Skip to main content
Log in

Evolution of Hematology Clinical Trial Adverse Event Reporting to Improve Care Delivery

  • Topical Collection on Health Economics (N Khera, Section Editor)
  • Published:
Current Hematologic Malignancy Reports Aims and scope Submit manuscript

Abstract

Purpose of Review

Reporting of adverse events on hematology clinical trials is crucial to understanding the safety of standard treatments and novel agents. However, despite the importance of understanding toxicities, challenges in capturing and reporting accurate adverse event data exist.

Recent Findings

Currently, adverse events are reported manually on most hematology clinical trials. Especially on phase III trials, the highest grade of each adverse event during a reporting period is typically reported. Despite the effort committed to AE reporting, studies have identified underreporting of adverse events on hematologic malignancy clinical trials, which raises concern about the true understanding of safety of treatment that clinicians have in order to guide patients about what to expect during therapy. In order to address these concerns, recent studies have piloted alternative methods for identification of adverse events. These methods include automated extraction of adverse event data from the electronic health record, implementation of trigger or alert tools into the medical record, and analytic tools to evaluate duration of adverse events rather than only the highest adverse event grade.

Summary

Adverse event reporting is a crucial component of clinical trials. Novel tools for identifying and reporting adverse events provide opportunities for honing and refining methods of toxicity capture and improving understanding of toxicities patients experience while enrolled on clinical trials.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References

  1. Miller TP, Fisher BT, Getz KD, Sack L, Razzaghi H, Seif AE, et al. Unintended consequences of evolution of the Common Terminology Criteria for Adverse Events. Pediatr Blood Cancer. 2019;66(7):e27747. https://doi.org/10.1002/pbc.27747.

    Article  PubMed  PubMed Central  Google Scholar 

  2. Miller TP, Li Y, Kavcic M, Troxel AB, Huang YS, Sung L, et al. Accuracy of adverse event ascertainment in clinical trials for pediatric acute myeloid leukemia. J Clin Oncol : official journal of the American Society of Clinical Oncology. 2016;34(13):1537–43. https://doi.org/10.1200/JCO.2015.65.5860.

    Article  Google Scholar 

  3. Farber S, Diamond LK. Temporary remissions in acute leukemia in children produced by folic acid antagonist, 4-aminopteroyl-glutamic acid. N Engl J Med. 1948;238(23):787–93. https://doi.org/10.1056/NEJM194806032382301.

    Article  CAS  PubMed  Google Scholar 

  4. Good MJ, Hurley P, Woo KM, Szczepanek C, Stewart T, Robert N, et al. Assessing clinical trial-associated workload in community-based research programs using the ASCO Clinical Trial Workload Assessment Tool. J Oncol Pract / Am Soc Clin Oncol. 2016;12(5):e536–47. https://doi.org/10.1200/JOP.2015.008920.

    Article  Google Scholar 

  5. Roche K, Paul N, Smuck B, Whitehead M, Zee B, Pater J, et al. Factors affecting workload of cancer clinical trials: results of a multicenter study of the National Cancer Insititue of Canada clinical trials group. J Clin Oncol : official journal of the American Society of Clinical Oncology. 2002;20:545–56.

    Article  Google Scholar 

  6. Trotti A, Bentzen SM. The need for adverse effects reporting standards in oncology clinical trials. J Clin Oncol : official journal of the American Society of Clinical Oncology. 2004;22(1):19–22. https://doi.org/10.1200/JCO.2004.10.911.

    Article  Google Scholar 

  7. Gwede CK, Johnson DJ, Daniels SS, Trotti A. Assessment of toxicity in cooperative oncology clinical trials: the long and short of it. J Oncol Manag : the official journal of the American College of Oncology Administrators. 2002;11(2):15–21.

    Google Scholar 

  8. Trotti A, Byhardt R, Stetz J, Gwede C, Corn B, Fu K, et al. Common toxicity criteria: version 2.0. an improved reference for grading the acute effects of cancer treatment: impact on radiotherapy. Int J Radiat Oncol Biol Phys. 2000;47(1):13–47.

    Article  CAS  Google Scholar 

  9. Thanarajasingam G, Hubbard JM, Sloan JA, Grothey A. The imperative for a new approach to toxicity analysis in oncology clinical trials. J Natl Cancer Inst. 2015;107(10):djv216. https://doi.org/10.1093/jnci/djv216.

    Article  PubMed  Google Scholar 

  10. Trotti A, Colevas AD, Setser A, Rusch V, Jaques D, Budach V, et al. CTCAE v3.0: development of a comprehensive grading system for the adverse effects of cancer treatment. Semin Radiat Oncol. 2003;13(3):176–81. https://doi.org/10.1016/S1053-4296(03)00031-6.

    Article  PubMed  Google Scholar 

  11. Chen AP, Setser A, Anadkat MJ, Cotliar J, Olsen EA, Garden BC, et al. Grading dermatologic adverse events of cancer treatments: the Common Terminology Criteria for Adverse Events Version 4.0. J Am Acad Dermatol. 2012;67(5):1025–39. https://doi.org/10.1016/j.jaad.2012.02.010.

    Article  PubMed  Google Scholar 

  12. NCI. National Cancer Institute Common Terminology Criteria for Adverse Events (CTCAE) v5.0. https://ctep.cancer.gov/protocolDevelopment/electronic_applications/ctc.htm. 2017. Accessed 26 Jan 2021.

  13. Basch E, Abernethy AP, Mullins CD, Reeve BB, Smith ML, Coons SJ, et al. Recommendations for incorporating patient-reported outcomes into clinical comparative effectiveness research in adult oncology. J Clin Oncol : official journal of the American Society of Clinical Oncology. 2012;30(34):4249–55. https://doi.org/10.1200/JCO.2012.42.5967.

    Article  Google Scholar 

  14. Basch E, Reeve BB, Mitchell SA, Clauser SB, Minasian LM, Dueck AC, et al. Development of the National Cancer Institute’s patient-reported outcomes version of the common terminology criteria for adverse events (PRO-CTCAE), Journal of the National Cancer Institute. 2014;106(9):dju244. https://doi.org/10.1093/jnci/dju244.

  15. Chung AE, Shoenbill K, Mitchell SA, Dueck AC, Schrag D, Bruner DW, et al. Patient free text reporting of symptomatic adverse events in cancer clinical research using the National Cancer Institute’s Patient-Reported Outcomes version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE). J Am Med Inform Assoc. 2019;26(4):276–85. https://doi.org/10.1093/jamia/ocy169.

    Article  PubMed  PubMed Central  Google Scholar 

  16. Dueck AC, Mendoza TR, Mitchell SA, Reeve BB, Castro KM, Rogak LJ, et al. Validity and reliability of the US National Cancer Institute’s Patient-Reported Outcomes Version of the Common Terminology Criteria for Adverse Events (PRO-CTCAE). JAMA Oncol. 2015;1:1051–9. https://doi.org/10.1001/jamaoncol.2015.2639.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Trotti A, Colevas AD, Setser A, Basch E. Patient-reported outcomes and the evolution of adverse event reporting in oncology. J Clin Oncol : official journal of the American Society of Clinical Oncology. 2007;25(32):5121–7. https://doi.org/10.1200/JCO.2007.12.4784.

    Article  Google Scholar 

  18. Reeve BB, McFatrich M, Pinheiro LC, Weaver MS, Sung L, Withycombe JS et al. Eliciting the child’s voice in adverse event reporting in oncology trials: cognitive interview findings from the Pediatric Patient-Reported Outcomes version of the Common Terminology Criteria for Adverse Events initiative. Ped Blood Cancer. 2017;64(3). https://doi.org/10.1002/pbc.26261.

  19. McFatrich M, Brondon J, Lucas NR, Hinds PS, Maurer SH, Mack JW, et al. Mapping child and adolescent self-reported symptom data to clinician-reported adverse event grading to improve pediatric oncology care and research. Cancer. 2019;126:140–7. https://doi.org/10.1002/cncr.32525.

    Article  PubMed  PubMed Central  Google Scholar 

  20. Sivendran S, Latif A, McBride RB, Stensland KD, Wisnivesky J, Haines L, et al. Adverse event reporting in cancer clinical trial publications. J Clin Oncol : official journal of the American Society of Clinical Oncology. 2014;32(2):83–9. https://doi.org/10.1200/JCO.2013.52.2219.

    Article  Google Scholar 

  21. Scharf O, Colevas AD. Adverse event reporting in publications compared with sponsor database for cancer clinical trials. J Clin Oncol : official journal of the American Society of Clinical Oncology. 2006;24(24):3933–8. https://doi.org/10.1200/JCO.2005.05.3959.

    Article  Google Scholar 

  22. Zhang S, Liang F, Tannock I. Use and misuse of common terminology criteria for adverse events in cancer clinical trials. BMC Cancer. 2016;16(1):392. https://doi.org/10.1186/s12885-016-2408-9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Miller TP, Li Y, Getz KD, Dudley J, Burrows E, Pennington J, et al. Using electronic medical record data to report laboratory adverse events. Br J Haematol. 2017;177(2):283–6. https://doi.org/10.1111/bjh.14538.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Miller TP, Getz KD, Demissei B, Rabin KR, Daves M, Lupo PJ, et al. Rates of laboratory adverse events by chemotherapy course for pediatric acute leukemia patients within the Leukemia Electronic Abstraction of Records Network (LEARN). Blood. 2019;134(Supplement_1):333. https://doi.org/10.1182/blood-2019-130656.

    Article  Google Scholar 

  25. Weeks HL, Beck C, McNeer E, Williams ML, Bejan CA, Denny JC, et al. medExtractR: a targeted, customizable approach to medication extraction from electronic health records. J Am Med Inform Assoc. 2020. https://doi.org/10.1093/jamia/ocz207.

  26. Hong JC, Fairchild AT, Tanksley JP, Palta M, Tenenbaum JD. Natural language processing for abstraction of cancer treatment toxicities: accuracy versus human experts. JAMIA Open. 2020;3(4):513–7. https://doi.org/10.1093/jamiaopen/ooaa064.

    Article  PubMed  PubMed Central  Google Scholar 

  27. Kehl KL, Elmarakeby H, Nishino M, Van Allen EM, Lepisto EM, Hassett MJ, et al. Assessment of deep natural language processing in ascertaining oncologic outcomes from radiology reports. JAMA Oncol. 2019;5:1421. https://doi.org/10.1001/jamaoncol.2019.1800.

    Article  PubMed  PubMed Central  Google Scholar 

  28. Kehl KL, Xu W, Lepisto E, Elmarakeby H, Hassett MJ, Van Allen EM, et al. Natural language processing to ascertain cancer outcomes from medical oncologist notes. JCO Clin Cancer Inform. 2020;4:680–90. https://doi.org/10.1200/CCI.20.00020.

    Article  PubMed  Google Scholar 

  29. Lencioni A, Hutchins L, Annis S, Chen W, Ermisoglu E, Feng Z, et al. An adverse event capture and management system for cancer studies. BMC Bioinform. 2015;16(Suppl 13):S6. https://doi.org/10.1186/1471-2105-16-S13-S6.

    Article  Google Scholar 

  30. Weingart SN, Nelson J, Koethe B, Yaghi O, Dunning S, Feldman A, et al. Developing a cancer-specific trigger tool to identify treatment-related adverse events using administrative data. Cancer Med. 2020;9:1462–72. https://doi.org/10.1002/cam4.2812.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Call RJ, Burlison JD, Robertson JJ, Scott JR, Baker DK, Rossi MG, et al. Adverse drug event detection in pediatric oncology and hematology patients: using medication triggers to identify patient harm in a specialized pediatric patient population. J Pediatr. 2014;165(3):447–52 e4. https://doi.org/10.1016/j.jpeds.2014.03.033.

    Article  PubMed  PubMed Central  Google Scholar 

  32. Lipitz-Snyderman A, Classen D, Pfister D, Killen A, Atoria CL, Fortier E, et al. Performance of a trigger tool for identifying adverse events in oncology. J Oncol Pract / Am Soc Clin Oncol. 2017;13(3):e223–e30. https://doi.org/10.1200/JOP.2016.016634.

    Article  Google Scholar 

  33. Musy SN, Ausserhofer D, Schwendimann R, Rothen HU, Jeitziner MM, Rutjes AW, et al. Trigger tool-based automated adverse event detection in electronic health records: systematic review. J Med Internet Res. 2018;20(5):e198. https://doi.org/10.2196/jmir.9901.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Co Z, Holmgren AJ, Classen DC, Newmark L, Seger DL, Danforth M, et al. The tradeoffs between safety and alert fatigue: data from a national evaluation of hospital medication-related clinical decision support. J Am Med Inform Assoc. 2020;27(8):1252–8. https://doi.org/10.1093/jamia/ocaa098.

    Article  PubMed  Google Scholar 

  35. Lamont EB, Herndon JE, Weeks JC, Henderson IC, Lilenbaumm R, Schilsky RL, et al. Measuring clinically significant chemotherapy-related toxicities using Medicare claims from Cancer and Leukemia Group B (CALGB) trial participants. Med Care. 2008;46(3):303–8.

    Article  Google Scholar 

  36. Han L, Ball R, Pamer CA, Altman RB, Proestel S. Development of an automated assessment tool for MedWatch reports in the FDA adverse event reporting system. J Am Med Inform Assoc. 2017;24(5):913–20. https://doi.org/10.1093/jamia/ocx022.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Hauben M, Reich L, Chung S. Postmarketing surveillance of potentially fatal reactions to oncology drugs: potential utility of two signal-detection algorithms. Eur J Clin Pharmacol. 2004;60(10):747–50. https://doi.org/10.1007/s00228-004-0834-0.

    Article  PubMed  Google Scholar 

  38. Levit LA, Perez RP, Smith DC, Schilsky RL, Hayes DF, Vose JM. Streamlining adverse events reporting in oncology: an American Society of Clinical Oncology Research Statement. J Clin Oncol : official journal of the American Society of Clinical Oncology. 2018;36(6):617–23. https://doi.org/10.1200/JCO.2017.75.8193.

    Article  Google Scholar 

  39. Thanarajasingam G, Minasian LM, Baron F, Cavalli F, De Claro RA, Dueck AC, et al. Beyond maximum grade: modernising the assessment and reporting of adverse events in haematological malignancies. Lancet Haematol. 2018;5:e563–98. https://doi.org/10.1016/S2352-3026(18)30051-6.

    Article  PubMed  PubMed Central  Google Scholar 

  40. Thanarajasingam G, Atherton PJ, Novotny PJ, Loprinzi CL, Sloan JA, Grothey A. Longitudinal adverse event assessment in oncology clinical trials: the Toxicity over Time (ToxT) analysis of Alliance trials NCCTG N9741 and 979254. Lancet Oncol. 2016;17:663–70. https://doi.org/10.1016/S1470-2045(16)00038-3.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Atkinson TM, Dueck AC, Satele DV, Thanarajasingam G, Lafky JM, Sloan JA, et al. Clinician vs patient reporting of baseline and postbaseline symptoms for adverse event assessment in cancer clinical trials. JAMA Oncol. 2019;6:437–9. https://doi.org/10.1001/jamaoncol.2019.5566.

    Article  PubMed Central  Google Scholar 

Download references

Funding

Dr. Miller received funding from the National Cancer Institute (K07CA211959). Dr. Aplenc received funding from an Alex’s Lemonade Stand Foundation Epidemiology Grant and the Children’s Hospital of Philadelphia Hematologic Malignancy Research Fund.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Tamara P. Miller.

Ethics declarations

Human and Animal Rights and Informed Consent

This article does not contain any studies with human or animal subjects performed by any of the authors.

Conflict of Interest

Dr. Miller and Dr. Aplenc declare that they have no conflicts of interest.

Additional information

Publisher’s Note

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

This article is part of the Topical Collection on Health Economics

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Miller, T.P., Aplenc, R. Evolution of Hematology Clinical Trial Adverse Event Reporting to Improve Care Delivery. Curr Hematol Malig Rep 16, 126–131 (2021). https://doi.org/10.1007/s11899-021-00627-3

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s11899-021-00627-3

Keywords

Navigation