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.

  • Review Article
  • Published:

Prevention and treatment of relapse after stem cell transplantation with immunotherapy

Abstract

Relapse has become the leading cause of treatment failure after stem cell transplantation. Besides cellular therapies and novel agents, immunotherapeutic strategies have entered clinical practice in order to reduce or prevent relapse. Here, we summarize the presentations on checkpoint inhibitors, vaccination strategies, and novel antibody therapies, which were presented and discussed at the third International Workshop on Biology, Prevention, and Treatment of Relapse after Stem Cell Transplantation.

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

Similar content being viewed by others

References

  1. Brahmer JR, Tykodi SS, Chow LQ, Hwu WJ, Topalian SL, Hwu P, et al. Safety and activity of anti-PD-L1 antibody in patients with advanced cancer. N Engl J Med. 2012;366:2455–65.

    Article  CAS  Google Scholar 

  2. Topalian SL, Sznol M, McDermott DF, Kluger HM, Carvajal RD, Sharfman WH, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol. 2014;32:1020–30.

    Article  CAS  Google Scholar 

  3. Borghaei H, Paz-Ares L, Horn L, Spigel DR, Steins M, Ready NE, et al. Nivolumab versus docetaxel in advanced nonsquamous non-small-cell lung cancer. N Engl J Med. 2015;373:1627–39.

    Article  CAS  Google Scholar 

  4. Hamanishi J, Mandai M, Ikeda T, Minami M, Kawaguchi A, Murayama T, et al. Safety and anti-tumor activity of anti-PD-1 antibody, nivolumab, in patients with platinum-resistant ovarian cancer. J Clin Oncol. 2015;33:4015–22.

    Article  CAS  Google Scholar 

  5. Robert C, Schachter J, Long GV, Arance A, Grob JJ, Mortier L, et al. Pembrolizumab versus ipilimumab in advanced melanoma. N Engl J Med. 2015;372:2521–32.

    Article  CAS  Google Scholar 

  6. Ansell SM, Lesokhin AM, Borrello I, Halwani A, Scott EC, Gutierrez M, et al. PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med. 2015;372:311–9.

    Article  Google Scholar 

  7. Younes A, Santoro A, Shipp M, Zinzani PL, Timmerman JM, Ansell S, et al. Nivolumab for classical Hodgkin’s lymphoma after failure of both autologous stem-cell transplantation and brentuximab vedotin: a multicentre, multicohort, single-arm phase 2 trial. Lancet Oncol. 2016;17:1283–94.

    Article  CAS  Google Scholar 

  8. Armand P, Shipp MA, Ribrag V, Michot JM, Zinzani PL, Kuruvilla J, et al. Programmed death-1 blockade with pembrolizumab in patients with classical Hodgkin lymphoma after brentuximab vedotin failure. J Clin Oncol. 2016;34:3733–9.

    Article  CAS  Google Scholar 

  9. Tsirigotis P, Savani BN, Nagler A. Programmed death-1 immune checkpoint blockade in the treatment of hematological malignancies. Ann Med. 2016;48:428–39.

    Article  CAS  Google Scholar 

  10. Rosenblatt J, Avigan D. Targeting the PD-1/PD-L1 axis in multiple myeloma: a dream or a reality? Blood. 2017;129:275–9.

    Article  CAS  Google Scholar 

  11. Krupka C,Kufer P,Kischel R,Zugmaier G,Köhnke T,Lichtenegger FS, et al. PD-1/PD-L1 blocking enhances CD33/CD3-bispecific BiTE® antibody (AMG 330) mediated lysis of primary AML cells. Blood. 2014;124:3738.

    Article  Google Scholar 

  12. Koestner W, Hapke M, Herbst J, Klein C, Welte K, Fruehauf J, et al. PD-L1 blockade effectively restores strong graft-versus-leukemia effects without graft-versus-host disease after delayed adoptive transfer of T-cell receptor gene-engineered allogeneic CD8+ T cells. Blood. 2011;117:1030–41.

    Article  CAS  Google Scholar 

  13. Li JM, Petersen CT, Li JX, Panjwani R, Chandra DJ, Giver CR, et al. Modulation of immune checkpoints and graft-versus-leukemia in allogeneic transplants by antagonizing vasoactive intestinal peptide signaling. Cancer Res. 2016;76:6802–15.

    Article  CAS  Google Scholar 

  14. Michonneau D, Sagoo P, Breart B, Garcia Z, Celli S, Bousso P. The PD-1 axis enforces an anatomical segregation of CTL activity that creates tumor niches after allogeneic hematopoietic stem cell transplantation. Immunity. 2016;44:143–54.

    Article  CAS  Google Scholar 

  15. Bashey A, Medina B, Corringham S, Pasek M, Carrier E, Vrooman L, et al. CTLA4 blockade with ipilimumab to treat relapse of malignancy after allogeneic hematopoietic cell transplantation. Blood. 2009;113:1581–8.

    Article  CAS  Google Scholar 

  16. Albring JC, Inselmann S, Sauer T, Schliemann C, Altvater B, Kailayangiri S, et al. PD-1 checkpoint blockade in patients with relapsed AML after allogeneic stem cell transplantation. Bone Marrow Transplant. 2017;52:317–20.

    Article  CAS  Google Scholar 

  17. Yared JA, Hardy N, Singh Z, Hajj S, Badros AZ, Kocoglu M, et al. Major clinical response to nivolumab in relapsed/refractory Hodgkin lymphoma after allogeneic stem cell transplantation. Bone Marrow Transplant. 2016;51:850–2.

    Article  CAS  Google Scholar 

  18. Chan TS, Khong PL, Kwong YL. Pembrolizumab for relapsed anaplastic large cell lymphoma after allogeneic haematopoietic stem cell transplantation: efficacy and safety. Ann Hematol. 2016;95:1913–5.

    Article  Google Scholar 

  19. Schnorfeil FM, Lichtenegger FS, Emmerig K, Schlueter M, Neitz JS, Draenert R, et al. T cells are functionally not impaired in AML: increased PD-1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J Hematol Oncol. 2015;8:93.

  20. Chung DJ, Pronschinske KB, Shyer JA, Sharma S, Leung S, Curran SA, et al. T-cell exhaustion in multiple myeloma relapse after autotransplant: optimal timing of immunotherapy. Cancer Immunol Res. 2016;4:61–71.

    Article  CAS  Google Scholar 

  21. Merryman RW, Kim HT, Zinzani PL, Carlo-Stella C, Ansell SM, Perales MA, et al. Safety and efficacy of allogeneic hematopoietic stem cell transplant after PD-1 blockade in relapsed/refractory lymphoma. Blood. 2017;129:1380–8.

    Article  CAS  Google Scholar 

  22. Schade H, Sen S, Neff CP, Freed BM, Gao D, Gutman JA, et al. Programmed death 1 expression on CD4+ T cells predicts mortality after allogeneic stem cell transplantation. Biol Blood Marrow Transplant. 2016;22:2172–9.

    Article  CAS  Google Scholar 

  23. Paiva B, Azpilikueta A, Puig N, Ocio EM, Sharma R, Oyajobi BO, et al. PD-L1/PD-1 presence in the tumor microenvironment and activity of PD-1 blockade in multiple myeloma. Leukemia. 2015;29:2110–3.

    Article  CAS  Google Scholar 

  24. Blazar BR, Carreno BM, Panoskaltsis-Mortari A, Carter L, Iwai Y, Yagita H, et al. Blockade of programmed death-1 engagement accelerates graft-versus-host disease lethality by an IFN-gamma-dependent mechanism. J Immunol. 2003;171:1272–7.

    Article  CAS  Google Scholar 

  25. Saha A, O’Connor RS, Thangavelu G, Lovitch SB, Dandamudi DB, Wilson CB, et al. Programmed death ligand-1 expression on donor T cells drives graft-versus-host disease lethality. J Clin Invest. 2016;126:2642–60.

    Article  Google Scholar 

  26. McDuffee E, Aue G, Cook L, Ramos-Delgado C, Shalabi R, Worthy T, et al. Tumor regression concomitant with steroid-refractory GvHD highlights the pitfalls of PD-1 blockade following allogeneic hematopoietic stem cell transplantation. Bone Marrow Transplant. 2017;52:759–61.

    Article  CAS  Google Scholar 

  27. Singh AK, Porrata LF, Aljitawi O, Lin T, Shune L, Ganguly S, et al. Fatal GvHD induced by PD-1 inhibitor pembrolizumab in a patient with Hodgkin’s lymphoma. Bone Marrow Transplant. 2016;51:1268–70.

    Article  CAS  Google Scholar 

  28. Davids MS, Kim HT, Bachireddy P, Costello C, Liguori R, Savell A, et al. Ipilimumab for patients with relapse after allogeneic transplantation. N Engl J Med. 2016;375:143–53.

    Article  CAS  Google Scholar 

  29. Herbaux C, Gauthier J, Brice P, Fornecker L, Bouabdallah K, Manson G, et al. Nivolumab is effective and reasonably safe in relapsed or refractory Hodgkin’s lymphoma after allogeneic hematopoietic cell transplantation: a study from the Lysa and SFGM-TC. Blood. 2015;126:3979.

  30. Haverkos BM, Schowinksy J, Kaplan J, Kamdar M, Kanate AS, Saad A, et al. Checkpoint blockade for treatment of relapsed lymphoma following allogeneic hematopoietic cell transplant: use may be complicated by onset of severe acute graft versus host disease. Blood. 2016;128:1163.

  31. Schoch LK, Borrello I, Fuchs EJ, Bolanos-Meade J, Huo JS, Gojo I, et al. Checkpoint inhibitor therapy and graft versus host disease in allogeneic bone marrow transplant recipients of haploidentical and matched products with post-transplant cyclophosphamide. Blood. 2016;128:4571.

  32. Champlin R. Reduced intensity allogeneic hematopoietic transplantation is an established standard of care for treatment of older patients with acute myeloid leukemia. Best Pract Res Clin Haematol. 2013;26:297–300.

    Article  Google Scholar 

  33. Yoon JH, Kim HJ, Park SS, Jeon YW, Lee SE, Cho BS, et al. Long-term clinical outcomes of hematopoietic cell transplantation for intermediate-to-poor-risk acute myeloid leukemia during first remission according to available donor types. Oncotarget. 2017;8:41590–604.

    PubMed  PubMed Central  Google Scholar 

  34. Guillerey C, Nakamura K, Vuckovic S, Hill GR, Smyth MJ. Immune responses in multiple myeloma: role of the natural immune surveillance and potential of immunotherapies. Cell Mol Life Sci. 2016;73:1569–89.

    Article  CAS  Google Scholar 

  35. Rashidi A, Walter RB. Antigen-specific immunotherapy for acute myeloid leukemia: where are we now, and where do we go from here? Expert Rev Hematol. 2016;9:335–50.

    Article  CAS  Google Scholar 

  36. Rosenblatt J, Bar-Natan M, Munshi NC, Avigan DE. Immunotherapy for multiple myeloma. Expert Rev Hematol. 2014;7:91–6.

    Article  CAS  Google Scholar 

  37. Saito Y, Kitamura H, Hijikata A, Tomizawa-Murasawa M, Tanaka S, Takagi S, et al. Identification of therapeutic targets for quiescent, chemotherapy-resistant human leukemia stem cells. Sci Transl Med. 2010;2:17ra19.

    Article  Google Scholar 

  38. Stroopinsky D, Rosenblatt J, Ito K, Mills H, Yin L, Rajabi H, et al. MUC1 is a potential target for the treatment of acute myeloid leukemia stem cells. Cancer Res. 2013;73:5569–79.

    Article  CAS  Google Scholar 

  39. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74.

    Article  CAS  Google Scholar 

  40. Rosenblatt J, Stone RM, Uhl L, Neuberg D, Joyce R, Levine JD, et al. Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions. Sci Transl Med. 2016;8:368ra171.

    Article  Google Scholar 

  41. Sabado RL, Balan S, Bhardwaj N. Dendritic cell-based immunotherapy. Cell Res. 2017;27:74–95.

    Article  CAS  Google Scholar 

  42. Nahas MR, Avigan D. Challenges in vaccine therapy in hematological malignancies and strategies to overcome them. Expert Opin Biol Ther. 2016;16:1093–104.

    Article  CAS  Google Scholar 

  43. Lacy MQ, Mandrekar S, Dispenzieri A, Hayman S, Kumar S, Buadi F, et al. Idiotype-pulsed antigen-presenting cells following autologous transplantation for multiple myeloma may be associated with prolonged survival. Am J Hematol. 2009;84:799–802.

    Article  Google Scholar 

  44. Rosenblatt J, Vasir B, Uhl L, Blotta S, Macnamara C, Somaiya P, et al. Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma. Blood. 2011;117:393–402.

    Article  CAS  Google Scholar 

  45. Rosenblatt J, Avivi I, Vasir B, Uhl L, Munshi NC, Katz T, et al. Vaccination with dendritic cell/tumor fusions following autologous stem cell transplant induces immunologic and clinical responses in multiple myeloma patients. Clin Cancer Res. 2013;19:3640–8.

    Article  CAS  Google Scholar 

  46. Rapoport AP, Stadtmauer EA, Aqui N, Badros A, Cotte J, Chrisley L, et al. Restoration of immunity in lymphopenic individuals with cancer by vaccination and adoptive T-cell transfer. Nat Med. 2005;11:1230–7.

    Article  CAS  Google Scholar 

  47. Rapoport AP, Aqui NA, Stadtmauer EA, Vogl DT, Fang HB, Cai L, et al. Combination immunotherapy using adoptive T-cell transfer and tumor antigen vaccination on the basis of hTERT and survivin after ASCT for myeloma. Blood. 2011;117:788–97.

    Article  CAS  Google Scholar 

  48. Noonan K, Rudraraju L, Ferguson A, Emerling A, Pasetti MF, Huff CA, et al. Lenalidomide-induced immunomodulation in multiple myeloma: impact on vaccines and antitumor responses. Clin Cancer Res. 2012;18:1426–34.

    Article  CAS  Google Scholar 

  49. Hosen N, Maeda T, Hashii Y, Tsuboi A, Nishida S, Nakata J, et al. Wilms tumor 1 peptide vaccination after hematopoietic stem cell transplant in leukemia patients. Stem Cell Investig. 2016;3:90.

    Article  Google Scholar 

  50. Shah NN, Loeb DM, Khuu H, Stroncek D, Ariyo T, Raffeld M, et al. Induction of immune response after allogeneic Wilms’ tumor 1 dendritic cell vaccination and donor lymphocyte infusion in patients with hematologic malignancies and post-transplantation relapse. Biol Blood Marrow Transplant. 2016;22:2149–54.

    Article  CAS  Google Scholar 

  51. Ho VT, Vanneman M, Kim H, Sasada T, Kang YJ, Pasek M, et al. Biologic activity of irradiated, autologous, GM-CSF-secreting leukemia cell vaccines early after allogeneic stem cell transplantation. Proc Natl Acad Sci USA. 2009;106:15825–30.

    Article  CAS  Google Scholar 

  52. Burkhardt UE, Hainz U, Stevenson K, Goldstein NR, Pasek M, Naito M, et al. Autologous CLL cell vaccination early after transplant induces leukemia-specific T cells. J Clin Invest. 2013;123:3756–65.

    Article  CAS  Google Scholar 

  53. Hoelzer D, Bassan R, Dombret H, Fielding A, Ribbera JM, Buske C. Acute lymphoblastic leukaemia in adult patients: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2016;27(suppl 5):v69–82.

    CAS  PubMed  Google Scholar 

  54. Gökbuget N, Dombret H, Ribera JM, Fielding AK, Advani A, Bassan R, et al. International reference analysis of outcomes in adults with B-precursor Ph-negative relapsed/refractory lymphoblastic leukemia. Haematologica. 2016;101:1524–33.

    Article  Google Scholar 

  55. Raponi S, De Propris MS, Intoppa S, Milani ML, Vitale A, Elia L, et al. Flow cytometric study of potential target antigens (CD19, CD20, CD22, CD33) for antibody-based immunotherapy in acute lymphoblastic leukemia: anlysis of 552 cases. Leuk Lymphoma. 2011;52:1098–107.

    Article  CAS  Google Scholar 

  56. Maury S, Chevret S, Thomas X, Heim D, Leguay T, Huguet F, et al. Rituximab in B-lineage adult acute lymphoblastic leukemia. N Engl J Med. 2016;375:1044–53.

    Article  CAS  Google Scholar 

  57. Chevallier P, Pigneux A, Robillard N, Ayari S, Guillaume T, Delaunay J, et al. Rituximab for the treatment of adult relapsed/refractory CD20 positive B-ALL patients: a pilot series. Leuk Res. 2012;36:311–5.

    Article  CAS  Google Scholar 

  58. Kantarjian H, Thomas D, Jorgensen J, Jabbour E, Kebriaei P, Rytting M, et al. Inotuzumab ozogamicin, an anti-CD22-calecheamicin conjugate, for refractory and relapsed acute lymphocytic leukaemia: a phase 2 study. Lancet Oncol. 2012;13:403–11.

    Article  CAS  Google Scholar 

  59. Kantarjian HM, DeAngelo DJ, Stelljes M, Martinelli G, Liedtke M, Stock W, et al. Inotuzumab ozogamicin versus standard therapy for acute lymphoblastic leukemia. N Engl J Med. 2016;375:740–53.

    Article  CAS  Google Scholar 

  60. Thota S, Advani A. Inotuzumab ozogamicin in relapsed B-cell acute lymphoblastic leukemia. Eur J Haematol. 2017;98:425–34.

    Article  CAS  Google Scholar 

  61. Goebeler ME, Bargou R. Blinatumomab: a CD19/CD3 bispecific T cell engager (BiTE) with unique anti-tumor efficacy. Leuk Lymphoma. 2016;57:1021–32.

    Article  CAS  Google Scholar 

  62. Topp MS, Gokbuget N, Zugmaier G, Klappers P, Stelljes M, Neumann S, et al. Phase II trial of the anti-CD19 bispecific T cell-engager blinatumomab shows hematologic and molecular remissions in patients with relapsed or refractory B-precursor acute lymphoblastic leukemia. J Clin Oncol. 2014;32:4134–40.

    Article  CAS  Google Scholar 

  63. Topp MS, Gokbuget N, Stein AS, Zugmaier G, O’Brien S, Bargou RC, et al. Safety and activity of blinatumomab for adult patients with relapsed or refractory B-precursor acute lymphoblastic leukemia: a multicentre, single-arm, phase 2 study. Lancet Oncol. 2015;16:57–66.

    Article  CAS  Google Scholar 

  64. Kantarjian H, Stein A, Gokbuget N, Fielding AK, Schuh AC, Ribera JM, et al. Blinatumomab versus chemotherapy for advanced acute lymphoblastic leukemia. N Engl J Med. 2017;376:836–47.

    Article  CAS  Google Scholar 

  65. Alcharakh M, Yun S, Dong Y, Vincelette ND, Daud M, Manzoor S, et al. Blinatumomab-induced donor T-cell activation for post-stem cell transplant-relapsed acute CD19-positive biphenotypic leukemia. Immunotherapy. 2016;8:847–52.

    Article  CAS  Google Scholar 

  66. Handgretinger R, Zugmaier G, Henze G, Kreyenberg H, Lang P, von Stackelberg A. Complete remission after blinatumomab-induced donor T-cell activation in three pediatric patients with post-transplant relapsed acute lymphoblastic leukemia. Leukemia. 2011;25:181–4.

    Article  CAS  Google Scholar 

  67. Ueda M, de Lima M, Caimi P, Tomlinson B, Little J, Creger R, et al. Concurrent blinatumomab and donor lymphocyte infusions for treatment of relapsed pre-B-cell ALL after allogeneic hematopoietic cell transplant. Bone Marrow Transplant. 2016;51:1253–5.

    Article  CAS  Google Scholar 

  68. Kebriaei P, Wilhelm K, Ravandi F, Brandt M, de Lima M, Ciurea S, et al. Feasibility of allografting in patients with advanced acute lymphoblastic leukemia after salvage therapy with inotuzumab ozogamicin. Clin Lymphoma Myeloma Leuk. 2013;13:296–301.

    Article  CAS  Google Scholar 

  69. Gökbuget N, Kneba M, Raff T, Trautmann H, Bartram CR, Arnold R, et al. Adult patients with acute lymphoblastic leukemia and molecular failure display a poor prognosis and are candidates for stem cell transplantation and targeted therapies. Blood. 2012;120:1868–76.

    Article  Google Scholar 

  70. Van Dongen JJ, van der Velde VH, Bruggemann M, Orfao A. Minimal residual disease diagnostics in acute lymphoblastic leukemia: need for sensitiove, fast, and standardized technologies. Blood. 2015;125:3996–4009.

    Article  Google Scholar 

  71. Topp MS, Kufer P, Gökbuget N, Goebeler M, Klinger M, Neumann S, et al. Targeted therapy with the T-cell-engaging antibody blinatumomab of chemotherapy-refractory minimal residual disease in B-lineage acute lymphoblastic leukemia patients results in high response rate and prolonged leukemia-free survival. J Clin Oncol. 2011;29:2493–8.

    Article  CAS  Google Scholar 

  72. Topp MS, Gokbuget N, Zugmaier G, Degenhard E, Goebeler ME, Klinger M, et al. Long-term follow-up of hematologic relapse-free survival in a phase 2 study of blinatumomab in patients with MRD in B-lineage ALL. Blood. 2012;120:5185–7.

    Article  CAS  Google Scholar 

  73. Gökbuget N, Brüggemann M, Topp M, Effects of the BiTE® antibody blinatumomab on molecular response in a phase 2 open-label, multicenter confirmatory study in relapsed refractory B-precursor acute lymphoblastic leukemia (R/R ALL). Haematologica. 2014;99:507.

  74. Gökbuget N, Dombret H, Bonifacio M, Reichle A, Graux C, Faul C, et al. Blinatumomab for minimal residual disease in adults with B-precursor acute lymphoblastic leukemia. Blood. 2018. https://doi.org/10.1182/blood-2017-08-798322.

    Article  Google Scholar 

  75. Gokbuget N, Zugmaier G, Klinger M, Kufer P, Stelljes M, Viardot A, et al. Long-term relapse-free survival in a phase 2 study of blinatumomab for the treatment of patients with minimal residual disease in B-lineage acute lymphoblastic leukemia. Haematologica. 2017;102:e132–5.

    Article  Google Scholar 

  76. Taksin AL, Legrand O, Raffoux E, de Revel T, Thomas X, Contentin N, et al. High efficacy and safety profile of fractionated doses of Mylotarg as induction therapy in patients with relapsed acute myeloblastic leukemia: a prospective study of the alfa group. Leukemia. 2007;21:66–71.

    Article  CAS  Google Scholar 

  77. Assi R, Kantarjian H, Ravandi F, Daver N. Immune therapies in acute myeloid leukemia: a focus on monoclonal antibodies and immune checkpoint inhibitors. Curr Opin Hematol. 2018;25:136–45.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank Birgit Ramme for formatting.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Nicolaus Kröger.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Additional information

These authors contributed equally: Nicola Gökbuget, Jonathan Canaani, Arnon Nagler, Michael Bishop, Nicolaus Kröger, David Avigan.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gökbuget, N., Canaani, J., Nagler, A. et al. Prevention and treatment of relapse after stem cell transplantation with immunotherapy. Bone Marrow Transplant 53, 664–672 (2018). https://doi.org/10.1038/s41409-018-0232-3

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41409-018-0232-3

This article is cited by

Search

Quick links