10.1093/hmg/ddq416
  5. Xu L, Seki M. Recent advances in the detection of base modifications using the Nanopore sequencer. J Hum Genet. 2020;65(1):25-33. doi:10.1038/s10038-019-0679-0
  6. Jain M, Olsen HE, Paten B, Akeson M. The Oxford Nanopore MinION: delivery of nanopore sequencing to the genomics community. Genome Biol. 2016;17(1). doi:10.1186/s13059-016-1103-0
  7. Wang Y, Zhao Y, Bollas A, Wang Y, Au KF. Nanopore sequencing technology, bioinformatics and applications. Nat Biotechnol. 2021;39(11):1348-1365. doi:10.1038/s41587-021-01108-x
  8. van Dijk EL, Jaszczyszyn Y, Naquin D, Thermes C. The Third Revolution in Sequencing Technology. Trends in Genetics. 2018;34(9):666-681. doi:10.1016/j.tig.2018.05.008
  9. Wescoe ZL, Schreiber J, Akeson M. Nanopores discriminate among five C5-cytosine variants in DNA. J Am Chem Soc. 2014;136(47):16582-16587. doi:10.1021/ja508527b
  10. Norris AL, Workman RE, Fan Y, Eshleman JR, Timp W. Nanopore sequencing detects structural variants in cancer. Cancer Biol Ther. 2016;17(3):246-253. doi:10.1080/15384047.2016.1139236
  11. DIRECT WHOLE METHYLOME SEQUENCING: WASATCH BIOLABS’ ARRAY REPLACEMENT Case Study. Accessed May 11, 2025. https://cdn.prod.website-files.com/66a0075ec4b0869dd3f73db6/67f6eedbc3b34c8fd249b36d_dWMS%20Case%20Study_v2.pdf
  12. Wasik K, Berisa T, Pickrell JK, et al. Comparing low-pass sequencing and genotyping for trait mapping in pharmacogenetics. BMC Genomics. 2021;22(1). doi:10.1186/s12864-021-07508-2
  13. Papaiakovou M, Fraija-Fernández N, James K, et al. Evaluation of genome skimming to detect and characterise human and livestock helminths. Int J Parasitol. 2023;53(2):69-79. doi:10.1016/j.ijpara.2022.12.002
  14. Faulk C. Genome Skimming with Nanopore Sequencing Precisely Determines Global and Transposon DNA Methylation in Vertebrates. Published online January 25, 2023. doi:10.1101/2023.01.25.525540
  15. Liau Y, Whibley A, Hill AM, et al. Low-pass nanopore sequencing for measurement of global methylation levels in plants. BMC Genomics. 2024;25(1):1235. doi:10.1186/s12864-024-11145-w
  16. Valle-Inclan JE, Stangl C, de Jong AC, et al. Optimizing Nanopore sequencing-based detection of structural variants enables individualized circulating tumor DNA-based disease monitoring in cancer patients. Genome Med. 2021;13(1). doi:10.1186/s13073-021-00899-7
  17. Rodon J, Soria JC, Berger R, et al. Genomic and transcriptomic profiling expands precision cancer medicine: the WINTHER trial. Nat Med. 2019;25(5):751-758. doi:10.1038/s41591-019-0424-4
  18. Ben-Hamo R, Jacob Berger A, Gavert N, et al. Predicting and affecting response to cancer therapy based on pathway-level biomarkers. Nat Commun. 2020;11(1). doi:10.1038/s41467-020-17090-y
  19. Wang X, Sun Z, Zimmermann MT, Bugrim A, Kocher JP. Predict drug sensitivity of cancer cells with pathway activity inference. BMC Med Genomics. 2019;12. doi:10.1186/s12920-018-0449-4
  20. Treangen TJ, Salzberg SL. Repetitive DNA and next-generation sequencing: Computational challenges and solutions. Nat Rev Genet. 2012;13(1):36-46. doi:10.1038/nrg3117
  21. Mock A, Braun M, Scholl C, Fröhling S, Erkut C. Transcriptome profiling for precision cancer medicine using shallow nanopore cDNA sequencing. Sci Rep. 2023;13(1). doi:10.1038/s41598-023-29550-8
  22. O’Neill K, Pleasance E, Fan J, et al. Long-read sequencing of an advanced cancer cohort resolves rearrangements, unravels haplotypes, and reveals methylation landscapes. Cell Genomics. 2024;4(11):100674. doi:10.1016/j.xgen.2024.100674
  23. Zhao EY, Shen Y, Pleasance E, et al. Homologous recombination deficiency and platinum-based therapy outcomes in advanced breast cancer. Clinical Cancer Research. 2017;23(24):7521-7530. doi:10.1158/1078-0432.CCR-17-1941
  24. O’Malley DM, Krivak TC, Kabil N, Munley J, Moore KN. PARP Inhibitors in Ovarian Cancer: A Review. Target Oncol. 2023;18(4):471-503. doi:10.1007/s11523-023-00970-w

Related Posts

Shedding Light on the Dark Genome with Long-Reads

NGS Liquid Biopsy Technologies: Transforming Clinical Diagnostics