Skip to main content
  • Research article
  • Open access
  • Published:

Functional parameters indicative of mild cognitive impairment: a systematic review using instrumented kinematic assessment

Abstract

Background

Patients with mild cognitive impairment (MCI) experience alterations of functional parameters, such as an impaired balance or gait. The current systematic review set out to investigate whether functional objective performance may predict a future risk of MCI; to compare functional objective parameters in patients with MCI and a control group; and to assess changes in these parameters after different physical activity interventions.

Methods

Electronic databases, including PubMed, AMED, CINAHL, EMBASE, PEDro and Web of Science as well as grey literature databases, were searched from inception to February 2020. Cohort studies and Randomized Controlled Trials (RCTs) were included. The risk of bias of the included studies was assessed independently by reviewers using quality assessment checklists. The level of evidence per outcome was assessed using the GRADE criteria.

Results

Seventeen studies met inclusion criteria including patients with MCI. Results from RCTs suggested that gait speed, gait variability and balance may be improved by different physical activity interventions. Cohort studies showed that slower gait speed, above all, under Dual Task (DT) conditions, was the main impaired parameter in patients with MCI in comparison with a Control Gorup. Furthermore, cohort studies suggested that gait variability could predict an incident MCI. Although most of included cohort studies reported low risk of bias, RCTs showed an unclear risk of bias.

Conclusions

Studies suggest that gait variability may predict an incident MCI. Moreover, different gait parameters, above all under DT conditions, could be impaired in patients with MCI. These parameters could be improved by some physical activity interventions. Although cohort studies reported low risk of bias, RCTs showed an unclear risk of bias and GRADE criteria showed a low level of evidence per outcome, so further studies are required to refute our findings.

Prospero

CRD42019119180.

Peer Review reports

Background

The global life expectancy is increasing in the last years. Consequently, morbidity, chronic individual diseases and the number of people affected by dementia are also increasing [1,2,3,4,5]. Thus, while dementias affected around 46.8 million people worldwide in 2016 [5], it is expected that in 2050 there will be 115–135 million people suffering from dementia [6, 7]. There is an increase in the interest of mild cognitive impairment (MCI), defined as a clinical stage accounting for cognitive impairment that often precedes dementia [5, 8,9,10,11,12,13,14,15,16,17], and whose prevalence in adults of ≥65 years old is 10–20%, increasing this prevalence with age [5, 8].

Accepted diagnosis criteria of MCI reported that patients with MCI were characterized by an objective impairment of cognition that is often not severe enough to interfere with activities of daily living (ADL), instrumental activities of daily living (IADL) or in social or occupational functioning [5, 8, 10,11,12,13,14, 17,18,19,20,21]. In the same way, Petersen [11] determined that patients with MCI presented very mild degrees of functional impairment that is difficult to distinguish from the functional problems of cognitively healthy individuals of the same age. However, patients with MCI may have problems in functional tasks [5] and it has been reported that these patients present the alteration of functional parameters, such as mobility, muscle strength, balance, gait dysfunction, or increased risk of falls [8, 22,23,24,25,26]. Slower gait speed has also been suggested as the mainly altered parameter in older populations [23,24,25, 27,28,29,30,31,32] which may be a marker for the preclinical stages of dementia [23, 30,31,32,33]. Thus, Doi et al. [34], Eggermont et al. [35] and Deshpande et al. [36] reported that slower gait speed could be indicative of MCI. Veronese et al. [30] showed an association between decreased gait speed and low performance in the Short Physical Performance Battery (SPPB) and cognitive decline. Other functional tests, such as Timed Up Go (TUG), Hand Grip Strength Test (HGST), Sit to Stand Test (STS), or Walking Speed ​​Test (WST), have also been used to demonstrate the association between the performance on functional tests and MCI [22, 30, 37, 38].

However, Mirelman et al. [39] reported that patients with MCI could have functional alterations only identifiable through a kinematic analysis conducted in their case with an inertial sensor. In this way, Bahureksa et al. [40] revealed that kinematic gait parameters such as velocity, stride length, and stride time best discriminated patients with MCI from cognitively healthy individuals under single task (ST) conditions and, above all, under dual task (DT) conditions. Balance parameters such as anterior-posterior and medio-lateral sway position also were identified as significant discriminators [40]. Kinematic measurements are frequently used by physicians and researchers to quantify normal and pathological movements and could allow to identify altered objective functional parameters in patients with MCI [41]. The identification of changes in functional objective parameters could be relevant for targeting specific interventions aiming to prevent further functional decline or to improve the functionality of patients with MCI. Currently, no drug has been shown to be effective for MCI [8, 17, 18, 42]. However, it has been reported that the combination of aerobic exercise, balance training, cognitive training, the Mediterranean diet and social commitment could reduce the risk of further cognitive impaiment and may improve cognition, mobility, balance and quality of life in patients with MCI [8, 17, 18, 43, 44]. Considering this, the main objectives of this systematic review were (1) to examine if functional kinematic parameters may predict a future risk of MCI; (2) to compare these functional objective parameters in patients with MCI and a control group; (3) to assess longitudinal changes in these parameters after different physical activity interventions. The secondary objectives were (1) to assess the risk of bias of the included studies using The Newcastle-Ottawa Quality Assessment Scale (NOS) and The Cochrane Collaboration’s tool; (2) to assess the level of evidence per outcome using the Grading of Recommendations Assessment, Development and Evaluation (GRADE).

Methods

This systematic review was carried out in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement [45]. The PRISMA checklist for this trial is available in supplementary appendix A. The systematic review protocol was registered at the International Prospective Register of Systematic Reviews (PROSPERO: CRD42019119180).

Data sources and search strategy

A systematic search was performed by two independent reviewers (IJ-FA and A-CV) from inception to February 2020 using optimised search strategies in the following electronic databases: PubMed, AMED, CINAHL, EMBASE, PEDro, Web of Science. A sensitive search strategy using relevant search terms that were developed from Medical Subject Headings (MeSH), and keywords from other similar studies were used: ‘mild cognitive impaiment’ (MeSH Terms), ‘kinetics’ (MeSH Terms), ‘acceleromet*’ (MeSH Terms), ‘walking speed’ (MeSH Terms), ‘kinematic’, ‘kinematic analysis’, ‘Timed Up and Go’, ‘TUG’, ‘gait speed’, ‘gait speed test’, ‘walking speed test’, ‘short physical performance battery’, ‘SPPB’, ‘six minute walk test’, ‘6 minute walk test’, ‘sit to stand test’, ‘single leg stance test’, ‘one leg stance test’, ‘functional reach test’, ‘romberg test’ and ‘functional task´. The complete search strategy report with all search terms is shown online in supplementary appendix B. The grey literature databases, such as New York Academy of Medicine Grey Literature Report, Grey Literature in Health Research and Open Grey were explored to detect any relevant unpublished data. References were exported, and duplicates were removed using citation management software (Mendeley desktop V.1.19.2).

Eligibility criteria

Only studies published in full-text papers were included. Abstracts in conference proceedings, poster presentations, notes or letters to the editor were excluded because they had insufficient detail to be evaluated. Each study had to meet the following inclusion criteria:

  1. 1.

    Cohort studies examining the relationship between functional kinematic parameters obtained by instrumented analysis (e.g., electronic walkways, wearable sensors, camera systems …) and incident MCI or comparing these functional objective parameters between confirmed MCI and a Control Group formed by cognitively healthy individuals or people with Alzheimer Disease.

  2. 2.

    RCTs assessing longitudinal changes in functional objective parameters after different physical activity interventions.

  3. 3.

    Studies that included patients with MCI diagnosed by a specialist or which used validated diagnostic criteria (e.g., Petersen’s et al. [11, 12, 14,15,16], Winblad et al. [13]), supported by a score of 0.5 on the Clinical Dementia Rating (CDR)) [46], < 26 on the Montreal Cognitive Assessment (MoCa) [47, 48], or > 24 Mini-Mental State Examination (MMSE) [48, 49], that permited to confirm the diagnosis of MCI.

  4. 4.

    Studies recruiting participants from any setting (general population, primary, secondary or tertiary care).

  5. 5.

    Studies written in English or Spanish.

The exclusion criteria were as follows:

  1. 1.

    All studies not including a longitudinal design (e.g cross-sectional studies).

  2. 2.

    Studies that included the relationship between functional parameters and incident MCI but did not include a kinematic instrumented analysis.

  3. 3.

    Studies exploring the relationship between functional kinematic parameters and cognitively healthy individuals or people with other neurologic diseases different from MCI.

  4. 4.

    Studies examining the relationship between MCI and other different kinematic parameters such as graphomotor functions, handwriting process variables, etc.

  5. 5.

    Studies that evaluated the relationship between functional kinematic parameters and brain structures in patients with MCI.

  6. 6.

    Studies that did not include validated diagnostic criteria of MCI, did not specify how those patients with MCI were diagnosed or used a diagnosis based on a MMSE score of less than 24, which could be indicative of a greater dementia than the MCI [48,49,50,51,52].

Study selection

All studies identified by the search strategy were screened using the eligibility criteria previously specified. Two independent reviewers (IJ-FA and A-CV) carried out the first stage, which involved the screening of titles and abstracts to identify potentially relevant records. If the reviewers were unable to determine a study’s eligibility based on title and abstract, the full text was retrieved. In this first stage, the two reviewers also excluded those documents that were not full-text papers. The same reviewers undertook the second stage, screening those articles that met all inclusion criteria. A short checklist was carried out to the present review in order to guide the selection of relevant studies (supplementary appendix C).

Data extraction

Two independent reviewers (IJ-FA and A-CV) extracted the following relevant data from each study: study details (first author, year of publication), study design, length of follow up, sample size and characteristics of participants (mean age), functional assessment or test used to assess functional parameters, physical activity intervention, instrument used to kinematic analysis and the methods used to diagnose or assess MCI.

Quality assessment

Two independent reviewers (IJ-FA and A-CV) assessed the risk of bias of the included cohort studies using the NOS [53]. The NOS is a reliable and valid tool for assessing the quality of non-randomized studies [53] and assigns up to a maximum of nine points for the least risk of bias in three domains: selection of study groups (four points); comparability of groups (two points); and ascertainment of exposure and outcomes (three points). The risk of bias of the included RCTs was assessed using The Cochrane Collaboration’s tool [54]. The Cochrane Collaboration’s tool includes seven domain or sources of risk or bias assessment: random sequence generation, allocation concealment, selective reporting, blinding of participants and personnel, blinding of outcome assessment, incomplete outcome data and other bias. For each domain, the risk is categorized as “low risk”, “high risk” or “unclear risk”. To assess the overall quality and the strength of the evidence per outcome, the GRADE approach was used [55]. In brief, the GRADE classification was carried out according to the presence, or not, of the following identified factors: (i) study design, (ii) risk of bias, (iii) inconsistency of results (iv) indirectness (v) imprecision, and (vi) other considerations (e.g. reporting bias). Two researchers (IJ-FA and A-CV) judged whether these factors were present for each outcome. The GRADE criteria was applied when each outcome was informed at least by two studies with the same design. The level of evidence per outcome based on the GRADE criteria is classified as: (1) high (further research is unlikely to change our confidence in the estimate of effect and there are no known or suspected reporting biases); (2) moderate (further research is likely to have an important effect on our confidence in the estimate of effect and might change the estimate); (3) low (further research is likely to have an important effect on our confidence in the estimate of effect and is likely to change the estimate); or (4) very low (we are uncertain about the estimate) [55].

Data synthesis and analysis

It was planned to conduct a meta-analysis of functional kinematic parameters such as gait, balance, posture or mobility, that could be indicative of MCI. However, due to an observed heterogeneity across studies in the type of design, methods of functional assessment, instruments used to conduct a kinematic analysis, duration of follow-up, statistical analysis, interventions and data presentation, the statistical pooling of results was deemed not appropriate. Therefore, a meta-analysis of results was not conducted, and a descriptive quantitative analysis was carried out. For this reason, a narrative synthesis of the most relevant summary measure and the main change from baseline was reported.

Results

Study characteristics

A total of 2239 citations were identified through electronic databases, with 0 additional studies identified through Grey Literature sources. One thousand one hundred fifty-seven titles and abstracts were screened, and 277 full-text papers were assessed. The number of studies retrieved from each database and the number of studies excluded in each screening phase are shown in Fig. 1. The full reference of excluded studies in the second stage (n = 260) is reported online in supplementary appendix D. The conflict of interests of included studies are shown online in supplementary appendix E. Of these, 17 studies (six RCTs, one pilot RCT study, one pilot cohort study, eight cohort studies and a reliability study) with a total of 478 participants with MCI and 1540 cognitively healthy individuals at baseline, were included in this review. The characteristics of the included RCTs and the main results are reported in Table 1. The results of cohort studies which compared functional objective parameters between confirmed MCI and a Control Group are reported in Table 2. The characteristics of cohort studies examining the relationship between functional kinematic parameters and incident MCI are showed in Table 3. Functional kinematic parameters were obtained by wearable sensors, tri-axial accelerometers, digital balance platform, motion and contact sensors, cameras and electronic walkways such as the GAITRite (see Table 4). The most frequently used diagnosis criteria of MCI were Petersen criteria (n = 7, 41%) and the combination of the CDR (n = 9, 53%) and MMSE (n = 11, 65%) (see Table 5).

figure 1

Fig. 1

Table 1 Summary of included RCT studies involved an Instrumented Functional Assessment as Outcome
Table 2 Summary of included Cohort studies which compared Instrumented Objective Functional Parameters between Confirmed MCI and a Control Group
Table 3 Summary of included Cohort studies examined the relationship between Kinematic Functional Parameters and an incident MCI
Table 4 Instruments used in kinematic analysis
Table 5 Criteria for MCI diagnosis reported in studies

Functional objective parameters after physical activity interventions

RCTs showed that gait speed, cadence, stride length, smoothness of trunk movement in the vertical direction could be improved by aerobic exercises (60% of aged predicted maximal heart rate), especially when aerobic exercises are performed alongside cognitive stimulation exercises or others physical exercises such as muscle strength training, postural balance retraining, or gait training [56, 57, 62]. Stride time and the total time to perform the TUG also could be improved by the same interventions [56, 57]. The center of mass sway in anterior-posterior and medial-lateral directions also may be improved by the balance training [58].

Functional objective parameters predicting MCI or discriminating MCI patients from a control group

Cohort studies suggested that a slower gait speed in ST condition and, above all, in DT conditions (counting backwards) was the parameter that best discriminated patients with MCI from cognitively healthy individuals [63, 67, 68]. Larger gait speed variability could also discriminate patients with MCI from cognitively healthy individuals [64, 67]. However, the total time to perform the TUG and the different subtask of the TUG, was the parameter which best discriminated patients with MCI from patients with Alzheimer disease instead of the gait speed, because patients with MCI took lower time in performing the TUG [65]. On the other hand, cohort studies showed that larger gait variability or larger gait speed variability could predict an incident MCI [70, 71]. Decreased cadence and walk-regularity also were associated with an incident MCI [73].

Methodological quality

The methodological quality assessment of RCTs included is shown in supplementary Table 1. In summary, RCTs showed an unclear risk of bias, being the allocation concealment and the blinding of outcome assessment the ítems worst defined. Most of the included cohort studies (60%) reported low risk of bias while the rest (40%) showed moderate risk of bias (supplementary Table 2). Comparability was the main source of bias in cohort studies. The GRADE criteria showed a low level of evidence per outcome (supplementary Table 3).

Discussion

Statement of principal findings and comparision with others studies

The objective of this study was to review the current state of knowledge on the presence of functional kinematic parameters which may predict a future risk of MCI, could discriminate patients with MCI from a control group and could even be improved after different physical activity interventions. To our knowledge, this is the first systematic review that provides a comprehensive overview of longitudinal studies (RCTs and cohort studies) using an instrumented kinematic assessment of functional task as outcome measures or as parameters which could be impaired in patients with MCI or may predict an incident MCI. Furthermore, most of the studies included in this review were published after 2015, which indicates the novelty of the topic [57,58,59,60,61,62,63, 65, 68, 70,71,72,73].

Cohort studies showed that slower gait speed in ST condition and DT conditions (counting backwards), as well as a larger gait speed variability were the parameters that best discriminated patients with MCI from cognitively healthy individuals [64, 67, 68]. Slower gait speed in ST and DT conditions, as well as the time to perform the TUG were the parameters that best discriminated patients with MCI from patients with Alzheimer disease [63, 65]. Some studies have identified a slower gait speed in patients with MCI in comparison with cognitively healthy individuals [25,26,27, 29, 34, 35, 40]. Bahureksa et al. [40] also showed that a shorter stride length, a longer time to perform the stride (stride time) and a larger variability of these parameters may discriminate patients with MCI from cognitively healthy individuals. Verghese et al. [29] added a decreased cadence, a larger swing time variability, and a longer time to perform the swing phase, stance phase and double support phase, as parameters which could be impaired in patients with MCI in comparison with cognitively healthy controls. A previous systematic review also demonstrated slower gait speed and larger gait speed variability in patients with MCI than in cognitively healthy individuals [74].

Thus, larger gait variability, above all in DT conditions, could be associated with MCI [26]. In our review, cohort studies showed that a larger gait variability or a larger gait speed variability could predict an incident MCI [70, 71]. Several studies have also reported that a slow gait speed and a larger gait variability seem to be the main parameters which could predict a future cognitive decline and may be useful in the early detection of MCI [24, 26, 28, 30,31,32,33, 36, 75]. Decreased cadence, walk-regularity and slower gait speed were other kinematic parameters in our review which may be associated with an incident MCI [70, 73].

In the current review, RCTs suggested that gait speed, stride length, stride time, balance and the time to perform the TUG may be improved by aerobic exercises (60% of aged predicted maximal heart rate), especially when aerobic exercises are performed alongside cognitive stimulation exercises or others physical exercises such as muscle strength training, postural balance retraining, or gait training [56,57,58, 61]. Nevertheless, sample sizes were small in most of the included studies [56,57,58,59, 61, 62]. A previous study showed limited evidence on intervention effects on stride time variability [76] although this parameter seems to be a important predictor of MCI [58, 61, 63]. The combination of aerobic exercise, balance training and cognitive training could help reduce the risk of further cognitive impairment and may improve cognition, mobility, balance and quality of life [41, 43]. Furthermore, some systematic reviews and meta-analysis showed that aerobic and resistance (strength) exercises, join cognitive training could improve cognitive function, activities in daily living and mood [77,78,79,80].

Strengths and weaknesses of the study

The strengths of this systematic review included the use of a pre-specified protocol registered on PROSPERO, the PRISMA checklist, the NOS and The Cochrane Collaboration’s tool to determine the risk of bias of included studies and the GRADE criteria to assess the level of evidence per outcome. Furthermore, this review only included studies which provided a validated diagnostic criteria of MCI. There are several limitations that should be mentioned. First, it is possible that some studies were not identified, although we conducted a robust search strategy in order to avoid it. Second, the lack of uniformity among the study design (e.g. walking distance, variables measured, different instruments used in kinematic analysis) should be taken into account when interpreting the results. In the literature, it has been demonstrated that participant walking strategy changes with walking distance, resulting in a significant effect on gait variability [81], so walking distance could be highly relevant in order to measure gait variability as a marker for MCI. Furthermore, studies did not report the reliability, validity or responsiveness of the instruments used in kinematic analysis, so we could not show what motion capture instrument is the most effective to perform the kinematic analysis. Third, RCTs reported an “unclear” risk of bias, so no firm conclusions should be drawn.

Implications for clinical practice

Our results showed kinematic gait parameters which could be impaired and may predict an incident MCI. This is an important step forward in developing a clinically validated approach for measuring MCI related functional deficits which could predict a future risk of MCI and could even help its early diagnosis, although further studies are required in order to validate the findings of this review. Findings of this systematic review also could be useful for promoting specific interventions which could revert the functional changes associated with MCI, since RCTs included in this systematic review have demonstrated that physical activity interventions could improve some functional objective parameters.

Implications for further research

Despite the promising results of the present study, some flaws observed in most of the included studies in this review should be resolved. Hence, there are some recommendations to guide future research: (i) studies should use the same instrument to perform the kinematic analysis which would allow a better comparison of data between studies; (ii) these instruments should be valid and reliable as established in the Cosmin taxonomy; (iii) RCTs and Cohort studies with high quality of evidence should be conducted since studies included in this systematic review often showed an unclear risk of bias and a low quality of evidence; (iv) Clinical trials which use functional objective parameters as outcome measures of physical activity interventions in MCI also should be conducted.

Conclusion

Slower gait speed in ST condition and, above all, in DT conditions and larger gait speed variability are the parameters that best discriminate patients with MCI from a control group. Slower gait speed and larger gait variability may also predict an incident MCI or could even help its early diagnosis. Some functional objective parameters such as gait speed, stride length, stride time, balance and the time to perform the TUG may be improved by aerobic exercises when aerobic exercises are performed alongside cognitive stimulation exercises or others physical exercises such as muscle strength training, postural balance retraining, or gait training. Although most of the cohort studies showed a low risk of bias, RCTs reported unclear risk of bias and GRADE criteria showed a low level of evidence per outcome, so further studies are required to confirm our findings.

Availability of data and materials

Not Applicable – this manuscript does not contain any data.

Abbreviations

MCI:

Mild Cognitive Impairment

AMED:

The Allied and Complementary Medicine Database

CINAHL:

Cumulative Index to Nursing and Allied Health Literature

PEDro:

Physiotherapy Evidence Database

RCTs:

Randomized Controlled Trials

DT:

Dual Task conditions

ADL:

Activities of Daily Living

IADL:

Instrumental Activities of Daily Living

SPPB:

Short Physical Performance Battery

TUG:

Timed Up and Go Test

HGST:

Hand Grip Strength Test

STS:

Sit-To Stand Test

WST:

Walking Speed ​​Test

ST:

Single Task condition

NOS:

Newcastle-Ottawa Quality Assessment Scale

GRADE:

Grading of Recommendations Assessment,Development and Evaluation

PRISMA:

Preferred Reporting Items for Systematic Reviews and Meta-Analyses

PROSPERO:

International Prospective Register of Systematic Reviews

MeSH:

Medical Subject Headings

CDR:

Clinical Dementia Rating

MoCA:

Montreal Cognitive Assessment

MMSE:

Mini-Mental State Examination

References

  1. Abajobir AA, Abate KH, Abbafati C, Abbas KM, Abd-Allah F, Abdulkader RS, et al. Global, regional, and national disability-adjusted life-years (DALYs) for 333 diseases and injuries and healthy life expectancy (HALE) for 195 countries and territories, 1990–2016: a systematic analysis for the global burden of disease study 2016. Lancet. 2017;390(10100):1260–344.

    Google Scholar 

  2. Abajobir AA, Abbafati C, Abbas KM, Abd-Allah F, Abera SF, Aboyans V, et al. Global, regional, and national age-sex specific mortality for 264 causes of death, 1980–2016: a systematic analysis for the global burden of disease study 2016. Lancet. 2017;390(10100):1151–210.

    Google Scholar 

  3. Wang H, Abajobir AA, Abate KH, Abbafati C, Abbas KM, Abd-Allah F, et al. Global, regional, and national under-5 mortality, adult mortality, age-specific mortality, and life expectancy, 1970-2016: a systematic analysis for the global burden of disease study 2016. Lancet. 2017;390(10100):1084–150.

    Google Scholar 

  4. Hoffman C, Rice D, Sung HY. Persons with chronic conditions: their prevalence and costs. JAMA. 1996;276(18):1473–9.

    CAS  PubMed  Google Scholar 

  5. Cornelis E, Gorus E, Beyer I, Bautmans I, De Vriendt P. Early diagnosis of mild cognitive impairment and mild dementia through basic and instrumental activities of daily living: development of a new evaluation tool. PLoS Med. 2017;14(3):1–22.

    Google Scholar 

  6. Ferretti-Rebustini RE d L, Balbinotti MAA, Jacob-Filho W, Rebustini F, Suemoto CK, Pasqualucci CAG, et al. Validity of the Katz index to assess activities of daily living by informants in neuropathological studies. Rev Esc Enferm USP. 2015;49(6):946–52.

    PubMed  Google Scholar 

  7. Limongi F, Siviero P, Noale M, Gesmundo A, Crepaldi G, Maggi S. Prevalence and conversion to dementia of mild cognitive impairment in an elderly Italian population. Aging Clin Exp Res. 2017;29(3):361–70.

    PubMed  Google Scholar 

  8. Langa K, Levine D. The diagnosis and Management of Mild Cognitive Impairment: a clinical review. JAMA. 2014;312(23):2551–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Brodaty H, Aerts L, Crawford JD, Heffernan M, Kochan NA, Reppermund S, et al. Operationalizing the diagnostic criteria for mild cognitive impairment: the salience of objective measures in predicting incident dementia. Am J Geriatr Psychiatry. 2017;25(5):485–97.

    PubMed  Google Scholar 

  10. Roberts R, Knopman DS. Classification and epidemiology of MCI. Clin Geriatr Med. 2013;29(4). https://doi.org/10.1016/j.cger.2013.07.003.

  11. Petersen RC. Mild cognitive impairment as a diagnostic entity. J Intern Med. 2004;256(9):183–94.

    CAS  PubMed  Google Scholar 

  12. Petersen RC, Caracciolo B, Brayne C, Gauthier S, Jelic V, Fratiglioni L. Mild cognitive impairment: a concept in evolution. J Intern Med. 2014;275(3):214–28.

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Winblad B, Palmer K, Kivipelto M, Jelic V, Fratiglioni L, Wahlund L-O, et al. Mild cognitive impairment--beyond controversies, towards a consensus: report of the international working group on mild cognitive impairment. J Intern Med. 2004;256(3):240–6.

    CAS  PubMed  Google Scholar 

  14. Petersen RC, Smith GE, Waring SC, Ivnik RJ, Tangalos EG, Kokmen E. Mild cognitive impairment: clinical characterization and outcome. Arch Neurol. 1999;56(3):303–8.

    CAS  PubMed  Google Scholar 

  15. Petersen RC, Doody R, Kurz A, Mohs RC, Morris JC, Rabins PV, et al. Current concepts in mild cognitive impairment. Arch Neurol. 2001;58:1985–92.

  16. Petersen R, Morris J. Mild cognitive impairment as a clinical entity and treatment target. Arch Neurol. 2005;62(7):1160–3.

    PubMed  Google Scholar 

  17. Sachs-Ericsson N, Blazer DG. The new DSM-5 diagnosis of mild neurocognitive disorder and its relation to research in mild cognitive impairment. Aging Ment Health. 2015;19(1):2–12.

    PubMed  Google Scholar 

  18. Lin JS, O’Connor E, Rossom RC, Perdue LA, Eckstrom E. Screening for cognitive impairment in older adults: a systematic review for the U.S. preventive services task force. Ann Intern Med. 2013;159(9):601–12.

  19. Mitchell AJ, Beaumont H, Ferguson D, Yadegarfar M, Stubbs B. Risk of dementia and mild cognitive impairment in older people with subjective memorycomplaints: meta-analysis. Acta Psychiatr Scand. 2014;130(6):439–51.

    CAS  PubMed  Google Scholar 

  20. Petersen RC, Knopman DS, Boeve BF, Yonas E, Ivnik RJ, Smith GE, et al. Mild cognitive impairment: ten years later. Arch Neurol. 2009;66(12):1447–55.

    PubMed  PubMed Central  Google Scholar 

  21. Portet F. Mild cognitive impairment (MCI) in medical practice: a critical review of the concept and new diagnostic procedure. Report of the MCI working Group of the European Consortium on Alzheimer’s disease. J Neurol Neurosurg Psychiatry. 2006;77(6):714–8 [cited 2019 Jan 21]. Available from: http://jnnp.bmj.com/cgi/doi/10.1136/jnnp.2005.085332.

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Lee SH, Han JH, Jin YY, Lee IH, Hong HR, Kang HS. Poor physical fitness is independently associated with mild cognitive impairment in elderly Koreans. Biol Sport. 2016;33(1):57–62.

    CAS  PubMed  Google Scholar 

  23. Waite LM, Grayson DA, Piguet O, Creasey H, Bennett HP, Broe GA. Gait slowing as a predictor of incident dementia: 6-year longitudinal data from the Sydney older persons study. J Neurol Sci. 2005;229–230:89–93.

    PubMed  Google Scholar 

  24. Buracchio T, Dodge H, Howieson D, Wasserman D, Kaye J. The trajectory of gait speed preceding MCI. Arch Neurol. 2010;67(8):980–6.

    PubMed  PubMed Central  Google Scholar 

  25. Tseng B, Cullum C, Zhang R. Older adults with amnestic mild cognitive impairment exhibit exacerbated gait slowing under dual-task challenges. Curr Alzheimer Res. 2014;11(5):494–500 [cited 2019 Feb 12]. Available from:http://www.eurekaselect.com/openurl/content.php?genre=article&issn=1567-2050&volume=11&issue=5&spage=494.

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Montero-Odasso M, Muir SW, Speechley M. Dual-task complexity affects gait in people with mild cognitive impairment: the interplay between gait variability, dual tasking, and risk of falls. Arch Phys Med Rehabil. 2012;93(2):293–9.

    PubMed  Google Scholar 

  27. Gillain S, Warzee E, Lekeu F, Wojtasik V, Maquet D, Croisier JL, et al. The value of instrumental gait analysis in elderly healthy, MCI or Alzheimer’s disease subjects and a comparison with other clinical tests used in single and dual-task conditions. Ann Phys Rehabil Med. 2009;52(6):453–74.

    CAS  PubMed  Google Scholar 

  28. Aggarwal NT, Wilson RS, Beck TL, Bienias JL, Bennett DA. Motor dysfunction in mild cognitive impairment and the risk of incident Alzheimer disease. Arch Neurol. 2006;63(12):1763–9.

    PubMed  Google Scholar 

  29. Verghese J, Robbins M, Holtzer R, Zimmerman M, Wang C, Xue X, et al. Gait dysfunction in mild cognitive impairment syndromes. J Am Geriatr Soc. 2009;56(7):1244–51.

    Google Scholar 

  30. Veronese N, Stubbs B, Trevisan C, Bolzetta F, De Rui M, Solmi M, et al. What physical performance measures predict incident cognitive decline among intact older adults? A 4.4 year follow up study. Exp Gerontol. 2016;81:110–8.

    PubMed  Google Scholar 

  31. Mielke MM, Roberts RO, Savica R, Cha R, Drubach DI, Christianson T, et al. Assessing the temporal relationship between cognition and gait: slow gait predicts cognitive decline in the Mayo Clinic study of aging. J Gerontol Med Sci. 2013;68(8):929–37.

    Google Scholar 

  32. Ojagbemi A, Este CD, Verdes E, Chatterji S, Gureje O. Gait & Posture Gait speed and cognitive decline over 2 years in the Ibadan study of aging. Gait Posture. 2015;41(2):736–40.

    PubMed  PubMed Central  Google Scholar 

  33. Verghese J, Wang C, Lipton RB, Holtzer R, Xue X. Quantitative gait dysfunction and risk of cognitive decline and dementia. J Neurol Neurosurg Psychiatry. 2007;78(9):929–35.

    PubMed  PubMed Central  Google Scholar 

  34. Doi T, Shimada H, Park H, Makizako H, Tsutsumimoto K, Uemura K, et al. Cognitive function and falling among older adults with mild cognitive impairment and slow gait. Geriatr Gerontol Int. 2015;15(8):1073–8.

    PubMed  Google Scholar 

  35. Eggermont LH, Gavett BE, Volkers KM, Blankevoort CG, Scherder EJ, Jefferson AL, et al. Lower-extremity function in cognitively healthy aging, mild cognitive impairment, and Alzheimer’s disease. Arch Phys Med Rehabil. 2010 Apr;91(4):584–8.

    PubMed  PubMed Central  Google Scholar 

  36. Deshpande N, Metter EJ, Bandinelli S, Guralnik J, Ferrucci L. Gait speed under varied challenges and cognitive decline in older persons: a prospective study. Age Ageing. 2009;38(5):509–14.

    PubMed  PubMed Central  Google Scholar 

  37. Narazaki K, Matsuo E, Honda T, Nofuji Y, Yonemoto K, Kumagai S. Physical fitness measures as potential markers of low cognitive function in Japanese community-dwelling older adults without apparent cognitive problems. J Sports Sci Med. 2014 Sep 1;13(3):590–6.

    PubMed  PubMed Central  Google Scholar 

  38. Ansai JH, de Andrade LP, Rossi PG, Nakagawa TH, Vale FAC, Rebelatto JR. Differences in timed up and go subtasks between older people with mild cognitive impairment and mild Alzheimer’s disease. Motor Control. 2019 Jan 1;23(1):1–12. https://doi.org/10.1123/mc.2017-0015.

    Article  PubMed  Google Scholar 

  39. Mirelman A, Weiss A, Buchman AS, Bennett DA, Nir Giladi JMH. Association between performance on timed up and go subtasks and mild cognitive impairment: further insights into the links between cognitive and motor function. J Am Geriatr Soc. 2014;62(4):673–8.

    PubMed  PubMed Central  Google Scholar 

  40. Bahureksa L, Najafi B, Saleh A, Sabbagh M, Coon D, Mohler J, et al. The impact of mild cognitive impairment on gait and balance: a systematic review and meta-analysis of studies using instrumented assessment. Gerontology. 2017;28(2):180–7.

    Google Scholar 

  41. Cuesta-Vargas AI, Galán-Mercant A, Williams JM. The use of inertial sensors system for human motion analysis. Phys Ther Rev. 2010;15(6):462–73.

    PubMed  PubMed Central  Google Scholar 

  42. Han JW, Lee H, Hong JW, Kim K, Kim T, Byun HJ, et al. Multimodal cognitive enhancement therapy for patients with mild cognitive impairment and mild dementia: a multi- center, randomized, controlled, double-blind, crossover trial. J Alzheimers Dis. 2017;55(2):787–96.

    PubMed  Google Scholar 

  43. Panza GA, Taylor BA, Macdonald HV, Johnson BT, Zaleski AL, Livingston J, et al. Can exercise improve cognitive symptoms of Alzheimer’s disease? A meta-analysis. J Am Geriatr Soc. 2018;66(3):487–95.

    PubMed  Google Scholar 

  44. Hagovská M, Olekszyová Z. Impact of the combination of cognitive and balance training on gait, fear and risk of falling and quality of life in seniors with mild cognitive impairment. Geriatr Gerontol Int. 2016 Sep;16(9):1043–50.

    PubMed  Google Scholar 

  45. Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gotzsche PC, Ioannidis JP, et al. The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate healthcare interventions: explanation and elaboration. BMJ. 2009;339:b2700.

    PubMed  PubMed Central  Google Scholar 

  46. Morris JC. The clinical dementia rating (CDR): current version and scoring rules. Neurology. 1993;43:2412–4.

    CAS  PubMed  Google Scholar 

  47. Nasreddine ZS, Phillips NA, Bédirian V, et al. The Montreal cognitive assessment, MoCA: a brief screening tool for mild cognitive impairment. J Am Geriatr Soc. 2005;53:695–9.

    PubMed  Google Scholar 

  48. Ciesielska N, Sokołowski R, Mazur E, Podhorecka M, Polak-Szabela A, Kędziora-Kornatowska K. Is the Montreal cognitive assessment (MoCA) test better suited than the mini-mental state examination (MMSE) in mild cognitive impairment (MCI) detection among people aged over 60? Meta-analysis. Psychiatr Pol. 2016;50(5):1039–52.

    PubMed  Google Scholar 

  49. Folstein MF, Folstein SE, McHugh PR. Mini-mental state: a practical method for grading the state of patients for the clinician. J Psychiatr Res. 1975;12:189–98.

    CAS  PubMed  Google Scholar 

  50. Tombaugh TN, Mclntyre NJ. The mini-mental state examination: a comprehensive review. J Am Geriatr Soc. 1992;40:922–35.

    CAS  PubMed  Google Scholar 

  51. Mitchell AJ. A meta-analysis of the accuracy of the mini-mental state examination in the detection of dementia and mild cognitive impairment. J Psychiatr Res. 2009;43(4):411–31.

    PubMed  Google Scholar 

  52. Cacho J, Benito-León J, García-García R, Fernández-Calvo B, Vicente-Villardón JL, Mitchell AJ. Does the combination of the MMSE and clock drawing test (mini-clock) improve the detection of mild Alzheimer’s disease and mild cognitive impairment? J Alzheimers Dis. 2010;22(3):889–96.

    PubMed  Google Scholar 

  53. Wells GA, Shea B, O’Connell D, Peterson J, Welch V, Losos M, et al. The Newcastle-Ottawa scale (NOS) for assessing the quality of non-randomised studies in meta-analyses; 2009. [cited 2019 September 24]. Available from: http://www.ohri.ca/programs/clinical_epidemiology/oxford.asp.

    Google Scholar 

  54. Higgins JPT, Altman DG, Gøtzsche PC, Jüni P, Moher D, Oxman AD, et al. The Cochrane Collaboration’s tool for assessing risk of bias in randomised trials. BMJ. 2011;343:d5928.

    PubMed  PubMed Central  Google Scholar 

  55. Atkins D, Best D, Briss PA, et al. Grading quality of evidence and strength of recommendations. BMJ. 2004;328:1490. https://doi.org/10.1136/bmj.328.7454.1490 [PMC free article] [PubMed].

    Article  PubMed  Google Scholar 

  56. Doi T, Makizako H, Shimada H, Yoshida D, Tsutsumimoto K, Sawa R, et al. Effects of multicomponent exercise on spatial–temporal gait parameters among the elderly with amnestic mild cognitive impairment (aMCI): preliminary results from a randomized controlled trial (RCT). Arch Gerontol Geriatr. 2013;56(1):104–8 [cited 2019 Mar 13]. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0167494312002002.

    PubMed  Google Scholar 

  57. Donnezan LC, Perrot A, Sylvie Belleville FB, Kemoun G. Effects of simultaneous aerobic and cognitive training on executive functions, cardiovascular fitness and functional abilities in older adults with mild cognitive impairment. Ment Health Phys Act. 2018;15:78–87. https://doi.org/10.1016/j.mhpa.2018.06.001.

    Article  Google Scholar 

  58. Schwenk M, Sabbagh M, Lin I, Morgan P, Grewal GS, Mohler J, et al. Sensor-based balance training with motion feedback in people with mild cognitive impairment. J Rehabil Res Dev. 2016;53(6):945–58.

    PubMed  PubMed Central  Google Scholar 

  59. Fogarty JN, Murphy KJ, McFarlane B, Montero-Odasso M, Wells J, Troyer AK, et al. Taoist tai chi® and memory intervention for individuals with mild cognitive impairment. J Aging Phys Act. 2016 Apr;24(2):169–80.

    PubMed  Google Scholar 

  60. Bae S, Sangyoon Lee SL, Jung S, Makino K, Kazuhiro Harada K, Harada YS, et al. The effect of a multicomponent intervention to promote community activity on cognitive function in older adults with mild cognitive impairment: a randomized controlled trial. Complement Ther Med. 2018;42:164–9.

    PubMed  Google Scholar 

  61. Delbroek T, Vermeylen W, Spildooren J. The effect of cognitive-motor dual task training with the biorescue force platform on cognition, balance and dual task performance in institutionalized older adults: a randomized controlled trial. J Phys Ther Sci. 2017;29(7):1137–43.

    PubMed  PubMed Central  Google Scholar 

  62. Liao Y-Y, Chen I-H, Lin Y-J, Chen Y, Hsu W-C. Effects of virtual reality-based physical and cognitive training on executive function and dual-task gait performance in older adults with mild cognitive impairment: a randomized control trial. Front Aging Neurosci. 2019;11:162.

    PubMed  PubMed Central  Google Scholar 

  63. Gillain S, Dramé M, Lekeu F, Wojtasik V, Ricour C, Croisier J-L, et al. Gait speed or gait variability, which one to use as a marker of risk to develop Alzheimer disease? A pilot study. Aging Clin Exp Res. 2015;28(2):249–55 [cited 2019 Mar 19]. Available from: http://link.springer.com/10.1007/s40520-015-0392-6.

    PubMed  Google Scholar 

  64. Tamara L, Hayes FA, Adami A, Pavel M, Zitzelberger JAK TA. Unobtrusive assessment of activity patterns associated with mild cognitive impairment. Alzheimers Dement. 2008;4(6):395–405.

    Google Scholar 

  65. Ansai JH, de Andrade LP, Nakagawa TH, Rebelatto JR. Performances on the timed up and go test and subtasks between fallers and non-fallers in older adults with cognitive impairment. Arq Neuropsiquiatr. 2018;76(6):381–6.

    PubMed  Google Scholar 

  66. Pfeiffer E. A short portable mental status questionnaire for the assessment of organic brain deficit in elderly patients. J Am Geriatr Soc. 1975;23(10):433–41.

    CAS  PubMed  Google Scholar 

  67. Dodge HH, Mattek NC, Austin D, Hayes TL, Kaye JA. In-home walking speeds and variability trajectories associated with mild cognitive impairment. Neurology. 2012 Jun 12;78(24):1946–52.

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Pieruccini-Faria F, Sarquis-Adamson Y, Montero-Odasso M. Mild cognitive impairment affects obstacle negotiation in older adults: results from “gait and brain study”. Gerontology. 2019;65(2):164–73.

    PubMed  Google Scholar 

  69. Montero-Odasso M, Alvaro Casas KTH, Bilski P, Iris Gutmanis JLW, M JB. Quantitative gait analysis under dual-task in older people with mild cognitive impairment: a reliability study. J Neuroeng Rehabil. 2009;6:35.

    PubMed  PubMed Central  Google Scholar 

  70. Byun S, Han JW, Kim TH, Kim K, Kim TH, Park JY, et al. Gait variability can predict the risk of cognitive decline in cognitively Normal older people. Dement Geriatr Cogn Disord. 2018;45(5–6):251–61.

    PubMed  Google Scholar 

  71. Akl A, Taati B, Mihailidis A. Autonomous unobtrusive detection of mild cognitive impairment in older adults. IEEE Trans Biomed Eng. 2015 May;62(5):1383–94.

    PubMed  PubMed Central  Google Scholar 

  72. Akl A, Mihailidis A. Estimating in-home walking speed distributions for unobtrusive detection of mild cognitive impairment in older adults. In: 37th annual international conference of the IEEE engineering in medicine and biology society (EMBC); 2015. p. 5175–8.

    Google Scholar 

  73. Buchman A-S, Dawe R-J, Leurgans S-E, Curran T-A, Truty T, Yu L, et al. Different combinations of mobility metrics derived from a wearable sensor are associated with distinct health outcomes in older adults. J Gerontol A Biol Sci Med Sci. 2020;75(6):1176–83. https://doi.org/10.1093/gerona/glz160.

  74. Lussier M, Lavoie M, Giroux S, Consel C, Guay M, Macoir J, et al. Early detection of mild cognitive impairment with in-home monitoring technologies using functional measures: a systematic review. IEEE J Biomed Heal Informatics. 2019;23(2):838–47.

    Google Scholar 

  75. Lamoth CJ, van Deudekom FJ, van Campen JP, Appels BA, de Vries OJ, Pijnappels M. Gait stability and variability measures show effects of impaired cognition and dual tasking in frail people. J Neuroeng Rehabil. 2011;8:2–2.

    PubMed  PubMed Central  Google Scholar 

  76. Schwenk M, Zieschang T, Englert S, Grewal G, Najafi B, Hauer K. Improvements in gait characteristics after intensive resistance and functional training in people with dementia: a randomised controlled trial. BMC Geriatr. 2014;14:73.

    PubMed  PubMed Central  Google Scholar 

  77. Gomes-Osman J, Cabral DF, Morris TP, McInerney K, Cahalin LP, Rundek T, et al. Exercise for cognitive brain health in aging. Neurol Clin Pract. 2018;8(3):257–65.

    PubMed  PubMed Central  Google Scholar 

  78. Karssemeijer EGA, Aaronson JA, Bossers WJ, Smits T, Rikkert MGM, Kessels RPC. Positive effects of combined cognitive and physical exercise training on cognitive function in older adults with mild cognitive impairment or dementia: a meta-analysis. Ageing Res Rev. 2017 Nov;40:75–83.

    PubMed  Google Scholar 

  79. Song D, Yu DSF, Li PWC, Lei Y. The effectiveness of physical exercise on cognitive and psychological outcomes in individuals with mild cognitive impairment: a systematic review and meta-analysis. Int J Nurs Stud. 2018;79:155–64.

    PubMed  Google Scholar 

  80. Wang C, Yu J-T, Wang H-F, Tan C-C, Meng X-F, Tan L. Non-pharmacological interventions for patients with mild cognitive impairment: a meta-analysis of randomized controlled trials of cognition-based and exercise interventions. J Alzheimers Dis. 2014 Aug 28;42(2):663–78.

    PubMed  Google Scholar 

  81. Najafi B, Helbostad JL, Moe-Nilssen R, Zijlstra W, Aminian K. Does walking strategy in older people change as a function of walking distance? Gait Posture. 2009;29:261–6.

    PubMed  Google Scholar 

Download references

Acknowledgements

Not Applicable.

Funding

Brendon Stubbs is supported by a Clinical Lectureship (ICA-CL-2017-03-001) jointly funded by Health Education England (HEE) and the National Institute for Health Research (NIHR). Brendon Stubbs is part funded by the NIHR Biomedical Research Centre at South London and Maudsley NHS Foundation Trust. Brendon Stubbs is also supported by the Maudsley Charity, King’s College London and the NIHR South London Collaboration for Leadership in Applied Health Research and Care (CLAHRC) funding. This paper presents independent research. The views expressed in this publication are those of the authors and not necessarily those of the acknowledged institutions.

María Rosa Bernal-López was supported by “Miguel Servet Type I” program (CP15/00028) from the ISCIII-Madrid (Spain), cofinanced by the Fondo Europeo de Desarrollo Regional-FEDER.

Author information

Authors and Affiliations

Authors

Contributions

IJ-FA and A-CV contributed to the conception of this study. IJ-FA and A-CV were involved in the selection of the included studies. IJ-FA, A-CV, B-S, LM-PB, MR-BL and R-GH were involved in the writing and in the review of the manuscript. The authors have read and approved the manuscript.

Corresponding author

Correspondence to Antonio Cuesta-Vargas.

Ethics declarations

Ethics approval and consent to participate

Not Applicable-this manuscript does not report on or involve the use of any animal or human data or tissue.

Consent for publication

Not Applicable-this manuscript does not contain data from any individual person.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

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

Supplementary information

Additional file 1 Supplementary Appendix A. PRISMA Checklist

. It includes the PRISMA checklist with the item reported in this trial.

Additional file 2 Supplementary Appendix B. Search Strategy.

It includes the complete search strategy which was carried out with all search terms.

Additional file 3 Supplementary Appendix C. Checklist for selection of studies.

It shows a checklist conducted to select studies which could be included in the present trial.

Additional file 4 Supplementary Appendix D. Excluded studies in the second screening.

It contains references of excluded studies in the second screening since they did not meet inclusion criteria.

Additional file 5 Supplementary Appendix E. Conflict of interest of included studies.

It shows the conflict of interests of included studies in the results.

Additional file 6 Supplementary Table 1. Methodological Quality of included RCT (The Cochrane Collaboration’s tool).

It includes the risk of bias of RCTs included in the manuscript.

Additional file 7 Supplementary Table 2. Methodological Quality of included prospective longitudinal studies (The Newcastle Ottawa Scale (NOS)).

It shows the risk of bias of included prospective non-randomized longitudinal studies.

Additional file 8 Supplementary Table 3. Summary of findings and Quality of evidence assessment (GRADE).

It contains the quality of the evidence of each outcome informed at least by two studies with the same design based on the GRADE criteria.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fuentes-Abolafio, I.J., Stubbs, B., Pérez-Belmonte, L.M. et al. Functional parameters indicative of mild cognitive impairment: a systematic review using instrumented kinematic assessment. BMC Geriatr 20, 282 (2020). https://doi.org/10.1186/s12877-020-01678-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12877-020-01678-6

Keywords