The Neurologic Assessment in Neuro-Oncology (NANO) scale: a tool to assess neurologic function for integration into the Response Assessment in Neuro-Oncology (RANO) criteria

Lakshmi Nayak, Lisa M DeAngelis, Alba A Brandes, David M Peereboom, Evanthia Galanis, Nancy U Lin, Riccardo Soffietti, David R Macdonald, Marc Chamberlain, James Perry, Kurt Jaeckle, Minesh Mehta, Roger Stupp, Alona Muzikansky, Elena Pentsova, Timothy Cloughesy, Fabio M Iwamoto, Joerg-Christian Tonn, Michael A Vogelbaum, Patrick Y Wen, Martin J van den Bent, David A Reardon, Lakshmi Nayak, Lisa M DeAngelis, Alba A Brandes, David M Peereboom, Evanthia Galanis, Nancy U Lin, Riccardo Soffietti, David R Macdonald, Marc Chamberlain, James Perry, Kurt Jaeckle, Minesh Mehta, Roger Stupp, Alona Muzikansky, Elena Pentsova, Timothy Cloughesy, Fabio M Iwamoto, Joerg-Christian Tonn, Michael A Vogelbaum, Patrick Y Wen, Martin J van den Bent, David A Reardon

Abstract

Background: The Macdonald criteria and the Response Assessment in Neuro-Oncology (RANO) criteria define radiologic parameters to classify therapeutic outcome among patients with malignant glioma and specify that clinical status must be incorporated and prioritized for overall assessment. But neither provides specific parameters to do so. We hypothesized that a standardized metric to measure neurologic function will permit more effective overall response assessment in neuro-oncology.

Methods: An international group of physicians including neurologists, medical oncologists, radiation oncologists, and neurosurgeons with expertise in neuro-oncology drafted the Neurologic Assessment in Neuro-Oncology (NANO) scale as an objective and quantifiable metric of neurologic function evaluable during a routine office examination. The scale was subsequently tested in a multicenter study to determine its overall reliability, inter-observer variability, and feasibility.

Results: The NANO scale is a quantifiable evaluation of 9 relevant neurologic domains based on direct observation and testing conducted during routine office visits. The score defines overall response criteria. A prospective, multinational study noted a >90% inter-observer agreement rate with kappa statistic ranging from 0.35 to 0.83 (fair to almost perfect agreement), and a median assessment time of 4 minutes (interquartile range, 3-5).

Conclusion: The NANO scale provides an objective clinician-reported outcome of neurologic function with high inter-observer agreement. It is designed to combine with radiographic assessment to provide an overall assessment of outcome for neuro-oncology patients in clinical trials and in daily practice. Furthermore, it complements existing patient-reported outcomes and cognition testing to combine for a global clinical outcome assessment of well-being among brain tumor patients.

Keywords: brain tumor; neurologic function; outcome; response criteria.

© The Author(s) 2017. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com

Figures

Fig. 1
Fig. 1
Neurologic Assessment in Neuro-Oncology (NANO) scale.
Fig. 1
Fig. 1
Neurologic Assessment in Neuro-Oncology (NANO) scale.
Fig. 2
Fig. 2
Inter-observer study schema.

References

    1. Booth CM, Tannock I. Reflections on medical oncology: 25 years of clinical trials—where have we come and where are we going? J Clin Oncol. 2008;26(1):6–8.
    1. Wilson MK, Karakasis K, Oza AM. Outcomes and endpoints in trials of cancer treatment: the past, present, and future. Lancet Oncol. 2015;16(1):e32–e42.
    1. Helfer JL, Wen PY, Blakeley J, Gilbert MR, Armstrong TS. Report of the Jumpstarting Brain Tumor Drug Development Coalition and FDA clinical trials clinical outcome assessment endpoints workshop (October 15, 2014, Bethesda MD). Neuro Oncol. 2016; 18Suppl 2: ii26–ii36.
    1. Armstrong TS, Bishof AM, Brown PD, Klein M, Taphoorn MJ, Theodore-Oklota C. Determining priority signs and symptoms for use as clinical outcomes assessments in trials including patients with malignant gliomas: Panel 1 Report. Neuro Oncol. 2016;18 Suppl 2:ii1–ii12.
    1. Wen PY, Macdonald DR, Reardon DA, et al. Updated response assessment criteria for high-grade gliomas: Response Assessment in Neuro-Oncology working group. J Clin Oncol. 2010;28(11):1963–1972.
    1. van den Bent MJ, Wefel JS, Schiff D, et al. Response assessment in neuro-oncology (a report of the RANO group): assessment of outcome in trials of diffuse low-grade gliomas. Lancet Oncol. 2011;12(6):583–593.
    1. Lin NU, Lee EQ, Aoyama H, et al. ; Response Assessment in Neuro-Oncology (RANO) group. Challenges relating to solid tumour brain metastases in clinical trials, part 1: patient population, response, and progression. A report from the RANO group. Lancet Oncol. 2013;14(10):e396–e406.
    1. Lin NU, Wefel JS, Lee EQ, et al. ; Response Assessment in Neuro-Oncology (RANO) group. Challenges relating to solid tumour brain metastases in clinical trials, part 2: neurocognitive, neurological, and quality-of-life outcomes. A report from the RANO group. Lancet Oncol. 2013;14(10):e407–e416.
    1. Lin NU, Lee EQ, Aoyama H, et al. ; Response Assessment in Neuro-Oncology (RANO) group. Response assessment criteria for brain metastases: proposal from the RANO group. Lancet Oncol. 2015;16(6):e270–e278.
    1. Macdonald DR, Cascino TL, Schold SC, Jr, Cairncross JG. Response criteria for phase II studies of supratentorial malignant glioma. J Clin Oncol. 1990;8(7):1277–1280.
    1. Armstrong TS, Gilbert MR. Net clinical benefit: functional endpoints in brain tumor clinical trials. Curr Oncol Rep. 2007;9(1):60–65.
    1. Armstrong TS, Mendoza T, Gning I, et al. Validation of the M.D. Anderson Symptom Inventory Brain Tumor module (MDASI-BT). J Neurooncol. 2006;80(1):27–35.
    1. Armstrong TS, Vera-Bolanos E, Gning I, et al. The impact of symptom interference using the MD Anderson Symptom Inventory–Brain Tumor module (MDASI-BT) on prediction of recurrence in primary brain tumor patients. Cancer. 2011;117(14):3222–3228.
    1. Walker M, Brown J, Brown K, Gregor A, Whittle IR, Grant R. Practical problems with the collection and interpretation of serial quality of life assessments in patients with malignant glioma. J Neurooncol. 2003;63(2):179–186.
    1. Orr ST, Aisner J. Performance status assessment among oncology patients: a review. Cancer Treat Rep. 1986;70(12):1423–1429.
    1. Roila F, Lupattelli M, Sassi M, et al. Intra and interobserver variability in cancer patients’ performance status assessed according to Karnofsky and ECOG scales. Ann Oncol. 1991;2(6):437–439.
    1. Hutchinson TA, Boyd NF, Feinstein AR, Gonda A, Hollomby D, Rowat B. Scientific problems in clinical scales, as demonstrated in the Karnofsky index of performance status. J Chronic Dis. 1979;32(9-10):661–666.
    1. Dick JP, Guiloff RJ, Stewart A, et al. Mini-Mental State Examination in neurological patients. J Neurol Neurosurg Psychiatry. 1984;47(5):496–499.
    1. Meyers CA, Brown PD. Role and relevance of neurocognitive assessment in clinical trials of patients with CNS tumors. J Clin Oncol. 2006;24(8):1305–1309.
    1. Meyers CA, Hess KR. Multifaceted end points in brain tumor clinical trials: cognitive deterioration precedes MRI progression. Neuro Oncol. 2003;5(2):89–95.
    1. Herndon RM. Handbook of Neurologic Rating Scales. 2nd ed. New York: Demos Medical Publishing; 2006.
    1. Brott T, Adams HP, Jr, Olinger CP, et al. Measurements of acute cerebral infarction: a clinical examination scale. Stroke. 1989;20(7):864–870.
    1. Kurtzke JF. Further notes on disability evaluation in multiple sclerosis, with scale modifications. Neurology. 1965;15:654–661.
    1. Cedarbaum JM, Stambler N, Malta E, et al. The ALSFRS-R: a revised ALS functional rating scale that incorporates assessments of respiratory function. BDNF ALS Study Group (Phase III). J Neurol Sci. 1999;169(1-2):13–21.
    1. Iwamoto FM, Cooper AR, Reiner AS, Nayak L, Abrey LE. Glioblastoma in the elderly: the Memorial Sloan-Kettering Cancer Center experience (1997-2007). Cancer. 2009;115(16):3758–3766.
    1. Armstrong TS, Cohen MZ, Eriksen LR, Hickey JV. Symptom clusters in oncology patients and implications for symptom research in people with primary brain tumors. J Nurs Scholarsh. 2004;36(3):197–206.
    1. Chamberlain M, Soffietti R, Raizer J, et al. Leptomeningeal metastasis: a Response Assessment in Neuro-Oncology critical review of endpoints and response criteria of published randomized clinical trials. Neuro Oncol. 2014;16(9):1176–1185.
    1. Chamberlain M, Junck L, Brandsma D, et al. Leptomeningeal metastases: a RANO proposal for response criteria. Neuro Oncol. 2016; 16(Suppl 2): ii56.
    1. McHugh ML. Interrater reliability: the kappa statistic. Biochem Med. 2012;22(3):276–282.
    1. Landis JR, Koch GG. The measurement of observer agreement for categorical data. Biometrics. 1977;33(1):159–174.
    1. Viera AJ, Garrett JM. Understanding interobserver agreement: the kappa statistic. Fam Med. 2005;37(5):360–363.
    1. Cohen J. A coefficient of agreement for nominal scales. Educ Psychol Meas. 1960; (20):37–46.
    1. Goldstein LB, Bertels C, Davis JN. Interrater reliability of the NIH stroke scale. Arch Neurol. 1989;46(6):660–662.
    1. Côté R, Battista RN, Wolfson C, Boucher J, Adam J, Hachinski V. The Canadian Neurological Scale: validation and reliability assessment. Neurology. 1989;39(5):638–643.
    1. D’Olhaberriague L, Litvan I, Mitsias P, Mansbach HH. A reappraisal of reliability and validity studies in stroke. Stroke. 1996;27(12):2331–2336.
    1. Shinar D, Gross CR, Mohr JP, et al. Interobserver variability in the assessment of neurologic history and examination in the stroke data bank. Arch Neurol. 1985;42(6):557–565.

Source: PubMed

3
Iratkozz fel