Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Opinion
  • Published:

Safe harbours for the integration of new DNA in the human genome

Abstract

Interactions between newly integrate DNA and the host genome limit the reliability and safety of transgene integration for therapeutic cell engineering and other applications. Although targeted gene delivery has made considerable progress, the question of where to insert foreign sequences in the human genome to maximize safety and efficacy has received little attention. In this Opinion article, we discuss 'genomic safe harbours' — chromosomal locations where therapeutic transgenes can integrate and function in a predictable manner without perturbing endogenous gene activity and promoting cancer.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Intragenic candidate GSHs.
Figure 2: Prospective extragenic GSHs in human iPS cell clones harbouring single-copy globin transgenes.

Similar content being viewed by others

References

  1. Cartier, N. et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 326, 818–823 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Aiuti, A. et al. Multilineage hematopoietic reconstitution without clonal selection in ADA-SCID patients treated with stem cell gene therapy. J. Clin. Invest. 117, 2233–2240 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Gaspar, H. B. et al. Hematopoietic stem cell gene therapy for adenosine deaminase-deficient severe combined immunodeficiency leads to long-term immunological recovery and metabolic correction. Sci. Transl. Med. 3, 97ra80 (2011).

    PubMed  Google Scholar 

  4. Riviere, I., Dunbar, C. & Sadelain, M. Hematopoietic stem cell engineering at a crossroads. Blood 17 Nov 2011 (doi:10.1182/blood-2011-09-349993).

  5. Martin, D. I. & Whitelaw, E. The vagaries of variegating transgenes. Bioessays 18, 919–923 (1996).

    Article  CAS  PubMed  Google Scholar 

  6. Kioussis, D. & Festenstein, R. Locus control regions: overcoming heterochromatin-induced gene inactivation in mammals. Curr. Opin. Genet. Dev. 7, 614–619 (1997).

    Article  CAS  PubMed  Google Scholar 

  7. Rivella, S. & Sadelain, M. Genetic treatment of severe hemoglobinopathies: the combat against transgene variegation and transgene silencing. Semin. Hematol. 35, 112–125 (1998).

    CAS  PubMed  Google Scholar 

  8. Bestor, T. H. Gene silencing as a threat to the success of gene therapy. J. Clin. Invest. 105, 409–411 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ellis, J. Silencing and variegation of gammaretrovirus and lentivirus vectors. Hum. Gene Ther. 16, 1241–1246 (2005).

    Article  CAS  PubMed  Google Scholar 

  10. Karpen, G. H. Position-effect variegation and the new biology of heterochromatin. Curr. Opin. Genet. Dev. 4, 281–291 (1994).

    Article  CAS  PubMed  Google Scholar 

  11. Weiler, K. S. & Wakimoto, B. T. Heterochromatin and gene expression in Drosophila. Annu. Rev. Genet. 29, 577–605 (1995).

    Article  CAS  PubMed  Google Scholar 

  12. Nusse, R., van Ooyen, A., Cox, D., Fung, Y. K. & Varmus, H. Mode of proviral activation of a putative mammary oncogene (int-1) on mouse chromosome 15. Nature 307, 131–136 (1984).

    Article  CAS  PubMed  Google Scholar 

  13. Schroder, A. R. et al. HIV-1 integration in the human genome favors active genes and local hotspots. Cell 110, 521–529 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Wu, X., Li, Y., Crise, B. & Burgess, S. M. Transcription start regions in the human genome are favored targets for MLV integration. Science 300, 1749–1751 (2003).

    Article  CAS  PubMed  Google Scholar 

  15. Mitchell, R. S. et al. Retroviral DNA integration: ASLV, HIV, and MLV show distinct target site preferences. PLoS Biol. 2, E234 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. HaceinBeyAbina, S. et al. LMO2-associated clonal T cell proliferation in two patients after gene therapy for SCID-X1. Science 302, 415–419 (2003).

    Article  CAS  Google Scholar 

  17. Ott, M. G. et al. Correction of X-linked chronic granulomatous disease by gene therapy, augmented by insertional activation of MDS1-EVI1, PRDM16 or SETBP1. Nature Med. 12, 401–409 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Stein, S. et al. Genomic instability and myelodysplasia with monosomy 7 consequent to EVI1 activation after gene therapy for chronic granulomatous disease. Nature Med. 16, 198–204 (2010).

    Article  CAS  PubMed  Google Scholar 

  19. Howe, S. J. et al. Insertional mutagenesis combined with acquired somatic mutations causes leukemogenesis following gene therapy of SCID-X1 patients. J. Clin. Invest. 118, 3143–3150 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Cavazzana-Calvo, M. et al. Transfusion independence and HMGA2 activation after gene therapy of human beta-thalassaemia. Nature 467, 318–322 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kustikova, O. S. et al. Dose finding with retroviral vectors: correlation of retroviral vector copy numbers in single cells with gene transfer efficiency in a cell population. Blood 102, 3934–3937 (2003).

    Article  CAS  PubMed  Google Scholar 

  22. Modlich, U. et al. Leukemias following retroviral transfer of multidrug resistance 1 (MDR1) are driven by combinatorial insertional mutagenesis. Blood 105, 4235–4246 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Chang, A. H. & Sadelain, M. The genetic engineering of hematopoietic stem cells: the rise of lentiviral vectors, the conundrum of the ltr, and the promise of lineage-restricted vectors. Mol. Ther. 15, 445–456 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Gijsbers, R. et al. LEDGF hybrids efficiently retarget lentiviral integration into heterochromatin. Mol. Ther. 18, 552–560 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Modlich, U. et al. Cell-culture assays reveal the importance of retroviral vector design for insertional genotoxicity. Blood 108, 2545–2553 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Montini, E. et al. The genotoxic potential of retroviral vectors is strongly modulated by vector design and integration site selection in a mouse model of HSC gene therapy. J. Clin. Invest. 119, 964–975 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Emery, D. W. The use of chromatin insulators to improve the expression and safety of integrating gene transfer vectors. Hum. Gene Ther. 22, 761–774 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Persons, D. A. & Baum, C. Solving the problem of gamma-retroviral vectors containing long terminal repeats. Mol. Ther. 19, 229–231 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Capecchi, M. R. Gene targeting in mice: functional analysis of the mammalian genome for the twenty-first century. Nature Rev. Genet. 6, 507–512 (2005).

    Article  CAS  PubMed  Google Scholar 

  30. Moynahan, M. E. & Jasin, M. Mitotic homologous recombination maintains genomic stability and suppresses tumorigenesis. Nature Rev. Mol. Cell Biol. 11, 196–207 (2010).

    Article  CAS  Google Scholar 

  31. Jasin, M. Genetic manipulation of genomes with rare-cutting endonucleases. Trends Genet. 12, 224–228 (1996).

    Article  CAS  PubMed  Google Scholar 

  32. Porteus, M. H. & Carroll, D. Gene targeting using zinc finger nucleases. Nature Biotechnol. 23, 967–973 (2005).

    Article  CAS  Google Scholar 

  33. Paques, F. & Duchateau, P. Meganucleases and DNA double-strand break-induced recombination: perspectives for gene therapy. Curr. Gene Ther. 7, 49–66 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Boch, J. TALEs of genome targeting. Nature Biotechnol. 29, 135–136 (2011).

    Article  CAS  Google Scholar 

  35. Rosenberg, N., Jolicouer, P., Coffin, J. M., Hughes, S. H. & Varmus, H. E. in Retroviruses 475–586 (Cold Spring Harbor Laboratory Press, Cold Spring Harbor, USA, 1997).

    Google Scholar 

  36. Akagi, K., Suzuki, T., Stephens, R. M., Jenkins, N. A. & Copeland, N. G. RTCGD: retroviral tagged cancer gene database. Nucleic Acids Res. 32, D523–D527 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kim, R. et al. Genome-based identification of cancer genes by proviral tagging in mouse retrovirus-induced T-cell lymphomas. J. Virol. 77, 2056–2062 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Kohn, D. B., Sadelain, M. & Glorioso, J. C. Occurrence of leukaemia following gene therapy of X-linked SCID. Nature Rev. Cancer 3, 477–488 (2003).

    Article  CAS  Google Scholar 

  39. HaceinBeyAbina, S. et al. Efficacy of gene therapy for X-linked severe combined immunodeficiency. N. Engl. J. Med. 363, 355–364 (2010).

    Article  CAS  Google Scholar 

  40. Higgins, M. E., Claremont, M., Major, J. E., Sander, C. & Lash, A. E. CancerGenes: a gene selection resource for cancer genome projects. Nucleic Acids Res. 35, D721–D726 (2007).

    Article  CAS  PubMed  Google Scholar 

  41. Huret, J. L., Minor, S. L., Dorkeld, F., Dessen, P. & Bernheim, A. Atlas of genetics and cytogenetics in oncology and haematology, an interactive database. Nucleic Acids Res. 28, 349–351 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Sjoblom, T. et al. The consensus coding sequences of human breast and colorectal cancers. Science 314, 268–274 (2006).

    Article  PubMed  CAS  Google Scholar 

  43. Futreal, P. A. et al. A census of human cancer genes. Nature Rev. Cancer 4, 177–183 (2004).

    Article  CAS  Google Scholar 

  44. The Cancer Genome Atlas Research Network. Integrated genomic analyses of ovarian carcinoma. Nature 474, 609–615 (2011).

  45. Lombardo, A. et al. Site-specific integration and tailoring of cassette design for sustainable gene transfer. Nature Methods 8, 861–869 (2011).

    Article  CAS  PubMed  Google Scholar 

  46. Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature 409, 860–921 (2001).

    Article  CAS  PubMed  Google Scholar 

  47. Smit, A. F. Interspersed repeats and other mementos of transposable elements in mammalian genomes. Curr. Opin. Genet. Dev. 9, 657–663 (1999).

    Article  CAS  PubMed  Google Scholar 

  48. Brady, T. et al. Integration target site selection by a resurrected human endogenous retrovirus. Genes Dev. 23, 633–642 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Barr, S. D., Leipzig, J., Shinn, P., Ecker, J. R. & Bushman, F. D. Integration targeting by avian sarcoma-leukosis virus and human immunodeficiency virus in the chicken genome. J. Virol. 79, 12035–12044 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Kotin, R. M., Linden, R. M. & Berns, K. I. Characterization of a preferred site on human chromosome 19q for integration of adeno-associated virus DNA by non-homologous recombination. EMBO J. 11, 5071–5078 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Henckaerts, E. & Linden, R. M. Adeno-associated virus: a key to the human genome? Future Virol. 5, 555–574 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Schnepp, B. C., Jensen, R. L., Chen, C. L., Johnson, P. R. & Clark, K. R. Characterization of adeno-associated virus genomes isolated from human tissues. J. Virol. 79, 14793–14803 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Gao, G. et al. Adeno-associated viruses undergo substantial evolution in primates during natural infections. Proc. Natl Acad. Sci. USA 100, 6081–6086 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Drew, H. R., Lockett, L. J. & Both, G. W. Increased complexity of wild-type adeno-associated virus-chromosomal junctions as determined by analysis of unselected cellular genomes. J. Gen. Virol. 88, 1722–1732 (2007).

    Article  CAS  PubMed  Google Scholar 

  55. Huser, D. et al. Integration preferences of wildtype AAV-2 for consensus rep-binding sites at numerous loci in the human genome. PLoS Pathog 6, e1000985 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. McCarty, D. M., Young, S. M., Jr. & Samulski, R. J. Integration of adeno-associated virus (AAV) and recombinant AAV vectors. Annu. Rev. Genet. 38, 819–845 (2004).

    Article  CAS  PubMed  Google Scholar 

  57. Mehrle, S., Rohde, V. & Schlehofer, J. R. Evidence of chromosomal integration of AAV DNA in human testis tissue. Virus Genes 28, 61–69 (2004).

    Article  CAS  PubMed  Google Scholar 

  58. Hernandez, Y. J. et al. Latent adeno-associated virus infection elicits humoral but not cell-mediated immune responses in a nonhuman primate model. J. Virol. 73, 8549–8558 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. DeKelver, R. C. et al. Functional genomics, proteomics, and regulatory DNA analysis in isogenic settings using zinc finger nuclease-driven transgenesis into a safe harbor locus in the human genome. Genome Res. 20, 1133–1142 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Zou, J. et al. Oxidase-deficient neutrophils from X-linked chronic granulomatous disease iPS cells: functional correction by zinc finger nuclease-mediated safe harbor targeting. Blood 117, 5561–5572 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Ramachandra, C. J. et al. Efficient recombinase-mediated cassette exchange at the AAVS1 locus in human embryonic stem cells using baculoviral vectors. Nucleic Acids Res. 39, e107 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Smith, J. R. et al. Robust, persistent transgene expression in human embryonic stem cells is achieved with AAVS1-targeted integration. Stem Cells 26, 496–504 (2008).

    Article  CAS  PubMed  Google Scholar 

  63. Yang, L. et al. Human cardiovascular progenitor cells develop from a KDR+ embryonicstemcell-derived population. Nature 453, 524–528 (2008).

    Article  CAS  PubMed  Google Scholar 

  64. Hockemeyer, D. et al. Efficient targeting of expressed and silent genes in human ESCs and iPSCs using zinc-finger nucleases. Nature Biotechnol. 27, 851–857 (2009).

    Article  CAS  Google Scholar 

  65. Henckaerts, E. et al. Site-specific integration of adeno-associated virus involves partial duplication of the target locus. Proc. Natl Acad. Sci. USA 106, 7571–7576 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Dutheil, N. et al. Characterization of the mouse adeno-associated virus AAVS1 ortholog. J. Virol. 78, 8917–8921 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Ogata, T., Kozuka, T. & Kanda, T. Identification of an insulator in AAVS1, a preferred region for integration of adeno-associated virus DNA. J. Virol. 77, 9000–9007 (2003).

    Article  CAS  Google Scholar 

  68. Liu, R. et al. Homozygous defect in HIV-1 coreceptor accounts for resistance of some multiply-exposed individuals to HIV-1 infection. Cell 86, 367–377 (1996).

    Article  CAS  PubMed  Google Scholar 

  69. Perez, E. E. et al. Establishment of HIV-1 resistance in CD4+ T cells by genome editing using zinc-finger nucleases. Nature Biotechnol. 26, 808–816 (2008).

    Article  CAS  Google Scholar 

  70. Rottman, J. B. et al. Cellular localization of the chemokine receptor CCR5. Correlation to cellular targets of HIV-1 infection. Am. J. Pathol. 151, 1341–1351 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Lim, J. K., Glass, W. G., McDermott, D. H. & Murphy, P. M. CCR5: no longer a “good for nothing” gene--chemokine control of West Nile virus infection. Trends Immunol. 27, 308–312 (2006).

    Article  CAS  PubMed  Google Scholar 

  72. Lombardo, A. et al. Gene editing in human stem cells using zinc finger nucleases and integrase-defective lentiviral vector delivery. Nature Biotechnol. 25, 1298–1306 (2007).

    Article  CAS  Google Scholar 

  73. Zambrowicz, B. P. et al. Disruption of overlapping transcripts in the ROSA beta geo 26 gene trap strain leads to widespread expression of beta-galactosidase in mouse embryos and hematopoietic cells. Proc. Natl Acad. Sci. USA 94, 3789–3794 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Irion, S. et al. Identification and targeting of the ROSA26 locus in human embryonic stem cells. Nature Biotechnol. 25, 1477–1482 (2007).

    Article  CAS  Google Scholar 

  75. Papapetrou, E. P. et al. Genomic safe harbors permit high beta-globin transgene expression in thalassemia induced pluripotent stem cells. Nature Biotechnol. 29, 73–78 (2011).

    Article  CAS  Google Scholar 

  76. Li, Q., Peterson, K. R., Fang, X. & Stamatoyannopoulos, G. Locus control regions. Blood 100, 3077–3086 (2002).

    Article  CAS  PubMed  Google Scholar 

  77. Bejerano, G. et al. Ultraconserved elements in the human genome. Science 304, 1321–1325 (2004).

    Article  CAS  PubMed  Google Scholar 

  78. Fraser, P. Transcriptional control thrown for a loop. Curr. Opin. Genet. Dev. 16, 490–495 (2006).

    Article  CAS  PubMed  Google Scholar 

  79. Gingeras, T. R. Origin of phenotypes: genes and transcripts. Genome Res. 17, 682–690 (2007).

    Article  CAS  PubMed  Google Scholar 

  80. Ciuffi, A. et al. Methods for integration site distribution analyses in animal cell genomes. Methods (San Diego, Calif.) 47, 261–268 (2009).

    Article  CAS  Google Scholar 

  81. Gabriel, R. et al. Comprehensive genomic access to vector integration in clinical gene therapy. Nature Med. 15, 1431–1436 (2009).

    Article  CAS  PubMed  Google Scholar 

  82. Brady, T. et al. A method to sequence and quantify DNA integration for monitoring outcome in gene therapy. Nucleic Acids Res. 39, e72 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Ogino, H., McConnell, W. B. & Grainger, R. M. High-throughput transgenesis in Xenopus using I-SceI meganuclease. Nature Protoc. 1, 1703–1710 (2006).

    Article  CAS  Google Scholar 

  84. Cermak, T. et al. Efficient design and assembly of custom TALEN and other TAL effector-based constructs for DNA targeting. Nucleic Acids Res. 39, e82 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Papapetrou, E. P. & Sadelain, M. Reconstructing blood from induced pluripotent stem cells. F1000 Med. Rep. 2, 44 (2010).

    PubMed  PubMed Central  Google Scholar 

  86. Wang, G. P. et al. Dynamics of gene-modified progenitor cells analyzed by tracking retroviral integration sites in a human SCID-X1 gene therapy trial. Blood 115, 4356–4366 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank N. Malani, S. Roth and A. Bailey for help with the Cancer Gene database. This work was supported by US NIH grants CA059350, HL053750, DK087923, AI052845 and AI082020 and by the New York State Stem Cell Science (NYSTEM) grant N08T060.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Michel Sadelain.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Related links

Related links

FURTHER INFORMATION

Michel Sadelain's homepage

Atlas of Genetics and Cytogenetics in Oncology and Haematology

Cancer Gene Census

Retroviral Tagged Cancer Gene Database (RTCGD)

The Bushman lab cancer gene list

Waldman gene database

Glossary

Euchromatic portion

A region of chromatin that has lighter packing than heterochromatin and that is generally considered to be richer in actively transcribed genes.

Gene trapping screen

A high-throughput approach used to report and/or inactivate the expression of multiple individual genes across the genome by introducing a reporter gene lacking a promoter (through plasmid or retroviral gene transfer). Selection for expression of the gene requires transcription from a cellular promoter.

Insulator elements

Regulatory DNA elements that create boundaries in chromatin, delineating the ranges over which neighbouring regulatory elements function. They can have enhancer-blocking activity, which prevents communication between discrete sequence elements (typically enhancers and promoters) when insulators are positioned between them, and/or barrier activity, which prevents the spread of heterochromatin.

Intergenic transcription

Transcription of chromosomal DNA sequences between known genes.

Locus control regions

A class of cis-acting DNA regulatory elements that confer high level, tissue-specific, site-of-integration-independent, copy number-dependent expression on linked transgenes located at ectopic chromatin sites.

Matrix attachment regions

AT-rich sequences of DNA that bind to a proteinaceous nuclear scaffold called the nuclear matrix.

Meganucleases

Sequence-specific endonucleases with long recognition sites (>12bp). They are naturally occurring enzymes that are harnessed as tools for targeted genome engineering by the modification of their recognition sequence.

Proviruses

The duplex DNA form of the retroviral genome linked to a cellular chromosome. The provirus is produced by reverse transcription of the RNA genome and subsequent integration into the chromosomal DNA of the host cell.

Retroviral pre-integration complexes

Complexes of viral and cellular proteins with retroviral DNA made by reverse transcription, which together are capable of integrating the viral DNA into a target DNA.

Sub-telomeric regions

Regions adjacent to the telomeres or tips of chromosomes that are often heterochromatic.

Transcription activator-like effector (TALE) nucleases

Artificial endonucleases generated by fusing a TALE DNA-binding domain to the catalytic domain of an endonuclease that introduces double-strand breaks. Similar to zinc-finger nucleases and meganucleases, TALE nucleases can also be engineered to target user-specified DNA sequences within complex genomes.

Zinc-finger nucleases

A class of synthetic proteins that are generated by fusing a zinc-finger DNA-binding domain to the cleavage domain of the FokI restriction endonuclease. The DNA-binding domain can be engineered to induce double-strand breaks in desired DNA sequences, thus facilitating site-specific homologous recombination by the endogenous DNA repair machinery and targeted editing of a genomic locus (insertion, deletion and single-base substitution).

Rights and permissions

Reprints and permissions

About this article

Cite this article

Sadelain, M., Papapetrou, E. & Bushman, F. Safe harbours for the integration of new DNA in the human genome. Nat Rev Cancer 12, 51–58 (2012). https://doi.org/10.1038/nrc3179

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nrc3179

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer