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.

  • Article
  • Published:

Assessment of cognitive and neural recovery in survivors of pediatric brain tumors in a pilot clinical trial using metformin

Abstract

We asked whether pharmacological stimulation of endogenous neural precursor cells (NPCs) may promote cognitive recovery and brain repair, focusing on the drug metformin, in parallel rodent and human studies of radiation injury. In the rodent cranial radiation model, we found that metformin enhanced the recovery of NPCs in the dentate gyrus, with sex-dependent effects on neurogenesis and cognition. A pilot double-blind, placebo-controlled crossover trial was conducted (ClinicalTrials.gov, NCT02040376) in survivors of pediatric brain tumors who had been treated with cranial radiation. Safety, feasibility, cognitive tests and MRI measures of white matter and the hippocampus were evaluated as endpoints. Twenty-four participants consented and were randomly assigned to complete 12-week cycles of metformin (A) and placebo (B) in either an AB or BA sequence with a 10-week washout period at crossover. Blood draws were conducted to monitor safety. Feasibility was assessed as recruitment rate, medication adherence and procedural adherence. Linear mixed modeling was used to examine cognitive and MRI outcomes as a function of cycle, sequence and treatment. We found no clinically relevant safety concerns and no serious adverse events associated with metformin. Sequence effects were observed for all cognitive outcomes in our linear mixed models. For the subset of participants with complete data in cycle 1, metformin was associated with better performance than placebo on tests of declarative and working memory. We present evidence that a clinical trial examining the effects of metformin on cognition and brain structure is feasible in long-term survivors of pediatric brain tumors and that metformin is safe to use and tolerable in this population. This pilot trial was not intended to test the efficacy of metformin for cognitive recovery and brain growth, but the preliminary results are encouraging and warrant further investigation in a large multicenter phase 3 trial.

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

Fig. 1: Pilot trial design and procedures.
Fig. 2: Cranial radiation leads to cellular and cognitive deficits and metformin is able to rescue these deficits in females.
Fig. 3: Pilot trial adverse events and adherence.
Fig. 4: Baseline and outcome data points for LSWM, average reaction time on the CANTAB tests and immediate recall on the CAVLT-2/RAVLT.
Fig. 5: Baseline and outcome data points for AWF within the corpus callosum.

Similar content being viewed by others

Data availability

Requests for data related to the rodent studies should be directed to, and will be fulfilled by, senior corresponding author C.M.M. Requests for resources from the pilot clinical trial, including a copy of the trial protocol and aggregate data, should be directed to, and will be fulfilled by, senior corresponding author D.J.M. Data from individual participants in the pilot trial are not available owing to privacy and confidentiality and participants did not explicitly consent for their data to be shared.

References

  1. Miller, F. D. & Kaplan, D. R. Mobilizing endogenous stem cells for repair and regeneration: are we there yet? Cell Stem Cell 10, 650–652 (2012).

    CAS  PubMed  Google Scholar 

  2. Ming, G. L. & Song, H. Adult neurogenesis in the mammalian brain: significant answers and significant questions. Neuron 70, 687–702 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Lazarini, F. & Lledo, P. M. Is adult neurogenesis essential for olfaction? Trends Neurosci. 34, 20–30 (2011).

    CAS  PubMed  Google Scholar 

  4. Wang, J. et al. Metformin activates an atypical PKC–CBP pathway to promote neurogenesis and enhance spatial memory formation. Cell Stem Cell 11, 23–35 (2012).

    CAS  PubMed  Google Scholar 

  5. Chow, A. & Morshead, C. M. Cyclosporin A enhances neurogenesis in the dentate gyrus of the hippocampus. Stem Cell Res. 16, 79–87 (2016).

    CAS  PubMed  Google Scholar 

  6. Scafidi, J. et al. Intranasal epidermal growth factor treatment rescues neonatal brain injury. Nature 506, 230–234 (2014).

    CAS  PubMed  Google Scholar 

  7. Bridger, T., MacDonald, S., Baltzer, F. & Rodd, C. Randomized placebo-controlled trial of metformin for adolescents with polycystic ovary syndrome. Arch. Pediatr. Adolesc. Med. 160, 241–246 (2006).

    PubMed  Google Scholar 

  8. Benavides, S., Striet, J., Germak, J. & Nahata, M. C. Efficacy and safety of hypoglycemic drugs in children with type 2 diabetes mellitus. Pharmacotherapy 25, 803–809 (2005).

    CAS  PubMed  Google Scholar 

  9. Sun, J., Wang, Y., Zhang, X. & He, H. The effects of metformin on insulin resistance in overweight or obese children and adolescents: a PRISMA-compliant systematic review and meta-analysis of randomized controlled trials. Medicine 98, e14249 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Bjornstad, P. et al. Metformin improves insulin sensitivity and vascular health in youth With type 1 diabetes mellitus. Circulation 138, 2895–2907 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Anagnostou, E. et al. Metformin for treatment of overweight induced by atypical antipsychotic medication in young people with autism spectrum disorder: a randomized clinical trial. JAMA Psychiatry 73, 928–937 (2016).

    PubMed  Google Scholar 

  12. Wang, J. et al. CBP histone acetyltransferase activity regulates embryonic neural differentiation in the normal and Rubinstein–Taybi syndrome brain. Dev. Cell 18, 114–125 (2010).

    CAS  PubMed  Google Scholar 

  13. Dadwal, P. et al. Activating endogenous neural precursor cells using metformin leads to neural repair and functional recovery in a model of childhood brain injury. Stem Cell Rep. 5, 166–173 (2015).

    CAS  Google Scholar 

  14. Ruddy, R. M., Adams, K. V. & Morshead, C. M. Age- and sex-dependent effects of metformin on neural precursor cells and cognitive recovery in a model of neonatal stroke. Sci. Adv. 5, eaax1912 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Tanokashira, D. et al. Metformin treatment ameliorates diabetes-associated decline in hippocampal neurogenesis and memory via phosphorylation of insulin receptor substrate 1. FEBS Open Biol. 8, 1104–1118 (2018).

    CAS  Google Scholar 

  16. Ould-Brahim, F. et al. Metformin preconditioning of human induced pluripotent stem cell-derived neural stem cells promotes their engraftment and improves post-stroke regeneration and recovery. Stem Cells Dev. 27, 1085–1096 (2018).

    CAS  PubMed  Google Scholar 

  17. Monje, M. L., Mizumatsu, S., Fike, J. R. & Palmer, T. D. Irradiation induces neural precursor-cell dysfunction. Nat. Med. 8, 955–962 (2002).

    CAS  PubMed  Google Scholar 

  18. Monje, M. L., Toda, H. & Palmer, T. D. Inflammatory blockade restores adult hippocampal neurogenesis. Science 302, 1760–1765 (2003).

    CAS  PubMed  Google Scholar 

  19. Redmond, K. J., Mahone, E. M. & Horska, A. Association between radiation dose to neuronal progenitor cell niches and temporal lobes and performance on neuropsychological testing in children: a prospective study. Neuro Oncol. 15, 1455 (2013).

    PubMed  PubMed Central  Google Scholar 

  20. Roughton, K., Kalm, M. & Blomgren, K. Sex-dependent differences in behavior and hippocampal neurogenesis after irradiation to the young mouse brain. Eur. J. Neurosci. 36, 2763–2772 (2012).

    PubMed  Google Scholar 

  21. Riggs, L. et al. Changes to memory structures in children treated for posterior fossa tumors. J. Int. Neuropsychol. Soc. 20, 168–180 (2014).

    PubMed  Google Scholar 

  22. Decker, A. L. et al. Smaller hippocampal subfield volumes predict verbal associative memory in pediatric brain tumor survivors. Hippocampus 27, 1140–1154 (2017).

    CAS  PubMed  Google Scholar 

  23. Scantlebury, N. et al. White matter and information processing speed following treatment with cranial-spinal radiation for pediatric brain tumor. Neuropsychology 30, 425–438 (2016).

    PubMed  Google Scholar 

  24. Krull, K. R., Hardy, K. K., Kahalley, L. S., Schuitema, I. & Kesler, S. R. Neurocognitive outcomes and interventions in long-term survivors of childhood cancer. J. Clin. Oncol. 36, 2181–2189 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Rola, R. et al. Radiation-induced impairment of hippocampal neurogenesis is associated with cognitive deficits in young mice. Exp. Neurol. 188, 316–330 (2004).

    CAS  PubMed  Google Scholar 

  26. Fukuda, A. et al. Age-dependent sensitivity of the developing brain to irradiation is correlated with the number and vulnerability of progenitor cells. J. Neurochem. 92, 569–584 (2005).

    CAS  PubMed  Google Scholar 

  27. Hellstrom, N. A., Bjork-Eriksson, T., Blomgren, K. & Kuhn, H. G. Differential recovery of neural stem cells in the subventricular zone and dentate gyrus after ionizing radiation. Stem Cells 27, 634–641 (2009).

    PubMed  Google Scholar 

  28. Ruddy, R. M., Derkach, D., Dadwal, P. & Morshead, C. M. Cranial irradiation in juvenile mice leads to early and sustained defects in the stem and progenitor cell pools and late cognitive impairments. Brain Res. 1727, 146548 (2019).

    PubMed  Google Scholar 

  29. Sorrells, S. F. et al. Human hippocampal neurogenesis drops sharply in children to undetectable levels in adults. Nature 555, 377–381 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Moreno-Jiménez, E. P. et al. Adult hippocampal neurogenesis is abundant in neurologically healthy subjects and drops sharply in patients with Alzheimer’s disease. Nat. Med. 25, 554–560 (2019).

    PubMed  Google Scholar 

  31. Lebel, C. & Beaulieu, C. Longitudinal development of human brain wiring continues from childhood into adulthood. J. Neurosci. 31, 10937–10947 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Chang, A., Nishiyama, A., Peterson, J., Prineas, J. & Trapp, B. D. NG2-positive oligodendrocyte progenitor cells in adult human brain and multiple sclerosis lesions. J. Neurosci. 20, 6404–6412 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Gershon, R. C. et al. NIH Toolbox for assessment of neurological and behavioral function. Neurology 80, S2–S6 (2013).

    PubMed  PubMed Central  Google Scholar 

  34. Talley, J. L. Children’s Auditory Verbal Learning Test-2. Professional Manual (Psychological Assessment Resources, 1993).

  35. Schmidt, M. Rey Auditory Verbal Learning Test: RAVLT: a handbook (Western Psychological Services, 1996).

  36. CANTAB® (cognitive assessment software) http://www.cantab.com/ (Cambridge Cognition, 2018).

  37. Grizzle, J. E. The two-period change-over design and its use in clinical trials. Biometrics 21, 467–480 (1965).

    CAS  PubMed  Google Scholar 

  38. Fieremans, E., Jensen, J. H. & Helpern, J. A. White matter characterization with diffusional kurtosis imaging. Neuroimage 58, 177–188 (2011).

    PubMed  PubMed Central  Google Scholar 

  39. Jelescu, I. O. et al. In vivo quantification of demyelination and recovery using compartment-specific diffusion MRI metrics validated by electron microscopy. Neuroimage 132, 104–114 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Smith, S. M. et al. Tract-based spatial statistics: voxelwise analysis of multi-subject diffusion data. Neuroimage 31, 1487–1505 (2006).

    PubMed  Google Scholar 

  41. Pereira, A. C. et al. An in vivo correlate of exercise-induced neurogenesis in the adult dentate gyrus. Proc. Natl Acad. Sci. USA 104, 5638–5643 (2007).

    CAS  PubMed  Google Scholar 

  42. Kahalley, L. S. et al. Superior intellectual outcomes after proton radiotherapy compared with photon radiotherapy for pediatric medulloblastoma. J. Clin. Oncol. 38, 454–461 (2020).

    PubMed  Google Scholar 

  43. Moxon-Emre, I. et al. Impact of craniospinal dose, boost volume, and neurologic complications on intellectual outcome in patients with medulloblastoma. J. Clin. Oncol. 32, 1760–1768 (2014).

    PubMed  Google Scholar 

  44. Wellek, S. & Blettner, M. On the proper use of the crossover design in clinical trials: part 18 of a series on evaluation of scientific publications. Dtsch. Arztebl. Int. 109, 276–281 (2012).

    PubMed  PubMed Central  Google Scholar 

  45. Zammit, A. R. et al. Roles of hippocampal subfields in verbal and visual episodic memory. Behav. Brain Res. 317, 157–162 (2017).

    PubMed  Google Scholar 

  46. Zheng, F. et al. The volume of hippocampal subfields in relation to decline of memory recall across the adult lifespan. Front. Aging Neurosci. 10, 320 (2018).

    PubMed  PubMed Central  Google Scholar 

  47. Leszczynski, M. How does hippocampus contribute to working memory processing?. Front. Hum. Neurosci. 5, 168 (2011).

    PubMed  PubMed Central  Google Scholar 

  48. Curtis, E. et al. A first-in-human, phase I study of neural stem cell transplantation for chronic spinal cord injury. Cell Stem Cell 22, 941–950 (2018).

    CAS  PubMed  Google Scholar 

  49. Gupta, N. et al. Neural stem cell engraftment and myelination in the human brain. Sci. Transl. Med. 4, 155ra137 (2012).

    PubMed  PubMed Central  Google Scholar 

  50. Ou, Z. et al. Metformin treatment prevents amyloid plaque deposition and memory impairment in APP/PS1 mice. Brain Behav. Immun. 69, 351–363 (2018).

    CAS  PubMed  Google Scholar 

  51. Gantois, I. et al. Metformin ameliorates core deficits in a mouse model of fragile X syndrome. Nat. Med. 23, 674–677 (2017).

    CAS  PubMed  Google Scholar 

  52. Luchsinger, J. A. et al. Metformin in amnestic mild cognitive impairment: results of a pilot randomized placebo controlled clinical trial. J. Alzheimers Dis. 51, 501–514 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Lin, Y. et al. Evaluation of metformin on cognitive improvement in patients with non-dementia vascular cognitive impairment and abnormal glucose metabolism. Front. Aging Neurosci. 10, 227 (2018).

    PubMed  PubMed Central  Google Scholar 

  54. Dy, A. B. C. et al. Metformin as targeted treatment in fragile X syndrome. Clin. Genet. 93, 216–222 (2018).

    CAS  PubMed  Google Scholar 

  55. Gibson, E. M. et al. Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science 344, 1252304 (2014).

    PubMed  PubMed Central  Google Scholar 

  56. Sato, Y. et al. Grafting neural stem and progenitor cells into the hippocampus of juvenile, irradiated mice normalizes behavior deficits. Front. Neurol. 9, 715 (2018).

    PubMed  PubMed Central  Google Scholar 

  57. Babona-Pilipos, R., Popovic, M. R. & Morshead, C. M. A galvanotaxis assay for analysis of neural precursor cell migration kinetics in an externally applied direct current electric field. J. Vis. Exp. 2012, 4193 (2012).

    Google Scholar 

  58. Coles-Takabe, B. L. et al. Don’t look: growing clonal versus nonclonal neural stem cell colonies. Stem Cells 26, 2938–2944 (2008).

    PubMed  Google Scholar 

  59. Nusrat, L. et al. Cyclosporin A-mediated activation of endogenous neural precursor cells promotes cognitive recovery in a mouse model of stroke. Front. Aging Neurosci. 10, 93 (2018).

    PubMed  PubMed Central  Google Scholar 

  60. Spiller, H. A. & Quadrani, D. A. Toxic effects from metformin exposure. Ann. Pharmacother. 38, 776–780 (2004).

    CAS  PubMed  Google Scholar 

  61. Hsia, Y. et al. Unlicensed use of metformin in children and adolescents in the UK. Br. J. Clin. Pharmacol. 73, 135–139 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Vakil, E., Greenstein, Y. & Blachstein, H. Normative data for composite scores for children and adults derived from the Rey Auditory Verbal Learning Test. Clin. Neuropsychol. 24, 662–677 (2010).

    PubMed  Google Scholar 

  63. Riggs, L. et al. Exercise training for neural recovery in a restricted sample of pediatric brain tumor survivors: a controlled clinical trial with crossover of training versus no training. Neuro Oncol. 19, 440–450 (2017).

    PubMed  Google Scholar 

  64. Dale, A. M., Fischl, B. & Sereno, M. I. Cortical surface-based analysis. I. Segmentation and surface reconstruction. Neuroimage 9, 179–194 (1999).

    CAS  PubMed  Google Scholar 

  65. Fischl, B., Liu, A. & Dale, A. M. Automated manifold surgery: constructing geometrically accurate and topologically correct models of the human cerebral cortex. IEEE Trans. Med. Imaging 20, 70–80 (2001).

    CAS  PubMed  Google Scholar 

  66. Ades-Aron, B. et al. Evaluation of the accuracy and precision of the diffusion parameter EStImation with Gibbs and NoisE removal pipeline. Neuroimage 183, 532–543 (2018).

    PubMed  PubMed Central  Google Scholar 

  67. Veraart, J. et al. Denoising of diffusion MRI using random matrix theory. Neuroimage 142, 394–406 (2016).

    PubMed  PubMed Central  Google Scholar 

  68. Veraart, J., Fieremans, E. & Novikov, D. S. Diffusion MRI noise mapping using random matrix theory. Magn. Reson. Med. 76, 1582–1593 (2016).

    CAS  PubMed  Google Scholar 

  69. Kellner, E., Dhital, B., Kiselev, V. G. & Reisert, M. Gibbs-ringing artifact removal based on local subvoxel-shifts. Magn. Reson. Med. 76, 1574–1581 (2016).

    PubMed  Google Scholar 

  70. Andersson, J. L. R. & Sotiropoulos, S. N. An integrated approach to correction for off-resonance effects and subject movement in diffusion MR imaging. Neuroimage 125, 1063–1078 (2016).

    PubMed  PubMed Central  Google Scholar 

  71. Collier, Q., Veraart, J., Jeurissen, B., den Dekker, A. J. & Sijbers, J. Iterative reweighted linear least squares for accurate, fast, and robust estimation of diffusion magnetic resonance parameters. Magn. Reson. Med. 73, 2174–2184 (2015).

    PubMed  Google Scholar 

  72. Avants, B. B. et al. The Insight Toolkit image registration framework. Front. Neuroinformatics 8, 44 (2014).

    Google Scholar 

  73. Engvig, A. et al. Memory training impacts short-term changes in aging white matter: a longitudinal diffusion tensor imaging study. Hum. Brain Mapp. 33, 2390–2406 (2012).

    PubMed  Google Scholar 

  74. Jenkinson, M. & Smith, S. A global optimisation method for robust affine registration of brain images. Med. Image Anal. 5, 143–156 (2001).

    CAS  PubMed  Google Scholar 

  75. Jenkinson, M., Bannister, P., Brady, M. & Smith, S. Improved optimization for the robust and accurate linear registration and motion correction of brain images. Neuroimage 17, 825–841 (2002).

    PubMed  Google Scholar 

  76. Shirzadi, Z., Crane, D. E., Robertson, A. D. & Maralani, P. J. Automated removal of spurious intermediate cerebral blood flow volumes improves image quality among older patients: a clinical arterial spin labeling investigation.J. Magn. Reson. Imaging 42, 1377–1385 (2015).

    PubMed  Google Scholar 

  77. van Osch, M. J. et al. Quantitative cerebral perfusion MRI and CO2 reactivity measurements in patients with symptomatic internal carotid artery occlusion. Neuroimage 17, 469–478 (2002).

    PubMed  Google Scholar 

  78. Hendrikse, J. et al. Internal carotid artery occlusion assessed at pulsed arterial spin-labeling perfusion MR imaging at multiple delay times. Radiology 233, 899–904 (2004).

    PubMed  Google Scholar 

  79. Faul, F., Erdfelder, E., Lang, A.-G. & Buchner, A. G*Power 3: a flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behavior Res, Methods 39, 175–191 (2007).

    Google Scholar 

Download references

Acknowledgements

We wish to thank A. Willan, Clinical Trials Methodologist, Ontario Child Health Support Unit, Research Institute, The Hospital for Sick Children, for consulting on and developing our analytic approach while blind to the trial data. We also wish to thank the research personnel and clinical staff who made this trial possible, particularly V. Ramaswamy, U. Bartels, U. Tabori and A. Huang, as well as N. Sarvaria, D. Thomas, E. Barlev, J. Dutton, J. Gammon, L. Lauer, A. Decker, A. Ferkul, D. McRae, T. Rayner, R. Weiss and M. Lalancette. We would also like to thank J. Wang, D. Kaplan and P. Frankland for frequent discussions and advice during the course of this work. Finally, we wish to thank J. Tseng for her technical support in improving the presentation of our figures. This work was supported by grants from Brain Canada (F.D.M., D.J.M., C.M.M.), the Stem Cell Network (F.D.M., D.J.M., C.M.M.), the Ontario Institute for Regenerative Medicine (F.D.M., D.J.M., C.M.M.), Medicine By Design (CFREF), and the Garron Family Cancer Centre (D.J.M., E.B.). R.M.R. was supported by a CIHR graduate student fellowship.

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, R.M.R., E.B., F.D.M., C.M.M. and D.J.M.; methodology, R.A., R.M.R., E.B., C.M.M. and D.J.M.; software, B.A., Z.S., E.F. and B.J.M.; formal analysis, R.A., R.M.R., E.C., A.O., D.D., C.B.d.M., E.B., C.M.M. and D.J.M.; investigation, R.M.R., C.B.d.M., J.S., E.B., C.M.M. and D.J.M.; resources, S.L., B.A., Z.S., E.F., B.J.M., E.B., C.M.M. and D.J.M.; data curation, R.A., R.M.R., D.D., C.B.d.M. and J.S.; writing—original draft, R.A., R.M.R., F.D.M., C.M.M. and D.J.M.; writing—review and editing, R.A., R.M.R., E.C., A.O., D.D., B.A., Z.S., E.F., B.J.M., C.B.d.M., J.S., E.B., F.D.M., C.M.M. and D.J.M.; supervision, C.M.M. and D.J.M.; project administration, C.B.d.M., J.S. and D.J.M.; funding acquisition, F.D.M., C.M.M. and D.J.M.

Corresponding authors

Correspondence to Cindi M. Morshead or Donald J. Mabbott.

Ethics declarations

Competing interests

E.F. is a co-inventor and the New York University Grossman School of Medicine is owner of the denoising technology used in this manuscript as part of the routine data image processing pipeline; a patent application has been filed and is pending. E.F., B.A. and the New York University Grossman School of Medicine are shareholders and have advisory roles at Microstructure Imaging, Inc. The remaining authors do not have any competing interests to declare.

Additional information

Peer review information Kate Gao and Javier Carmona were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Metformin in the absence of injury has no significant effect on the number of neurospheres or on behavioural assays.

a, b, Fold change in the number of neurospheres from the (a) SVZ (n = 21 Ctrl, 19 Met mice over 10 independent experiments; t(38) = 0.10, p = 0.92) and (b) DG (n = 12 Ctrl, 10 Met mice over 5 independent experiments; t(20) = 0.90, p = 0.38) 5 weeks post-radiation. c, Spontaneous alternation performance measured using the Y maze at P43 (n = 38 Ctrl, 31 Met mice over 14 independent experiments; t(67) = 0.34, p = 0.74). d, Percentage of time spent exploring objects in a novel place measured using the novel place recognition task from P44-46 (n = 34 Ctrl, 27 Met over 13 independent experiments; t(59) = 0.70, p = 0.48). Two-sided unpaired t-test was used for all analyses. Data is presented as mean ± SEM.

Extended Data Fig. 2 Metformin’s effects on NSC pool recovery following juvenile cranial radiation are not sex-dependent.

a, b, Fold change in the number of neurospheres from the SVZ 5 weeks post-radiation in (a) females (n = 8 Ctrl, 9 IR, 6 IR + Met mice over 6 independent experiments; F(2,20) = 0.56, p = 0.58) and (b) males (n = 9 Ctrl, 7 IR, 12 IR+Met mice over 6 independent experiments; F(2,25) = 0.03, p = 0.97). (c-d) Fold change in the number of neurospheres from the DG 5 weeks post-radiation in (c) females (n = 6 Ctrl, 5 IR, 6 IR+Met mice over 4 independent experiments; F(2,14) = 3.47, p = 0.06; Ctrl vs. IR, p = 0.0496) and (d) males (n = 4 Ctrl, 5 IR, 4 IR+Met over 4 independent experiments; F(2,10) = 6.47, p = 0.02; Ctrl vs. IR, p = 0.04; IR vs. IR+Met, p = 0.02). *p < 0.05, one-way ANOVA with Tukey’s test was used for all analyses. Data is presented as mean ± SEM.

Extended Data Fig. 3 Cranial radiation does not lead to impairments in the open field test or in the elevated plus maze.

a, Experimental paradigm. IR=cranial radiation. b, c, Path length travelled over 10 minutes in an open arena by (b) females (n = 17 Ctrl, 15 IR, 13 IR+Met mice over 13 independent experiments; F(2,42) = 0.62, p = 0.54) and (c) males (n = 17 Ctrl, 19 IR, 19 IR+Met mice over 13 independent experiments; F(2,52) = 0.40, p = 0.67). d, e, Percentage of time spent in the open arms of the elevated plus maze over 10 minutes by (d) females (n = 5 Ctrl, 5 IR, 6 IR+Met mice over 4 independent experiments; F(2,13) = 1.26, p = 0.32) and (e) males (n = 5 Ctrl, 5 IR, 6 IR+Met mice over 4 independent experiments; F(2,13) = 0.78, p = 0.48). One-way ANOVA with Tukey’s test was used for all analyses. Data is presented as mean ± SEM.

Extended Data Fig. 4 Consort table.

Eligible participants were identified via database review. Randomization was conducted by the Research Support Pharmacy and all research personnel remained blind to treatment assignment until all participants had completed the trial and data processing and scoring was completed. Initially, due to the pilot nature of the trial, neuroimaging and neuropsychological assessments were acquired at three time points (Baseline 1 and 2; Outcome 2). With an amendment to the study protocol, subsequent participants were assessed at four time points (Baseline 1 and 2; Outcome 1 and 2). Therefore, fewer Outcome 1 than Outcome 2 data points were acquired. Linear mixed modeling can be used in the context of such missing data. The single participant who consented but did not complete the trial was not included in the analyses.

Extended Data Fig. 5 Estimated marginal means for linear mixed models of cognitive and WMTI outcomes.

Data are presented as estimated marginal means from seperate general linear mixed models (two sided) with two sets of outcomes (Outcome 1 and 2 corresponding to the end of the first and second 12-week treatment cycles, respectively). We examined the fixed effects of cycle (the first versus second 12-week treatment cycle), treatment (metformin versus placebo), and sequence (metformin first, placebo second [AB] versus placebo first, metformin second [BA]). Bar graphs show estimated means+/- SEMs from the following model: Outcome measure = cycle + treatment + sequence + covariate (Baseline measure) + (1| participant ID)+ε, where cycle, treatment, and sequence are independent fixed effects and where the measures are: a) total correct on the LSWM (n = 23); b) CANTAB mean latency (n = 22); c) total number of words recall for immediate recall (n = 23); and d) AWF. Standard error bars are shown for each estimated mean. All models were corrected for multiple comparisons (False Discovery Rate (FDR) q < .10): * p < 0.05, ** q < 0.10 from the linear mixed models (Panel a-c, qs = 0.09; Panel d, q = 0.08).

Extended Data Fig. 6 Voxel wise analyses of treatment effects.

We used a longitudinal voxel wise approach to test for clusters of significant changes in AWF and De, following metformin in all participants using Tract Based Spatial Statistics (TBSS). For 2 sided comparisons across treatment conditions, individual difference maps for AWF and De, (post-metformin minus pre-metformin) were projected onto the skeleton and tested for voxels where change was significantly different from zero using threshold-free cluster enhancement (TFCE). For these analyses, the null distribution of the cluster-size statistic was built up over 5000 random permutations. Cluster size was thresholded at P < 0.05, which is family wise fully corrected for multiple comparisons across space. Images are presented in the axial frame in radiological convention within Montreal Neurological Institute (MNI) Z-coordinates. The white matter skeleton is displayed in blue. No significant clusters of change were evident for AWF (p = .90) or De, (p = .47) across the white matter skeleton.

Extended Data Fig. 7 Arterial Spin Labelling and Cerebral Blood Flow within the Hippocampus as a function of cycle, treatment, and sequence effects for the right and left hippocampi and adjusted for baseline hippocampal CBF.

a, Axial T1-weighted image with FreeSurfer hippocampus segmentation shown. b, PASL image processing pipeline shown in the axial plane, including PASL Control, PASL Labeled image, Perfusion weighted Image and Cerebral Blood Flow Map. c, Segmented hippocampi registered to the CBF map. Boxplots showing all data points at baseline and outcome assessment with the mean (dashed line) and median (solid line) sequence group observations for CBF (ml/100 g/min) for: the left hippocampus at d) Cycle 1 and e) Cycle 2; and the right hippocampus at f) Cycle 1 and g) Cycle 2. Metformin treatment condition is shown in red and placebo in blue. The upper and lower limits of the box plots are the third and first quartiles (75th and 25th percentile), respectively. The whiskers extend up to 1.5 times the interquartile range from the top (bottom) of the box to the furthest datum within that distance: Data beyond this distance are represented individually as points.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ayoub, R., Ruddy, R.M., Cox, E. et al. Assessment of cognitive and neural recovery in survivors of pediatric brain tumors in a pilot clinical trial using metformin. Nat Med 26, 1285–1294 (2020). https://doi.org/10.1038/s41591-020-0985-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41591-020-0985-2

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing