U.S. flag

An official website of the United States government

The .gov means it’s official. Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

The site is secure. The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

  • Publications
  • Account settings

Preview improvements coming to the PMC website in October 2024. Learn More or Try it out now .

  • Advanced Search
  • Journal List
  • Korean J Anesthesiol
  • v.71(2); 2018 Apr

Introduction to systematic review and meta-analysis

1 Department of Anesthesiology and Pain Medicine, Inje University Seoul Paik Hospital, Seoul, Korea

2 Department of Anesthesiology and Pain Medicine, Chung-Ang University College of Medicine, Seoul, Korea

Systematic reviews and meta-analyses present results by combining and analyzing data from different studies conducted on similar research topics. In recent years, systematic reviews and meta-analyses have been actively performed in various fields including anesthesiology. These research methods are powerful tools that can overcome the difficulties in performing large-scale randomized controlled trials. However, the inclusion of studies with any biases or improperly assessed quality of evidence in systematic reviews and meta-analyses could yield misleading results. Therefore, various guidelines have been suggested for conducting systematic reviews and meta-analyses to help standardize them and improve their quality. Nonetheless, accepting the conclusions of many studies without understanding the meta-analysis can be dangerous. Therefore, this article provides an easy introduction to clinicians on performing and understanding meta-analyses.

Introduction

A systematic review collects all possible studies related to a given topic and design, and reviews and analyzes their results [ 1 ]. During the systematic review process, the quality of studies is evaluated, and a statistical meta-analysis of the study results is conducted on the basis of their quality. A meta-analysis is a valid, objective, and scientific method of analyzing and combining different results. Usually, in order to obtain more reliable results, a meta-analysis is mainly conducted on randomized controlled trials (RCTs), which have a high level of evidence [ 2 ] ( Fig. 1 ). Since 1999, various papers have presented guidelines for reporting meta-analyses of RCTs. Following the Quality of Reporting of Meta-analyses (QUORUM) statement [ 3 ], and the appearance of registers such as Cochrane Library’s Methodology Register, a large number of systematic literature reviews have been registered. In 2009, the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement [ 4 ] was published, and it greatly helped standardize and improve the quality of systematic reviews and meta-analyses [ 5 ].

An external file that holds a picture, illustration, etc.
Object name is kjae-2018-71-2-103f1.jpg

Levels of evidence.

In anesthesiology, the importance of systematic reviews and meta-analyses has been highlighted, and they provide diagnostic and therapeutic value to various areas, including not only perioperative management but also intensive care and outpatient anesthesia [6–13]. Systematic reviews and meta-analyses include various topics, such as comparing various treatments of postoperative nausea and vomiting [ 14 , 15 ], comparing general anesthesia and regional anesthesia [ 16 – 18 ], comparing airway maintenance devices [ 8 , 19 ], comparing various methods of postoperative pain control (e.g., patient-controlled analgesia pumps, nerve block, or analgesics) [ 20 – 23 ], comparing the precision of various monitoring instruments [ 7 ], and meta-analysis of dose-response in various drugs [ 12 ].

Thus, literature reviews and meta-analyses are being conducted in diverse medical fields, and the aim of highlighting their importance is to help better extract accurate, good quality data from the flood of data being produced. However, a lack of understanding about systematic reviews and meta-analyses can lead to incorrect outcomes being derived from the review and analysis processes. If readers indiscriminately accept the results of the many meta-analyses that are published, incorrect data may be obtained. Therefore, in this review, we aim to describe the contents and methods used in systematic reviews and meta-analyses in a way that is easy to understand for future authors and readers of systematic review and meta-analysis.

Study Planning

It is easy to confuse systematic reviews and meta-analyses. A systematic review is an objective, reproducible method to find answers to a certain research question, by collecting all available studies related to that question and reviewing and analyzing their results. A meta-analysis differs from a systematic review in that it uses statistical methods on estimates from two or more different studies to form a pooled estimate [ 1 ]. Following a systematic review, if it is not possible to form a pooled estimate, it can be published as is without progressing to a meta-analysis; however, if it is possible to form a pooled estimate from the extracted data, a meta-analysis can be attempted. Systematic reviews and meta-analyses usually proceed according to the flowchart presented in Fig. 2 . We explain each of the stages below.

An external file that holds a picture, illustration, etc.
Object name is kjae-2018-71-2-103f2.jpg

Flowchart illustrating a systematic review.

Formulating research questions

A systematic review attempts to gather all available empirical research by using clearly defined, systematic methods to obtain answers to a specific question. A meta-analysis is the statistical process of analyzing and combining results from several similar studies. Here, the definition of the word “similar” is not made clear, but when selecting a topic for the meta-analysis, it is essential to ensure that the different studies present data that can be combined. If the studies contain data on the same topic that can be combined, a meta-analysis can even be performed using data from only two studies. However, study selection via a systematic review is a precondition for performing a meta-analysis, and it is important to clearly define the Population, Intervention, Comparison, Outcomes (PICO) parameters that are central to evidence-based research. In addition, selection of the research topic is based on logical evidence, and it is important to select a topic that is familiar to readers without clearly confirmed the evidence [ 24 ].

Protocols and registration

In systematic reviews, prior registration of a detailed research plan is very important. In order to make the research process transparent, primary/secondary outcomes and methods are set in advance, and in the event of changes to the method, other researchers and readers are informed when, how, and why. Many studies are registered with an organization like PROSPERO ( http://www.crd.york.ac.uk/PROSPERO/ ), and the registration number is recorded when reporting the study, in order to share the protocol at the time of planning.

Defining inclusion and exclusion criteria

Information is included on the study design, patient characteristics, publication status (published or unpublished), language used, and research period. If there is a discrepancy between the number of patients included in the study and the number of patients included in the analysis, this needs to be clearly explained while describing the patient characteristics, to avoid confusing the reader.

Literature search and study selection

In order to secure proper basis for evidence-based research, it is essential to perform a broad search that includes as many studies as possible that meet the inclusion and exclusion criteria. Typically, the three bibliographic databases Medline, Embase, and Cochrane Central Register of Controlled Trials (CENTRAL) are used. In domestic studies, the Korean databases KoreaMed, KMBASE, and RISS4U may be included. Effort is required to identify not only published studies but also abstracts, ongoing studies, and studies awaiting publication. Among the studies retrieved in the search, the researchers remove duplicate studies, select studies that meet the inclusion/exclusion criteria based on the abstracts, and then make the final selection of studies based on their full text. In order to maintain transparency and objectivity throughout this process, study selection is conducted independently by at least two investigators. When there is a inconsistency in opinions, intervention is required via debate or by a third reviewer. The methods for this process also need to be planned in advance. It is essential to ensure the reproducibility of the literature selection process [ 25 ].

Quality of evidence

However, well planned the systematic review or meta-analysis is, if the quality of evidence in the studies is low, the quality of the meta-analysis decreases and incorrect results can be obtained [ 26 ]. Even when using randomized studies with a high quality of evidence, evaluating the quality of evidence precisely helps determine the strength of recommendations in the meta-analysis. One method of evaluating the quality of evidence in non-randomized studies is the Newcastle-Ottawa Scale, provided by the Ottawa Hospital Research Institute 1) . However, we are mostly focusing on meta-analyses that use randomized studies.

If the Grading of Recommendations, Assessment, Development and Evaluations (GRADE) system ( http://www.gradeworkinggroup.org/ ) is used, the quality of evidence is evaluated on the basis of the study limitations, inaccuracies, incompleteness of outcome data, indirectness of evidence, and risk of publication bias, and this is used to determine the strength of recommendations [ 27 ]. As shown in Table 1 , the study limitations are evaluated using the “risk of bias” method proposed by Cochrane 2) . This method classifies bias in randomized studies as “low,” “high,” or “unclear” on the basis of the presence or absence of six processes (random sequence generation, allocation concealment, blinding participants or investigators, incomplete outcome data, selective reporting, and other biases) [ 28 ].

The Cochrane Collaboration’s Tool for Assessing the Risk of Bias [ 28 ]

Data extraction

Two different investigators extract data based on the objectives and form of the study; thereafter, the extracted data are reviewed. Since the size and format of each variable are different, the size and format of the outcomes are also different, and slight changes may be required when combining the data [ 29 ]. If there are differences in the size and format of the outcome variables that cause difficulties combining the data, such as the use of different evaluation instruments or different evaluation timepoints, the analysis may be limited to a systematic review. The investigators resolve differences of opinion by debate, and if they fail to reach a consensus, a third-reviewer is consulted.

Data Analysis

The aim of a meta-analysis is to derive a conclusion with increased power and accuracy than what could not be able to achieve in individual studies. Therefore, before analysis, it is crucial to evaluate the direction of effect, size of effect, homogeneity of effects among studies, and strength of evidence [ 30 ]. Thereafter, the data are reviewed qualitatively and quantitatively. If it is determined that the different research outcomes cannot be combined, all the results and characteristics of the individual studies are displayed in a table or in a descriptive form; this is referred to as a qualitative review. A meta-analysis is a quantitative review, in which the clinical effectiveness is evaluated by calculating the weighted pooled estimate for the interventions in at least two separate studies.

The pooled estimate is the outcome of the meta-analysis, and is typically explained using a forest plot ( Figs. 3 and ​ and4). 4 ). The black squares in the forest plot are the odds ratios (ORs) and 95% confidence intervals in each study. The area of the squares represents the weight reflected in the meta-analysis. The black diamond represents the OR and 95% confidence interval calculated across all the included studies. The bold vertical line represents a lack of therapeutic effect (OR = 1); if the confidence interval includes OR = 1, it means no significant difference was found between the treatment and control groups.

An external file that holds a picture, illustration, etc.
Object name is kjae-2018-71-2-103f3.jpg

Forest plot analyzed by two different models using the same data. (A) Fixed-effect model. (B) Random-effect model. The figure depicts individual trials as filled squares with the relative sample size and the solid line as the 95% confidence interval of the difference. The diamond shape indicates the pooled estimate and uncertainty for the combined effect. The vertical line indicates the treatment group shows no effect (OR = 1). Moreover, if the confidence interval includes 1, then the result shows no evidence of difference between the treatment and control groups.

An external file that holds a picture, illustration, etc.
Object name is kjae-2018-71-2-103f4.jpg

Forest plot representing homogeneous data.

Dichotomous variables and continuous variables

In data analysis, outcome variables can be considered broadly in terms of dichotomous variables and continuous variables. When combining data from continuous variables, the mean difference (MD) and standardized mean difference (SMD) are used ( Table 2 ).

Summary of Meta-analysis Methods Available in RevMan [ 28 ]

The MD is the absolute difference in mean values between the groups, and the SMD is the mean difference between groups divided by the standard deviation. When results are presented in the same units, the MD can be used, but when results are presented in different units, the SMD should be used. When the MD is used, the combined units must be shown. A value of “0” for the MD or SMD indicates that the effects of the new treatment method and the existing treatment method are the same. A value lower than “0” means the new treatment method is less effective than the existing method, and a value greater than “0” means the new treatment is more effective than the existing method.

When combining data for dichotomous variables, the OR, risk ratio (RR), or risk difference (RD) can be used. The RR and RD can be used for RCTs, quasi-experimental studies, or cohort studies, and the OR can be used for other case-control studies or cross-sectional studies. However, because the OR is difficult to interpret, using the RR and RD, if possible, is recommended. If the outcome variable is a dichotomous variable, it can be presented as the number needed to treat (NNT), which is the minimum number of patients who need to be treated in the intervention group, compared to the control group, for a given event to occur in at least one patient. Based on Table 3 , in an RCT, if x is the probability of the event occurring in the control group and y is the probability of the event occurring in the intervention group, then x = c/(c + d), y = a/(a + b), and the absolute risk reduction (ARR) = x − y. NNT can be obtained as the reciprocal, 1/ARR.

Calculation of the Number Needed to Treat in the Dichotomous table

Fixed-effect models and random-effect models

In order to analyze effect size, two types of models can be used: a fixed-effect model or a random-effect model. A fixed-effect model assumes that the effect of treatment is the same, and that variation between results in different studies is due to random error. Thus, a fixed-effect model can be used when the studies are considered to have the same design and methodology, or when the variability in results within a study is small, and the variance is thought to be due to random error. Three common methods are used for weighted estimation in a fixed-effect model: 1) inverse variance-weighted estimation 3) , 2) Mantel-Haenszel estimation 4) , and 3) Peto estimation 5) .

A random-effect model assumes heterogeneity between the studies being combined, and these models are used when the studies are assumed different, even if a heterogeneity test does not show a significant result. Unlike a fixed-effect model, a random-effect model assumes that the size of the effect of treatment differs among studies. Thus, differences in variation among studies are thought to be due to not only random error but also between-study variability in results. Therefore, weight does not decrease greatly for studies with a small number of patients. Among methods for weighted estimation in a random-effect model, the DerSimonian and Laird method 6) is mostly used for dichotomous variables, as the simplest method, while inverse variance-weighted estimation is used for continuous variables, as with fixed-effect models. These four methods are all used in Review Manager software (The Cochrane Collaboration, UK), and are described in a study by Deeks et al. [ 31 ] ( Table 2 ). However, when the number of studies included in the analysis is less than 10, the Hartung-Knapp-Sidik-Jonkman method 7) can better reduce the risk of type 1 error than does the DerSimonian and Laird method [ 32 ].

Fig. 3 shows the results of analyzing outcome data using a fixed-effect model (A) and a random-effect model (B). As shown in Fig. 3 , while the results from large studies are weighted more heavily in the fixed-effect model, studies are given relatively similar weights irrespective of study size in the random-effect model. Although identical data were being analyzed, as shown in Fig. 3 , the significant result in the fixed-effect model was no longer significant in the random-effect model. One representative example of the small study effect in a random-effect model is the meta-analysis by Li et al. [ 33 ]. In a large-scale study, intravenous injection of magnesium was unrelated to acute myocardial infarction, but in the random-effect model, which included numerous small studies, the small study effect resulted in an association being found between intravenous injection of magnesium and myocardial infarction. This small study effect can be controlled for by using a sensitivity analysis, which is performed to examine the contribution of each of the included studies to the final meta-analysis result. In particular, when heterogeneity is suspected in the study methods or results, by changing certain data or analytical methods, this method makes it possible to verify whether the changes affect the robustness of the results, and to examine the causes of such effects [ 34 ].

Heterogeneity

Homogeneity test is a method whether the degree of heterogeneity is greater than would be expected to occur naturally when the effect size calculated from several studies is higher than the sampling error. This makes it possible to test whether the effect size calculated from several studies is the same. Three types of homogeneity tests can be used: 1) forest plot, 2) Cochrane’s Q test (chi-squared), and 3) Higgins I 2 statistics. In the forest plot, as shown in Fig. 4 , greater overlap between the confidence intervals indicates greater homogeneity. For the Q statistic, when the P value of the chi-squared test, calculated from the forest plot in Fig. 4 , is less than 0.1, it is considered to show statistical heterogeneity and a random-effect can be used. Finally, I 2 can be used [ 35 ].

I 2 , calculated as shown above, returns a value between 0 and 100%. A value less than 25% is considered to show strong homogeneity, a value of 50% is average, and a value greater than 75% indicates strong heterogeneity.

Even when the data cannot be shown to be homogeneous, a fixed-effect model can be used, ignoring the heterogeneity, and all the study results can be presented individually, without combining them. However, in many cases, a random-effect model is applied, as described above, and a subgroup analysis or meta-regression analysis is performed to explain the heterogeneity. In a subgroup analysis, the data are divided into subgroups that are expected to be homogeneous, and these subgroups are analyzed. This needs to be planned in the predetermined protocol before starting the meta-analysis. A meta-regression analysis is similar to a normal regression analysis, except that the heterogeneity between studies is modeled. This process involves performing a regression analysis of the pooled estimate for covariance at the study level, and so it is usually not considered when the number of studies is less than 10. Here, univariate and multivariate regression analyses can both be considered.

Publication bias

Publication bias is the most common type of reporting bias in meta-analyses. This refers to the distortion of meta-analysis outcomes due to the higher likelihood of publication of statistically significant studies rather than non-significant studies. In order to test the presence or absence of publication bias, first, a funnel plot can be used ( Fig. 5 ). Studies are plotted on a scatter plot with effect size on the x-axis and precision or total sample size on the y-axis. If the points form an upside-down funnel shape, with a broad base that narrows towards the top of the plot, this indicates the absence of a publication bias ( Fig. 5A ) [ 29 , 36 ]. On the other hand, if the plot shows an asymmetric shape, with no points on one side of the graph, then publication bias can be suspected ( Fig. 5B ). Second, to test publication bias statistically, Begg and Mazumdar’s rank correlation test 8) [ 37 ] or Egger’s test 9) [ 29 ] can be used. If publication bias is detected, the trim-and-fill method 10) can be used to correct the bias [ 38 ]. Fig. 6 displays results that show publication bias in Egger’s test, which has then been corrected using the trim-and-fill method using Comprehensive Meta-Analysis software (Biostat, USA).

An external file that holds a picture, illustration, etc.
Object name is kjae-2018-71-2-103f5.jpg

Funnel plot showing the effect size on the x-axis and sample size on the y-axis as a scatter plot. (A) Funnel plot without publication bias. The individual plots are broader at the bottom and narrower at the top. (B) Funnel plot with publication bias. The individual plots are located asymmetrically.

An external file that holds a picture, illustration, etc.
Object name is kjae-2018-71-2-103f6.jpg

Funnel plot adjusted using the trim-and-fill method. White circles: comparisons included. Black circles: inputted comparisons using the trim-and-fill method. White diamond: pooled observed log risk ratio. Black diamond: pooled inputted log risk ratio.

Result Presentation

When reporting the results of a systematic review or meta-analysis, the analytical content and methods should be described in detail. First, a flowchart is displayed with the literature search and selection process according to the inclusion/exclusion criteria. Second, a table is shown with the characteristics of the included studies. A table should also be included with information related to the quality of evidence, such as GRADE ( Table 4 ). Third, the results of data analysis are shown in a forest plot and funnel plot. Fourth, if the results use dichotomous data, the NNT values can be reported, as described above.

The GRADE Evidence Quality for Each Outcome

N: number of studies, ROB: risk of bias, PON: postoperative nausea, POV: postoperative vomiting, PONV: postoperative nausea and vomiting, CI: confidence interval, RR: risk ratio, AR: absolute risk.

When Review Manager software (The Cochrane Collaboration, UK) is used for the analysis, two types of P values are given. The first is the P value from the z-test, which tests the null hypothesis that the intervention has no effect. The second P value is from the chi-squared test, which tests the null hypothesis for a lack of heterogeneity. The statistical result for the intervention effect, which is generally considered the most important result in meta-analyses, is the z-test P value.

A common mistake when reporting results is, given a z-test P value greater than 0.05, to say there was “no statistical significance” or “no difference.” When evaluating statistical significance in a meta-analysis, a P value lower than 0.05 can be explained as “a significant difference in the effects of the two treatment methods.” However, the P value may appear non-significant whether or not there is a difference between the two treatment methods. In such a situation, it is better to announce “there was no strong evidence for an effect,” and to present the P value and confidence intervals. Another common mistake is to think that a smaller P value is indicative of a more significant effect. In meta-analyses of large-scale studies, the P value is more greatly affected by the number of studies and patients included, rather than by the significance of the results; therefore, care should be taken when interpreting the results of a meta-analysis.

When performing a systematic literature review or meta-analysis, if the quality of studies is not properly evaluated or if proper methodology is not strictly applied, the results can be biased and the outcomes can be incorrect. However, when systematic reviews and meta-analyses are properly implemented, they can yield powerful results that could usually only be achieved using large-scale RCTs, which are difficult to perform in individual studies. As our understanding of evidence-based medicine increases and its importance is better appreciated, the number of systematic reviews and meta-analyses will keep increasing. However, indiscriminate acceptance of the results of all these meta-analyses can be dangerous, and hence, we recommend that their results be received critically on the basis of a more accurate understanding.

1) http://www.ohri.ca .

2) http://methods.cochrane.org/bias/assessing-risk-bias-included-studies .

3) The inverse variance-weighted estimation method is useful if the number of studies is small with large sample sizes.

4) The Mantel-Haenszel estimation method is useful if the number of studies is large with small sample sizes.

5) The Peto estimation method is useful if the event rate is low or one of the two groups shows zero incidence.

6) The most popular and simplest statistical method used in Review Manager and Comprehensive Meta-analysis software.

7) Alternative random-effect model meta-analysis that has more adequate error rates than does the common DerSimonian and Laird method, especially when the number of studies is small. However, even with the Hartung-Knapp-Sidik-Jonkman method, when there are less than five studies with very unequal sizes, extra caution is needed.

8) The Begg and Mazumdar rank correlation test uses the correlation between the ranks of effect sizes and the ranks of their variances [ 37 ].

9) The degree of funnel plot asymmetry as measured by the intercept from the regression of standard normal deviates against precision [ 29 ].

10) If there are more small studies on one side, we expect the suppression of studies on the other side. Trimming yields the adjusted effect size and reduces the variance of the effects by adding the original studies back into the analysis as a mirror image of each study.

Log in using your username and password

  • Search More Search for this keyword Advanced search
  • Latest content
  • Current issue
  • Write for Us
  • BMJ Journals More You are viewing from: Google Indexer

You are here

  • Volume 14, Issue 3
  • What is a systematic review?
  • Article Text
  • Article info
  • Citation Tools
  • Rapid Responses
  • Article metrics

Download PDF

  • Jane Clarke
  • Correspondence to Jane Clarke 4 Prime Road, Grey Lynn, Auckland, New Zealand; janeclarkehome{at}gmail.com

https://doi.org/10.1136/ebn.2011.0049

Statistics from Altmetric.com

Request permissions.

If you wish to reuse any or all of this article please use the link below which will take you to the Copyright Clearance Center’s RightsLink service. You will be able to get a quick price and instant permission to reuse the content in many different ways.

A high-quality systematic review is described as the most reliable source of evidence to guide clinical practice. The purpose of a systematic review is to deliver a meticulous summary of all the available primary research in response to a research question. A systematic review uses all the existing research and is sometime called ‘secondary research’ (research on research). They are often required by research funders to establish the state of existing knowledge and are frequently used in guideline development. Systematic review findings are often used within the healthcare setting but may be applied elsewhere. For example, the Campbell Collaboration advocates the application of systematic reviews for policy-making in education, justice and social work.

Systematic reviews can be conducted on all types of primary research. Many are reviews of randomised trials (addressing questions of effectiveness), cross-sectional studies (addressing questions about prevalence or diagnostic accuracy, for example) or cohort studies (addressing questions about prognosis). When qualitative research is reviewed systematically, it may be described as a systematic review, but more often other terms such as meta-synthesis are used.

Systematic review methodology is explicit and precise and aims to minimise bias, thus enhancing the reliability of the conclusions drawn. 1 , 2 The features of a systematic review include:

■ clear aims with predetermined eligibility and relevance criteria for studies;

■ transparent, reproducible methods;

■ rigorous search designed to locate all eligible studies;

■ an assessment of the validity of the findings of the included studies and

■ a systematic presentation, and synthesis, of the included studies. 3

The first step in a systematic review is a meticulous search of all sources of evidence for relevant studies. The databases and citation indexes searched are listed in the methodology section of the review. Next, using predetermined reproducible criteria to screen for eligibility and relevance assessment of titles and the abstracts is completed. Each study is then assessed in terms of methodological quality.

Finally, the evidence is synthesised. This process may or may not include a meta-analysis. A meta-analysis is a statistical summary of the findings of independent studies. 4 Meta-analyses can potentially present more precise estimates of the effects of interventions than those derived from the individual studies alone. These strategies are used to limit bias and random error which may arise during this process. Without these safeguards, then, reviews can mislead, such that we gain an unreliable summary of the available knowledge.

The Cochrane Collaboration is a leader in the production of systematic reviews. Cochrane reviews are published on a monthly basis in the Cochrane Database of Systematic Reviews in The Cochrane Library (see: http://www.thecochranelibrary.com ).

  • Antman EM ,
  • Kupelnick B ,
  • Higgins JPT ,

Competing interests None.

Read the full text or download the PDF:

Introduction to Systematic Reviews

  • Reference work entry
  • First Online: 20 July 2022
  • pp 2159–2177
  • Cite this reference work entry

the purpose of a systematic review

  • Tianjing Li 3 ,
  • Ian J. Saldanha 4 &
  • Karen A. Robinson 5  

223 Accesses

A systematic review identifies and synthesizes all relevant studies that fit prespecified criteria to answer a research question. Systematic review methods can be used to answer many types of research questions. The type of question most relevant to trialists is the effects of treatments and is thus the focus of this chapter. We discuss the motivation for and importance of performing systematic reviews and their relevance to trialists. We introduce the key steps in completing a systematic review, including framing the question, searching for and selecting studies, collecting data, assessing risk of bias in included studies, conducting a qualitative synthesis and a quantitative synthesis (i.e., meta-analysis), grading the certainty of evidence, and writing the systematic review report. We also describe how to identify systematic reviews and how to assess their methodological rigor. We discuss the challenges and criticisms of systematic reviews, and how technology and innovations, combined with a closer partnership between trialists and systematic reviewers, can help identify effective and safe evidence-based practices more quickly.

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

Access this chapter

  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
  • Available as EPUB and PDF
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

AHRQ (2015) Methods guide for effectiveness and comparative effectiveness reviews. Available from https://effectivehealthcare.ahrq.gov/products/cer-methods-guide/overview . Accessed on 27 Oct 2019

Andersen MZ, Gülen S, Fonnes S, Andresen K, Rosenberg J (2020) Half of Cochrane reviews were published more than two years after the protocol. J Clin Epidemiol 124:85–93. https://doi.org/10.1016/j.jclinepi.2020.05.011

Article   Google Scholar  

Berkman ND, Lohr KN, Ansari MT, Balk EM, Kane R, McDonagh M, Morton SC, Viswanathan M, Bass EB, Butler M, Gartlehner G, Hartling L, McPheeters M, Morgan LC, Reston J, Sista P, Whitlock E, Chang S (2015) Grading the strength of a body of evidence when assessing health care interventions: an EPC update. J Clin Epidemiol 68(11):1312–1324

Borah R, Brown AW, Capers PL, Kaiser KA (2017) Analysis of the time and workers needed to conduct systematic reviews of medical interventions using data from the PROSPERO registry. BMJ Open 7(2):e012545. https://doi.org/10.1136/bmjopen-2016-012545

Chalmers I, Bracken MB, Djulbegovic B, Garattini S, Grant J, Gülmezoglu AM, Howells DW, Ioannidis JP, Oliver S (2014) How to increase value and reduce waste when research priorities are set. Lancet 383(9912):156–165. https://doi.org/10.1016/S0140-6736(13)62229-1

Clarke M, Chalmers I (1998) Discussion sections in reports of controlled trials published in general medical journals: islands in search of continents? JAMA 280(3):280–282

Cooper NJ, Jones DR, Sutton AJ (2005) The use of systematic reviews when designing studies. Clin Trials 2(3):260–264

Djulbegovic B, Kumar A, Magazin A, Schroen AT, Soares H, Hozo I, Clarke M, Sargent D, Schell MJ (2011) Optimism bias leads to inconclusive results-an empirical study. J Clin Epidemiol 64(6):583–593. https://doi.org/10.1016/j.jclinepi.2010.09.007

Elliott JH, Synnot A, Turner T, Simmonds M, Akl EA, McDonald S, Salanti G, Meerpohl J, MacLehose H, Hilton J, Tovey D, Shemilt I, Thomas J (2017) Living systematic review network. Living systematic review: 1. Introduction-the why, what, when, and how. J Clin Epidemiol 91:23–30

Equator Network. Reporting guidelines for systematic reviews. Available from https://www.equator-network.org/?post_type=eq_guidelines&eq_guidelines_study_design=systematic-reviews-and-meta-analyses&eq_guidelines_clinical_specialty=0&eq_guidelines_report_section=0&s=+ . Accessed 9 Mar 2020

Garner P, Hopewell S, Chandler J, MacLehose H, Schünemann HJ, Akl EA, Beyene J, Chang S, Churchill R, Dearness K, Guyatt G, Lefebvre C, Liles B, Marshall R, Martínez García L, Mavergames C, Nasser M, Qaseem A, Sampson M, Soares-Weiser K, Takwoingi Y, Thabane L, Trivella M, Tugwell P, Welsh E, Wilson EC, Schünemann HJ (2016) Panel for updating guidance for systematic reviews (PUGs). When and how to update systematic reviews: consensus and checklist. BMJ 354:i3507. https://doi.org/10.1136/bmj.i3507 . Erratum in: BMJ 2016 Sep 06 354:i4853

Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J, Norris S, Falck-Ytter Y, Glasziou P, DeBeer H, Jaeschke R, Rind D, Meerpohl J, Dahm P, Schünemann HJ (2011) GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J Clin Epidemiol 64(4):383–394

Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA (eds) (2019a) Cochrane handbook for systematic reviews of interventions, 2nd edn. Wiley, Chichester

Google Scholar  

Higgins JPT, Lasserson T, Chandler J, Tovey D, Thomas J, Flemyng E, Churchill R (2019b) Standards for the conduct of new Cochrane intervention reviews. In: JPT H, Lasserson T, Chandler J, Tovey D, Thomas J, Flemyng E, Churchill R (eds) Methodological expectations of Cochrane intervention reviews. Cochrane, London

IOM (2011) Committee on standards for systematic reviews of comparative effectiveness research, board on health care services. In: Eden J, Levit L, Berg A, Morton S (eds) Finding what works in health care: standards for systematic reviews. National Academies Press, Washington, DC

Jonnalagadda SR, Goyal P, Huffman MD (2015) Automating data extraction in systematic reviews: a systematic review. Syst Rev 4:78

Krnic Martinic M, Pieper D, Glatt A, Puljak L (2019) Definition of a systematic review used in overviews of systematic reviews, meta-epidemiological studies and textbooks. BMC Med Res Methodol 19(1):203. Published 4 Nov 2019. https://doi.org/10.1186/s12874-019-0855-0

Lasserson TJ, Thomas J, Higgins JPT (2019) Chapter 1: Starting a review. In: Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA (eds) Cochrane handbook for systematic reviews of interventions version 6.0 (updated July 2019). Cochrane. Available from www.training.cochrane.org/handbook

Lau J, Antman EM, Jimenez-Silva J, Kupelnick B, Mosteller F, Chalmers TC (1992) Cumulative meta-analysis of therapeutic trials for myocardial infarction. N Engl J Med 327(4):248–254

Lau J (2019) Editorial: systematic review automation thematic series. Syst Rev 8(1):70. Published 11 Mar 2019. https://doi.org/10.1186/s13643-019-0974-z

Liberati A, Altman DG, Tetzlaff J, Mulrow C, Gøtzsche PC, Ioannidis JP, Clarke M, Devereaux PJ, Kleijnen J, Moher D (2009) The PRISMA statement for reporting systematic reviews and meta-analyses of studies that evaluate health care interventions: explanation and elaboration. PLoS Med 6(7):e1000100. https://doi.org/10.1371/journal.pmed.1000100

Lund H, Brunnhuber K, Juhl C, Robinson K, Leenaars M, Dorch BF, Jamtvedt G, Nortvedt MW, Christensen R, Chalmers I (2016) Towards evidence based research. BMJ 355:i5440. https://doi.org/10.1136/bmj.i5440

Marshall IJ, Noel-Storr A, Kuiper J, Thomas J, Wallace BC (2018) Machine learning for identifying randomized controlled trials: an evaluation and practitioner’s guide. Res Synth Methods 9(4):602–614. https://doi.org/10.1002/jrsm.1287

Michelson M, Reuter K (2019) The significant cost of systematic reviews and meta-analyses: a call for greater involvement of machine learning to assess the promise of clinical trials. Contemp Clin Trials Commun 16:100443. https://doi.org/10.1016/j.conctc.2019.100443 . Erratum in: Contemp Clin Trials Commun 2019 16:100450

Moher D, Liberati A, Tetzlaff J (2009) Altman DG; PRISMA group. Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. Ann Intern Med 151(4):264–269. W64

Moher D, Shamseer L, Clarke M, Ghersi D, Liberati A, Petticrew M, Shekelle P, Stewart LA, PRISMA-P Group (2015) Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev 4(1):1. https://doi.org/10.1186/2046-4053-4-1

NIHR HTA Stage 1 guidance notes. Available from https://www.nihr.ac.uk/documents/hta-stage-1-guidance-notes/11743 ; Accessed 10 Mar 2020

Page MJ, Shamseer L, Altman DG, Tetzlaff J, Sampson M, Tricco AC, Catalá-López F, Li L, Reid EK, Sarkis-Onofre R, Moher D (2016) Epidemiology and reporting characteristics of systematic reviews of biomedical research: a cross-sectional study. PLoS Med 13(5):e1002028. https://doi.org/10.1371/journal.pmed.1002028

Page MJ, Higgins JPT, Sterne JAC (2019) Chapter 13: assessing risk of bias due to missing results in a synthesis. In: Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ et al (eds) Cochrane handbook for systematic reviews of interventions, 2nd edn. Wiley, Chichester, pp 349–374

Chapter   Google Scholar  

Robinson KA (2009) Use of prior research in the justification and interpretation of clinical trials. Johns Hopkins University

Robinson KA, Goodman SN (2011) A systematic examination of the citation of prior research in reports of randomized, controlled trials. Ann Intern Med 154(1):50–55. https://doi.org/10.7326/0003-4819-154-1-201101040-00007

Rouse B, Cipriani A, Shi Q, Coleman AL, Dickersin K, Li T (2016) Network meta-analysis for clinical practice guidelines – a case study on first-line medical therapies for primary open-angle glaucoma. Ann Intern Med 164(10):674–682. https://doi.org/10.7326/M15-2367

Saldanha IJ, Lindsley K, Do DV et al (2017) Comparison of clinical trial and systematic review outcomes for the 4 most prevalent eye diseases. JAMA Ophthalmol 135(9):933–940. https://doi.org/10.1001/jamaophthalmol.2017.2583

Shea BJ, Grimshaw JM, Wells GA, Boers M, Andersson N, Hamel C, Porter AC, Tugwell P, Moher D, Bouter LM (2007) Development of AMSTAR: a measurement tool to assess the methodological quality of systematic reviews. BMC Med Res Methodol 7:10

Shea BJ, Reeves BC, Wells G, Thuku M, Hamel C, Moran J, Moher D, Tugwell P, Welch V, Kristjansson E, Henry DA (2017) AMSTAR 2: a critical appraisal tool for systematic reviews that include randomised or non-randomised studies of healthcare interventions, or both. BMJ 358:j4008. https://doi.org/10.1136/bmj.j4008

Shojania KG, Sampson M, Ansari MT, Ji J, Doucette S, Moher D (2007) How quickly do systematic reviews go out of date? A survival analysis. Ann Intern Med 147(4):224–233

Sterne JA, Hernán MA, Reeves BC, Savović J, Berkman ND, Viswanathan M, Henry D, Altman DG, Ansari MT, Boutron I, Carpenter JR, Chan AW, Churchill R, Deeks JJ, Hróbjartsson A, Kirkham J, Jüni P, Loke YK, Pigott TD, Ramsay CR, Regidor D, Rothstein HR, Sandhu L, Santaguida PL, Schünemann HJ, Shea B, Shrier I, Tugwell P, Turner L, Valentine JC, Waddington H, Waters E, Wells GA, Whiting PF, Higgins JP (2016) ROBINS-I: a tool for assessing risk of bias in non-randomised studies of interventions. BMJ 355:i4919. https://doi.org/10.1136/bmj.i4919

Sterne JAC, Savović J, Page MJ, Elbers RG, Blencowe NS, Boutron I, Cates CJ, Cheng HY, Corbett MS, Eldridge SM, Emberson JR, Hernán MA, Hopewell S, Hróbjartsson A, Junqueira DR, Jüni P, Kirkham JJ, Lasserson T, Li T, McAleenan A, Reeves BC, Shepperd S, Shrier I, Stewart LA, Tilling K, White IR, Whiting PF, Higgins JPT (2019) RoB 2: a revised tool for assessing risk of bias in randomised trials. BMJ 366:l4898. https://doi.org/10.1136/bmj.l4898

Thomas J, Kneale D, McKenzie JE, Brennan SE, Bhaumik S (2019) Chapter 2: determining the scope of the review and the questions it will address. In: Higgins JPT, Thomas J, Chandler J, Cumpston M, Li T, Page MJ, Welch VA (eds) Cochrane handbook for systematic reviews of interventions version 6.0 (updated July 2019). Cochrane. Available from www.training.cochrane.org/handbook

USPSTF U.S. Preventive Services Task Force Procedure Manual (2017). Available from: https://www.uspreventiveservicestaskforce.org/uspstf/sites/default/files/inline-files/procedure-manual2017_update.pdf . Accessed 21 May 2020

Whitaker (2015) UCSF guides: systematic review: when will i be finished? https://guides.ucsf.edu/c.php?g=375744&p=3041343 , Accessed 13 May 2020

Whiting P, Savović J, Higgins JP, Caldwell DM, Reeves BC, Shea B, Davies P, Kleijnen J (2016) Churchill R; ROBIS group. ROBIS: a new tool to assess risk of bias in systematic reviews was developed. J Clin Epidemiol 69:225–234. https://doi.org/10.1016/j.jclinepi.2015.06.005

Download references

Author information

Authors and affiliations.

Department of Ophthalmology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA

Tianjing Li

Department of Health Services, Policy, and Practice and Department of Epidemiology, Brown University School of Public Health, Providence, RI, USA

Ian J. Saldanha

Department of Medicine, Johns Hopkins University, Baltimore, MD, USA

Karen A. Robinson

You can also search for this author in PubMed   Google Scholar

Corresponding author

Correspondence to Tianjing Li .

Editor information

Editors and affiliations.

Department of Surgery, Division of Surgical Oncology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA

Steven Piantadosi

Department of Epidemiology, School of Public Health, Johns Hopkins University, Baltimore, MD, USA

Curtis L. Meinert

Section Editor information

Department of Epidemiology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO, USA

The Johns Hopkins Center for Clinical Trials and Evidence Synthesis, Johns Hopkins University, Baltimore, MD, USA

Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA

Rights and permissions

Reprints and permissions

Copyright information

© 2022 Springer Nature Switzerland AG

About this entry

Cite this entry.

Li, T., Saldanha, I.J., Robinson, K.A. (2022). Introduction to Systematic Reviews. In: Piantadosi, S., Meinert, C.L. (eds) Principles and Practice of Clinical Trials. Springer, Cham. https://doi.org/10.1007/978-3-319-52636-2_194

Download citation

DOI : https://doi.org/10.1007/978-3-319-52636-2_194

Published : 20 July 2022

Publisher Name : Springer, Cham

Print ISBN : 978-3-319-52635-5

Online ISBN : 978-3-319-52636-2

eBook Packages : Mathematics and Statistics Reference Module Computer Science and Engineering

Share this entry

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Publish with us

Policies and ethics

  • Find a journal
  • Track your research

Jump to navigation

  • Bahasa Malaysia

Home

What are systematic reviews?

Watch this video from   Cochrane Consumers and Communication to learn what systematic reviews are, how researchers prepare them, and why they’re an important part of making informed decisions about health - for everyone. 

Cochrane evidence, including our systematic reviews, provides a powerful tool to enhance your healthcare knowledge and decision making. This video from Cochrane Sweden explains a bit about how we create health evidence and what Cochrane does. 

  • Search our Plain Language Summaries of health evidence
  • Learn more about Cochrane and our work

X

Library Services

UCL LIBRARY SERVICES

  • Guides and databases
  • Library skills
  • Systematic reviews

What are systematic reviews?

  • Types of systematic reviews
  • Formulating a research question
  • Identifying studies
  • Searching databases
  • Describing and appraising studies
  • Synthesis and systematic maps
  • Software for systematic reviews
  • Online training and support
  • Live and face to face training
  • Individual support
  • Further help

Searching for information

Systematic reviews are a type of literature review of research which require equivalent standards of rigour as primary research. They have a clear, logical rationale that is reported to the reader of the review. They are used in research and policymaking to inform evidence-based decisions and practice. They differ from traditional literature reviews particularly in the following elements of conduct and reporting.

Systematic reviews: 

  • use explicit and transparent methods
  • are a piece of research following a standard set of stages
  • are accountable, replicable and updateable
  • involve users to ensure a review is relevant and useful.

For example, systematic reviews (like all research) should have a clear research question, and the perspective of the authors in their approach to addressing the question is described. There are clearly described methods on how each study in a review was identified, how that study was appraised for quality and relevance and how it is combined with other studies in order to address the review question. A systematic review usually involves more than one person in order to increase the objectivity and trustworthiness of the reviews methods and findings.

Research protocols for systematic reviews may be peer-reviewed and published or registered in a suitable repository to help avoid duplication of reviews and for comparisons to be made with the final review and the planned review.

  • History of systematic reviews to inform policy (EPPI-Centre)
  • Six reasons why it is important to be systematic (EPPI-Centre)
  • Evidence Synthesis International (ESI): Position Statement Describes the issues, principles and goals in synthesising research evidence to inform policy, practice and decisions

On this page

Should all literature reviews be 'systematic reviews', different methods for systematic reviews, reporting standards for systematic reviews.

Literature reviews provide a more complete picture of research knowledge than is possible from individual pieces of research. This can be used to: clarify what is known from research, provide new perspectives, build theory, test theory, identify research gaps or inform research agendas.

A systematic review requires a considerable amount of time and resources, and is one type of literature review.

If the purpose of a review is to make justifiable evidence claims, then it should be systematic, as a systematic review uses rigorous explicit methods. The methods used can depend on the purpose of the review, and the time and resources available.

A 'non-systematic review' might use some of the same methods as systematic reviews, such as systematic approaches to identify studies or quality appraise the literature. There may be times when this approach can be useful. In a student dissertation, for example, there may not be the time to be fully systematic in a review of the literature if this is only one small part of the thesis. In other types of research, there may also be a need to obtain a quick and not necessarily thorough overview of a literature to inform some other work (including a systematic review). Another example, is where policymakers, or other people using research findings, want to make quick decisions and there is no systematic review available to help them. They have a choice of gaining a rapid overview of the research literature or not having any research evidence to help their decision-making. 

Just like any other piece of research, the methods used to undertake any literature review should be carefully planned to justify the conclusions made. 

Finding out about different types of systematic reviews and the methods used for systematic reviews, and reading both systematic and other types of review will help to understand some of the differences. 

Typically, a systematic review addresses a focussed, structured research question in order to inform understanding and decisions on an area. (see the  Formulating a research question  section for examples). 

Sometimes systematic reviews ask a broad research question, and one strategy to achieve this is the use of several focussed sub-questions each addressed by sub-components of the review.  

Another strategy is to develop a map to describe the type of research that has been undertaken in relation to a research question. Some maps even describe over 2,000 papers, while others are much smaller. One purpose of a map is to help choose a sub-set of studies to explore more fully in a synthesis. There are also other purposes of maps: see the box on  systematic evidence maps  for further information. 

Reporting standards specify minimum elements that need to go into the reporting of a review. The reporting standards refer mainly to methodological issues but they are not as detailed or specific as critical appraisal for the methodological standards of conduct of a review.

A number of organisations have developed specific guidelines and standards for both the conducting and reporting on systematic reviews in different topic areas.  

  • PRISMA PRISMA is a reporting standard and is an acronym for Preferred Reporting Items for Systematic Reviews and Meta-Analyses. The Key Documents section of the PRISMA website links to a checklist, flow diagram and explanatory notes. PRISMA is less useful for certain types of reviews, including those that are iterative.
  • eMERGe eMERGe is a reporting standard that has been developed for meta-ethnographies, a qualitative synthesis method.
  • ROSES: RepOrting standards for Systematic Evidence Syntheses Reporting standards, including forms and flow diagram, designed specifically for systematic reviews and maps in the field of conservation and environmental management.

Useful books about systematic reviews

the purpose of a systematic review

Systematic approaches to a successful literature review

the purpose of a systematic review

An introduction to systematic reviews

the purpose of a systematic review

Cochrane handbook for systematic reviews of interventions

Systematic reviews: crd's guidance for undertaking reviews in health care.

the purpose of a systematic review

Finding what works in health care: Standards for systematic reviews

Book cover image

Systematic Reviews in the Social Sciences

Meta-analysis and research synthesis.

Book cover image

Research Synthesis and Meta-Analysis

Book cover image

Doing a Systematic Review

Literature reviews.

  • What is a literature review?
  • Why are literature reviews important?
  • << Previous: Systematic reviews
  • Next: Types of systematic reviews >>
  • Last Updated: Apr 4, 2024 10:09 AM
  • URL: https://library-guides.ucl.ac.uk/systematic-reviews
  • - Google Chrome

Intended for healthcare professionals

  • Access provided by Google Indexer
  • My email alerts
  • BMA member login
  • Username * Password * Forgot your log in details? Need to activate BMA Member Log In Log in via OpenAthens Log in via your institution

Home

Search form

  • Advanced search
  • Search responses
  • Search blogs
  • The PRISMA 2020...

The PRISMA 2020 statement: an updated guideline for reporting systematic reviews

PRISMA 2020 explanation and elaboration: updated guidance and exemplars for reporting systematic reviews

  • Related content
  • Peer review
  • Matthew J Page , senior research fellow 1 ,
  • Joanne E McKenzie , associate professor 1 ,
  • Patrick M Bossuyt , professor 2 ,
  • Isabelle Boutron , professor 3 ,
  • Tammy C Hoffmann , professor 4 ,
  • Cynthia D Mulrow , professor 5 ,
  • Larissa Shamseer , doctoral student 6 ,
  • Jennifer M Tetzlaff , research product specialist 7 ,
  • Elie A Akl , professor 8 ,
  • Sue E Brennan , senior research fellow 1 ,
  • Roger Chou , professor 9 ,
  • Julie Glanville , associate director 10 ,
  • Jeremy M Grimshaw , professor 11 ,
  • Asbjørn Hróbjartsson , professor 12 ,
  • Manoj M Lalu , associate scientist and assistant professor 13 ,
  • Tianjing Li , associate professor 14 ,
  • Elizabeth W Loder , professor 15 ,
  • Evan Mayo-Wilson , associate professor 16 ,
  • Steve McDonald , senior research fellow 1 ,
  • Luke A McGuinness , research associate 17 ,
  • Lesley A Stewart , professor and director 18 ,
  • James Thomas , professor 19 ,
  • Andrea C Tricco , scientist and associate professor 20 ,
  • Vivian A Welch , associate professor 21 ,
  • Penny Whiting , associate professor 17 ,
  • David Moher , director and professor 22
  • 1 School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
  • 2 Department of Clinical Epidemiology, Biostatistics and Bioinformatics, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam, Netherlands
  • 3 Université de Paris, Centre of Epidemiology and Statistics (CRESS), Inserm, F 75004 Paris, France
  • 4 Institute for Evidence-Based Healthcare, Faculty of Health Sciences and Medicine, Bond University, Gold Coast, Australia
  • 5 University of Texas Health Science Center at San Antonio, San Antonio, Texas, USA; Annals of Internal Medicine
  • 6 Knowledge Translation Program, Li Ka Shing Knowledge Institute, Toronto, Canada; School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Canada
  • 7 Evidence Partners, Ottawa, Canada
  • 8 Clinical Research Institute, American University of Beirut, Beirut, Lebanon; Department of Health Research Methods, Evidence, and Impact, McMaster University, Hamilton, Ontario, Canada
  • 9 Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, Oregon, USA
  • 10 York Health Economics Consortium (YHEC Ltd), University of York, York, UK
  • 11 Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada; School of Epidemiology and Public Health, University of Ottawa, Ottawa, Canada; Department of Medicine, University of Ottawa, Ottawa, Canada
  • 12 Centre for Evidence-Based Medicine Odense (CEBMO) and Cochrane Denmark, Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Open Patient data Exploratory Network (OPEN), Odense University Hospital, Odense, Denmark
  • 13 Department of Anesthesiology and Pain Medicine, The Ottawa Hospital, Ottawa, Canada; Clinical Epidemiology Program, Blueprint Translational Research Group, Ottawa Hospital Research Institute, Ottawa, Canada; Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Canada
  • 14 Department of Ophthalmology, School of Medicine, University of Colorado Denver, Denver, Colorado, United States; Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, USA
  • 15 Division of Headache, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA; Head of Research, The BMJ , London, UK
  • 16 Department of Epidemiology and Biostatistics, Indiana University School of Public Health-Bloomington, Bloomington, Indiana, USA
  • 17 Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, UK
  • 18 Centre for Reviews and Dissemination, University of York, York, UK
  • 19 EPPI-Centre, UCL Social Research Institute, University College London, London, UK
  • 20 Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, Toronto, Canada; Epidemiology Division of the Dalla Lana School of Public Health and the Institute of Health Management, Policy, and Evaluation, University of Toronto, Toronto, Canada; Queen's Collaboration for Health Care Quality Joanna Briggs Institute Centre of Excellence, Queen's University, Kingston, Canada
  • 21 Methods Centre, Bruyère Research Institute, Ottawa, Ontario, Canada; School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Canada
  • 22 Centre for Journalology, Clinical Epidemiology Program, Ottawa Hospital Research Institute, Ottawa, Canada; School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, Canada
  • Correspondence to: M J Page matthew.page{at}monash.edu
  • Accepted 4 January 2021

The Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement, published in 2009, was designed to help systematic reviewers transparently report why the review was done, what the authors did, and what they found. Over the past decade, advances in systematic review methodology and terminology have necessitated an update to the guideline. The PRISMA 2020 statement replaces the 2009 statement and includes new reporting guidance that reflects advances in methods to identify, select, appraise, and synthesise studies. The structure and presentation of the items have been modified to facilitate implementation. In this article, we present the PRISMA 2020 27-item checklist, an expanded checklist that details reporting recommendations for each item, the PRISMA 2020 abstract checklist, and the revised flow diagrams for original and updated reviews.

Systematic reviews serve many critical roles. They can provide syntheses of the state of knowledge in a field, from which future research priorities can be identified; they can address questions that otherwise could not be answered by individual studies; they can identify problems in primary research that should be rectified in future studies; and they can generate or evaluate theories about how or why phenomena occur. Systematic reviews therefore generate various types of knowledge for different users of reviews (such as patients, healthcare providers, researchers, and policy makers). 1 2 To ensure a systematic review is valuable to users, authors should prepare a transparent, complete, and accurate account of why the review was done, what they did (such as how studies were identified and selected) and what they found (such as characteristics of contributing studies and results of meta-analyses). Up-to-date reporting guidance facilitates authors achieving this. 3

The Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement published in 2009 (hereafter referred to as PRISMA 2009) 4 5 6 7 8 9 10 is a reporting guideline designed to address poor reporting of systematic reviews. 11 The PRISMA 2009 statement comprised a checklist of 27 items recommended for reporting in systematic reviews and an “explanation and elaboration” paper 12 13 14 15 16 providing additional reporting guidance for each item, along with exemplars of reporting. The recommendations have been widely endorsed and adopted, as evidenced by its co-publication in multiple journals, citation in over 60 000 reports (Scopus, August 2020), endorsement from almost 200 journals and systematic review organisations, and adoption in various disciplines. Evidence from observational studies suggests that use of the PRISMA 2009 statement is associated with more complete reporting of systematic reviews, 17 18 19 20 although more could be done to improve adherence to the guideline. 21

Many innovations in the conduct of systematic reviews have occurred since publication of the PRISMA 2009 statement. For example, technological advances have enabled the use of natural language processing and machine learning to identify relevant evidence, 22 23 24 methods have been proposed to synthesise and present findings when meta-analysis is not possible or appropriate, 25 26 27 and new methods have been developed to assess the risk of bias in results of included studies. 28 29 Evidence on sources of bias in systematic reviews has accrued, culminating in the development of new tools to appraise the conduct of systematic reviews. 30 31 Terminology used to describe particular review processes has also evolved, as in the shift from assessing “quality” to assessing “certainty” in the body of evidence. 32 In addition, the publishing landscape has transformed, with multiple avenues now available for registering and disseminating systematic review protocols, 33 34 disseminating reports of systematic reviews, and sharing data and materials, such as preprint servers and publicly accessible repositories. To capture these advances in the reporting of systematic reviews necessitated an update to the PRISMA 2009 statement.

Summary points

To ensure a systematic review is valuable to users, authors should prepare a transparent, complete, and accurate account of why the review was done, what they did, and what they found

The PRISMA 2020 statement provides updated reporting guidance for systematic reviews that reflects advances in methods to identify, select, appraise, and synthesise studies

The PRISMA 2020 statement consists of a 27-item checklist, an expanded checklist that details reporting recommendations for each item, the PRISMA 2020 abstract checklist, and revised flow diagrams for original and updated reviews

We anticipate that the PRISMA 2020 statement will benefit authors, editors, and peer reviewers of systematic reviews, and different users of reviews, including guideline developers, policy makers, healthcare providers, patients, and other stakeholders

Development of PRISMA 2020

A complete description of the methods used to develop PRISMA 2020 is available elsewhere. 35 We identified PRISMA 2009 items that were often reported incompletely by examining the results of studies investigating the transparency of reporting of published reviews. 17 21 36 37 We identified possible modifications to the PRISMA 2009 statement by reviewing 60 documents providing reporting guidance for systematic reviews (including reporting guidelines, handbooks, tools, and meta-research studies). 38 These reviews of the literature were used to inform the content of a survey with suggested possible modifications to the 27 items in PRISMA 2009 and possible additional items. Respondents were asked whether they believed we should keep each PRISMA 2009 item as is, modify it, or remove it, and whether we should add each additional item. Systematic review methodologists and journal editors were invited to complete the online survey (110 of 220 invited responded). We discussed proposed content and wording of the PRISMA 2020 statement, as informed by the review and survey results, at a 21-member, two-day, in-person meeting in September 2018 in Edinburgh, Scotland. Throughout 2019 and 2020, we circulated an initial draft and five revisions of the checklist and explanation and elaboration paper to co-authors for feedback. In April 2020, we invited 22 systematic reviewers who had expressed interest in providing feedback on the PRISMA 2020 checklist to share their views (via an online survey) on the layout and terminology used in a preliminary version of the checklist. Feedback was received from 15 individuals and considered by the first author, and any revisions deemed necessary were incorporated before the final version was approved and endorsed by all co-authors.

The PRISMA 2020 statement

Scope of the guideline.

The PRISMA 2020 statement has been designed primarily for systematic reviews of studies that evaluate the effects of health interventions, irrespective of the design of the included studies. However, the checklist items are applicable to reports of systematic reviews evaluating other interventions (such as social or educational interventions), and many items are applicable to systematic reviews with objectives other than evaluating interventions (such as evaluating aetiology, prevalence, or prognosis). PRISMA 2020 is intended for use in systematic reviews that include synthesis (such as pairwise meta-analysis or other statistical synthesis methods) or do not include synthesis (for example, because only one eligible study is identified). The PRISMA 2020 items are relevant for mixed-methods systematic reviews (which include quantitative and qualitative studies), but reporting guidelines addressing the presentation and synthesis of qualitative data should also be consulted. 39 40 PRISMA 2020 can be used for original systematic reviews, updated systematic reviews, or continually updated (“living”) systematic reviews. However, for updated and living systematic reviews, there may be some additional considerations that need to be addressed. Where there is relevant content from other reporting guidelines, we reference these guidelines within the items in the explanation and elaboration paper 41 (such as PRISMA-Search 42 in items 6 and 7, Synthesis without meta-analysis (SWiM) reporting guideline 27 in item 13d). Box 1 includes a glossary of terms used throughout the PRISMA 2020 statement.

Glossary of terms

Systematic review —A review that uses explicit, systematic methods to collate and synthesise findings of studies that address a clearly formulated question 43

Statistical synthesis —The combination of quantitative results of two or more studies. This encompasses meta-analysis of effect estimates (described below) and other methods, such as combining P values, calculating the range and distribution of observed effects, and vote counting based on the direction of effect (see McKenzie and Brennan 25 for a description of each method)

Meta-analysis of effect estimates —A statistical technique used to synthesise results when study effect estimates and their variances are available, yielding a quantitative summary of results 25

Outcome —An event or measurement collected for participants in a study (such as quality of life, mortality)

Result —The combination of a point estimate (such as a mean difference, risk ratio, or proportion) and a measure of its precision (such as a confidence/credible interval) for a particular outcome

Report —A document (paper or electronic) supplying information about a particular study. It could be a journal article, preprint, conference abstract, study register entry, clinical study report, dissertation, unpublished manuscript, government report, or any other document providing relevant information

Record —The title or abstract (or both) of a report indexed in a database or website (such as a title or abstract for an article indexed in Medline). Records that refer to the same report (such as the same journal article) are “duplicates”; however, records that refer to reports that are merely similar (such as a similar abstract submitted to two different conferences) should be considered unique.

Study —An investigation, such as a clinical trial, that includes a defined group of participants and one or more interventions and outcomes. A “study” might have multiple reports. For example, reports could include the protocol, statistical analysis plan, baseline characteristics, results for the primary outcome, results for harms, results for secondary outcomes, and results for additional mediator and moderator analyses

PRISMA 2020 is not intended to guide systematic review conduct, for which comprehensive resources are available. 43 44 45 46 However, familiarity with PRISMA 2020 is useful when planning and conducting systematic reviews to ensure that all recommended information is captured. PRISMA 2020 should not be used to assess the conduct or methodological quality of systematic reviews; other tools exist for this purpose. 30 31 Furthermore, PRISMA 2020 is not intended to inform the reporting of systematic review protocols, for which a separate statement is available (PRISMA for Protocols (PRISMA-P) 2015 statement 47 48 ). Finally, extensions to the PRISMA 2009 statement have been developed to guide reporting of network meta-analyses, 49 meta-analyses of individual participant data, 50 systematic reviews of harms, 51 systematic reviews of diagnostic test accuracy studies, 52 and scoping reviews 53 ; for these types of reviews we recommend authors report their review in accordance with the recommendations in PRISMA 2020 along with the guidance specific to the extension.

How to use PRISMA 2020

The PRISMA 2020 statement (including the checklists, explanation and elaboration, and flow diagram) replaces the PRISMA 2009 statement, which should no longer be used. Box 2 summarises noteworthy changes from the PRISMA 2009 statement. The PRISMA 2020 checklist includes seven sections with 27 items, some of which include sub-items ( table 1 ). A checklist for journal and conference abstracts for systematic reviews is included in PRISMA 2020. This abstract checklist is an update of the 2013 PRISMA for Abstracts statement, 54 reflecting new and modified content in PRISMA 2020 ( table 2 ). A template PRISMA flow diagram is provided, which can be modified depending on whether the systematic review is original or updated ( fig 1 ).

Noteworthy changes to the PRISMA 2009 statement

Inclusion of the abstract reporting checklist within PRISMA 2020 (see item #2 and table 2 ).

Movement of the ‘Protocol and registration’ item from the start of the Methods section of the checklist to a new Other section, with addition of a sub-item recommending authors describe amendments to information provided at registration or in the protocol (see item #24a-24c).

Modification of the ‘Search’ item to recommend authors present full search strategies for all databases, registers and websites searched, not just at least one database (see item #7).

Modification of the ‘Study selection’ item in the Methods section to emphasise the reporting of how many reviewers screened each record and each report retrieved, whether they worked independently, and if applicable, details of automation tools used in the process (see item #8).

Addition of a sub-item to the ‘Data items’ item recommending authors report how outcomes were defined, which results were sought, and methods for selecting a subset of results from included studies (see item #10a).

Splitting of the ‘Synthesis of results’ item in the Methods section into six sub-items recommending authors describe: the processes used to decide which studies were eligible for each synthesis; any methods required to prepare the data for synthesis; any methods used to tabulate or visually display results of individual studies and syntheses; any methods used to synthesise results; any methods used to explore possible causes of heterogeneity among study results (such as subgroup analysis, meta-regression); and any sensitivity analyses used to assess robustness of the synthesised results (see item #13a-13f).

Addition of a sub-item to the ‘Study selection’ item in the Results section recommending authors cite studies that might appear to meet the inclusion criteria, but which were excluded, and explain why they were excluded (see item #16b).

Splitting of the ‘Synthesis of results’ item in the Results section into four sub-items recommending authors: briefly summarise the characteristics and risk of bias among studies contributing to the synthesis; present results of all statistical syntheses conducted; present results of any investigations of possible causes of heterogeneity among study results; and present results of any sensitivity analyses (see item #20a-20d).

Addition of new items recommending authors report methods for and results of an assessment of certainty (or confidence) in the body of evidence for an outcome (see items #15 and #22).

Addition of a new item recommending authors declare any competing interests (see item #26).

Addition of a new item recommending authors indicate whether data, analytic code and other materials used in the review are publicly available and if so, where they can be found (see item #27).

PRISMA 2020 item checklist

  • View inline

PRISMA 2020 for Abstracts checklist*

Fig 1

PRISMA 2020 flow diagram template for systematic reviews. The new design is adapted from flow diagrams proposed by Boers, 55 Mayo-Wilson et al. 56 and Stovold et al. 57 The boxes in grey should only be completed if applicable; otherwise they should be removed from the flow diagram. Note that a “report” could be a journal article, preprint, conference abstract, study register entry, clinical study report, dissertation, unpublished manuscript, government report or any other document providing relevant information.

  • Download figure
  • Open in new tab
  • Download powerpoint

We recommend authors refer to PRISMA 2020 early in the writing process, because prospective consideration of the items may help to ensure that all the items are addressed. To help keep track of which items have been reported, the PRISMA statement website ( http://www.prisma-statement.org/ ) includes fillable templates of the checklists to download and complete (also available in the data supplement on bmj.com). We have also created a web application that allows users to complete the checklist via a user-friendly interface 58 (available at https://prisma.shinyapps.io/checklist/ and adapted from the Transparency Checklist app 59 ). The completed checklist can be exported to Word or PDF. Editable templates of the flow diagram can also be downloaded from the PRISMA statement website.

We have prepared an updated explanation and elaboration paper, in which we explain why reporting of each item is recommended and present bullet points that detail the reporting recommendations (which we refer to as elements). 41 The bullet-point structure is new to PRISMA 2020 and has been adopted to facilitate implementation of the guidance. 60 61 An expanded checklist, which comprises an abridged version of the elements presented in the explanation and elaboration paper, with references and some examples removed, is available in the data supplement on bmj.com. Consulting the explanation and elaboration paper is recommended if further clarity or information is required.

Journals and publishers might impose word and section limits, and limits on the number of tables and figures allowed in the main report. In such cases, if the relevant information for some items already appears in a publicly accessible review protocol, referring to the protocol may suffice. Alternatively, placing detailed descriptions of the methods used or additional results (such as for less critical outcomes) in supplementary files is recommended. Ideally, supplementary files should be deposited to a general-purpose or institutional open-access repository that provides free and permanent access to the material (such as Open Science Framework, Dryad, figshare). A reference or link to the additional information should be included in the main report. Finally, although PRISMA 2020 provides a template for where information might be located, the suggested location should not be seen as prescriptive; the guiding principle is to ensure the information is reported.

Use of PRISMA 2020 has the potential to benefit many stakeholders. Complete reporting allows readers to assess the appropriateness of the methods, and therefore the trustworthiness of the findings. Presenting and summarising characteristics of studies contributing to a synthesis allows healthcare providers and policy makers to evaluate the applicability of the findings to their setting. Describing the certainty in the body of evidence for an outcome and the implications of findings should help policy makers, managers, and other decision makers formulate appropriate recommendations for practice or policy. Complete reporting of all PRISMA 2020 items also facilitates replication and review updates, as well as inclusion of systematic reviews in overviews (of systematic reviews) and guidelines, so teams can leverage work that is already done and decrease research waste. 36 62 63

We updated the PRISMA 2009 statement by adapting the EQUATOR Network’s guidance for developing health research reporting guidelines. 64 We evaluated the reporting completeness of published systematic reviews, 17 21 36 37 reviewed the items included in other documents providing guidance for systematic reviews, 38 surveyed systematic review methodologists and journal editors for their views on how to revise the original PRISMA statement, 35 discussed the findings at an in-person meeting, and prepared this document through an iterative process. Our recommendations are informed by the reviews and survey conducted before the in-person meeting, theoretical considerations about which items facilitate replication and help users assess the risk of bias and applicability of systematic reviews, and co-authors’ experience with authoring and using systematic reviews.

Various strategies to increase the use of reporting guidelines and improve reporting have been proposed. They include educators introducing reporting guidelines into graduate curricula to promote good reporting habits of early career scientists 65 ; journal editors and regulators endorsing use of reporting guidelines 18 ; peer reviewers evaluating adherence to reporting guidelines 61 66 ; journals requiring authors to indicate where in their manuscript they have adhered to each reporting item 67 ; and authors using online writing tools that prompt complete reporting at the writing stage. 60 Multi-pronged interventions, where more than one of these strategies are combined, may be more effective (such as completion of checklists coupled with editorial checks). 68 However, of 31 interventions proposed to increase adherence to reporting guidelines, the effects of only 11 have been evaluated, mostly in observational studies at high risk of bias due to confounding. 69 It is therefore unclear which strategies should be used. Future research might explore barriers and facilitators to the use of PRISMA 2020 by authors, editors, and peer reviewers, designing interventions that address the identified barriers, and evaluating those interventions using randomised trials. To inform possible revisions to the guideline, it would also be valuable to conduct think-aloud studies 70 to understand how systematic reviewers interpret the items, and reliability studies to identify items where there is varied interpretation of the items.

We encourage readers to submit evidence that informs any of the recommendations in PRISMA 2020 (via the PRISMA statement website: http://www.prisma-statement.org/ ). To enhance accessibility of PRISMA 2020, several translations of the guideline are under way (see available translations at the PRISMA statement website). We encourage journal editors and publishers to raise awareness of PRISMA 2020 (for example, by referring to it in journal “Instructions to authors”), endorsing its use, advising editors and peer reviewers to evaluate submitted systematic reviews against the PRISMA 2020 checklists, and making changes to journal policies to accommodate the new reporting recommendations. We recommend existing PRISMA extensions 47 49 50 51 52 53 71 72 be updated to reflect PRISMA 2020 and advise developers of new PRISMA extensions to use PRISMA 2020 as the foundation document.

We anticipate that the PRISMA 2020 statement will benefit authors, editors, and peer reviewers of systematic reviews, and different users of reviews, including guideline developers, policy makers, healthcare providers, patients, and other stakeholders. Ultimately, we hope that uptake of the guideline will lead to more transparent, complete, and accurate reporting of systematic reviews, thus facilitating evidence based decision making.

Acknowledgments

We dedicate this paper to the late Douglas G Altman and Alessandro Liberati, whose contributions were fundamental to the development and implementation of the original PRISMA statement.

We thank the following contributors who completed the survey to inform discussions at the development meeting: Xavier Armoiry, Edoardo Aromataris, Ana Patricia Ayala, Ethan M Balk, Virginia Barbour, Elaine Beller, Jesse A Berlin, Lisa Bero, Zhao-Xiang Bian, Jean Joel Bigna, Ferrán Catalá-López, Anna Chaimani, Mike Clarke, Tammy Clifford, Ioana A Cristea, Miranda Cumpston, Sofia Dias, Corinna Dressler, Ivan D Florez, Joel J Gagnier, Chantelle Garritty, Long Ge, Davina Ghersi, Sean Grant, Gordon Guyatt, Neal R Haddaway, Julian PT Higgins, Sally Hopewell, Brian Hutton, Jamie J Kirkham, Jos Kleijnen, Julia Koricheva, Joey SW Kwong, Toby J Lasserson, Julia H Littell, Yoon K Loke, Malcolm R Macleod, Chris G Maher, Ana Marušic, Dimitris Mavridis, Jessie McGowan, Matthew DF McInnes, Philippa Middleton, Karel G Moons, Zachary Munn, Jane Noyes, Barbara Nußbaumer-Streit, Donald L Patrick, Tatiana Pereira-Cenci, Ba’ Pham, Bob Phillips, Dawid Pieper, Michelle Pollock, Daniel S Quintana, Drummond Rennie, Melissa L Rethlefsen, Hannah R Rothstein, Maroeska M Rovers, Rebecca Ryan, Georgia Salanti, Ian J Saldanha, Margaret Sampson, Nancy Santesso, Rafael Sarkis-Onofre, Jelena Savović, Christopher H Schmid, Kenneth F Schulz, Guido Schwarzer, Beverley J Shea, Paul G Shekelle, Farhad Shokraneh, Mark Simmonds, Nicole Skoetz, Sharon E Straus, Anneliese Synnot, Emily E Tanner-Smith, Brett D Thombs, Hilary Thomson, Alexander Tsertsvadze, Peter Tugwell, Tari Turner, Lesley Uttley, Jeffrey C Valentine, Matt Vassar, Areti Angeliki Veroniki, Meera Viswanathan, Cole Wayant, Paul Whaley, and Kehu Yang. We thank the following contributors who provided feedback on a preliminary version of the PRISMA 2020 checklist: Jo Abbott, Fionn Büttner, Patricia Correia-Santos, Victoria Freeman, Emily A Hennessy, Rakibul Islam, Amalia (Emily) Karahalios, Kasper Krommes, Andreas Lundh, Dafne Port Nascimento, Davina Robson, Catherine Schenck-Yglesias, Mary M Scott, Sarah Tanveer and Pavel Zhelnov. We thank Abigail H Goben, Melissa L Rethlefsen, Tanja Rombey, Anna Scott, and Farhad Shokraneh for their helpful comments on the preprints of the PRISMA 2020 papers. We thank Edoardo Aromataris, Stephanie Chang, Toby Lasserson and David Schriger for their helpful peer review comments on the PRISMA 2020 papers.

Contributors: JEM and DM are joint senior authors. MJP, JEM, PMB, IB, TCH, CDM, LS, and DM conceived this paper and designed the literature review and survey conducted to inform the guideline content. MJP conducted the literature review, administered the survey and analysed the data for both. MJP prepared all materials for the development meeting. MJP and JEM presented proposals at the development meeting. All authors except for TCH, JMT, EAA, SEB, and LAM attended the development meeting. MJP and JEM took and consolidated notes from the development meeting. MJP and JEM led the drafting and editing of the article. JEM, PMB, IB, TCH, LS, JMT, EAA, SEB, RC, JG, AH, TL, EMW, SM, LAM, LAS, JT, ACT, PW, and DM drafted particular sections of the article. All authors were involved in revising the article critically for important intellectual content. All authors approved the final version of the article. MJP is the guarantor of this work. The corresponding author attests that all listed authors meet authorship criteria and that no others meeting the criteria have been omitted.

Funding: There was no direct funding for this research. MJP is supported by an Australian Research Council Discovery Early Career Researcher Award (DE200101618) and was previously supported by an Australian National Health and Medical Research Council (NHMRC) Early Career Fellowship (1088535) during the conduct of this research. JEM is supported by an Australian NHMRC Career Development Fellowship (1143429). TCH is supported by an Australian NHMRC Senior Research Fellowship (1154607). JMT is supported by Evidence Partners Inc. JMG is supported by a Tier 1 Canada Research Chair in Health Knowledge Transfer and Uptake. MML is supported by The Ottawa Hospital Anaesthesia Alternate Funds Association and a Faculty of Medicine Junior Research Chair. TL is supported by funding from the National Eye Institute (UG1EY020522), National Institutes of Health, United States. LAM is supported by a National Institute for Health Research Doctoral Research Fellowship (DRF-2018-11-ST2-048). ACT is supported by a Tier 2 Canada Research Chair in Knowledge Synthesis. DM is supported in part by a University Research Chair, University of Ottawa. The funders had no role in considering the study design or in the collection, analysis, interpretation of data, writing of the report, or decision to submit the article for publication.

Competing interests: All authors have completed the ICMJE uniform disclosure form at http://www.icmje.org/conflicts-of-interest/ and declare: EL is head of research for the BMJ ; MJP is an editorial board member for PLOS Medicine ; ACT is an associate editor and MJP, TL, EMW, and DM are editorial board members for the Journal of Clinical Epidemiology ; DM and LAS were editors in chief, LS, JMT, and ACT are associate editors, and JG is an editorial board member for Systematic Reviews . None of these authors were involved in the peer review process or decision to publish. TCH has received personal fees from Elsevier outside the submitted work. EMW has received personal fees from the American Journal for Public Health , for which he is the editor for systematic reviews. VW is editor in chief of the Campbell Collaboration, which produces systematic reviews, and co-convenor of the Campbell and Cochrane equity methods group. DM is chair of the EQUATOR Network, IB is adjunct director of the French EQUATOR Centre and TCH is co-director of the Australasian EQUATOR Centre, which advocates for the use of reporting guidelines to improve the quality of reporting in research articles. JMT received salary from Evidence Partners, creator of DistillerSR software for systematic reviews; Evidence Partners was not involved in the design or outcomes of the statement, and the views expressed solely represent those of the author.

Provenance and peer review: Not commissioned; externally peer reviewed.

Patient and public involvement: Patients and the public were not involved in this methodological research. We plan to disseminate the research widely, including to community participants in evidence synthesis organisations.

This is an Open Access article distributed in accordance with the terms of the Creative Commons Attribution (CC BY 4.0) license, which permits others to distribute, remix, adapt and build upon this work, for commercial use, provided the original work is properly cited. See: http://creativecommons.org/licenses/by/4.0/ .

  • Gurevitch J ,
  • Koricheva J ,
  • Nakagawa S ,
  • Liberati A ,
  • Tetzlaff J ,
  • Altman DG ,
  • PRISMA Group
  • Tricco AC ,
  • Sampson M ,
  • Shamseer L ,
  • Leoncini E ,
  • de Belvis G ,
  • Ricciardi W ,
  • Fowler AJ ,
  • Leclercq V ,
  • Beaudart C ,
  • Ajamieh S ,
  • Rabenda V ,
  • Tirelli E ,
  • O’Mara-Eves A ,
  • McNaught J ,
  • Ananiadou S
  • Marshall IJ ,
  • Noel-Storr A ,
  • Higgins JPT ,
  • Chandler J ,
  • McKenzie JE ,
  • López-López JA ,
  • Becker BJ ,
  • Campbell M ,
  • Sterne JAC ,
  • Savović J ,
  • Sterne JA ,
  • Hernán MA ,
  • Reeves BC ,
  • Whiting P ,
  • Higgins JP ,
  • ROBIS group
  • Hultcrantz M ,
  • Stewart L ,
  • Bossuyt PM ,
  • Flemming K ,
  • McInnes E ,
  • France EF ,
  • Cunningham M ,
  • Rethlefsen ML ,
  • Kirtley S ,
  • Waffenschmidt S ,
  • PRISMA-S Group
  • ↵ Higgins JPT, Thomas J, Chandler J, et al, eds. Cochrane Handbook for Systematic Reviews of Interventions : Version 6.0. Cochrane, 2019. Available from https://training.cochrane.org/handbook .
  • Dekkers OM ,
  • Vandenbroucke JP ,
  • Cevallos M ,
  • Renehan AG ,
  • ↵ Cooper H, Hedges LV, Valentine JV, eds. The Handbook of Research Synthesis and Meta-Analysis. Russell Sage Foundation, 2019.
  • IOM (Institute of Medicine)
  • PRISMA-P Group
  • Salanti G ,
  • Caldwell DM ,
  • Stewart LA ,
  • PRISMA-IPD Development Group
  • Zorzela L ,
  • Ioannidis JP ,
  • PRISMAHarms Group
  • McInnes MDF ,
  • Thombs BD ,
  • and the PRISMA-DTA Group
  • Beller EM ,
  • Glasziou PP ,
  • PRISMA for Abstracts Group
  • Mayo-Wilson E ,
  • Dickersin K ,
  • MUDS investigators
  • Stovold E ,
  • Beecher D ,
  • Noel-Storr A
  • McGuinness LA
  • Sarafoglou A ,
  • Boutron I ,
  • Giraudeau B ,
  • Porcher R ,
  • Chauvin A ,
  • Schulz KF ,
  • Schroter S ,
  • Stevens A ,
  • Weinstein E ,
  • Macleod MR ,
  • IICARus Collaboration
  • Kirkham JJ ,
  • Petticrew M ,
  • Tugwell P ,
  • PRISMA-Equity Bellagio group

the purpose of a systematic review

Newsletter Sign Up

Archie login.

  • Campbell Website Campbell Library -->

What is a systematic review?

A systematic review is an academic research paper, also called a report, that uses a method called 'evidence synthesis' to look for answers to a pre-defined question.

The purpose of a systematic review is to sum up the best available research on that specific question. This is done by synthesizing the results of several studies.

A systematic review uses transparent procedures to find, evaluate and synthesize the results of relevant research. Procedures are explicitly defined in advance, to ensure that the exercise is transparent and can be replicated. This practice is also designed to minimize bias.

Studies included in a review are screened for quality, so that the findings of a large number of studies can be combined. Peer review is a key part of the process; qualified independent researchers review the author's methods and results.

A systematic review must have:

  • Clear inclusion and exclusion criteria
  • An explicit search strategy
  • Systematic coding and analysis of included studies
  • Meta-analysis (where possible)

How do Campbell systematic reviews differ from less reliable systematic reviews?

  • Campbell reviews must include a systematic search for unpublished reports, to avoid publication bias.
  • Campbell reviews are usually international in scope.
  • A protocol (project plan) for the review is developed in advance and also undergoes peer review.
  • Study inclusion and coding decisions are carried out by at least two reviewers who work independently and compare results.
  • Study quality is appraised.
  • Campbell reviews undergo peer review and editorial review.
  • Campbell reviews provide answers for decisionmakers by using rigorous methods to synthesize evidence, including, where appropriate, statistical meta-analysis of quantitative evidence and theory-based analysis of qualitative evidence.

See all the Campbell systematic reviews on our journal website: click here .

  Donate to Campbell

Useful links

  • Evidence portals
  • Privacy policy
  • Terms of use

Lastest publications

  • New plain language summaries
  • About evidence and gap maps

Follow @campbellreviews

In order to provide you with the best online experience this website uses cookies.

We use cookies to improve our website. By continuing to use this website, you are giving consent to cookies being used by Google Analytics, and those social media channels you expressly select by clicking on buttons (links). Cookie policy . I accept cookies from this site.

GDPR compliance/Cookies

  • What are cookies?
  • How do we use cookies?
  • Cookies used on our site
  • How to delete cookies or control them

[divider type="stripes" margin="20px 0 20px 0" ]

At the top of our website we have displayed a message to warn you that our website makes use of cookies and that one has already been set. By displaying this message we hope that we are providing you with the information you require about our use of cookies, and presenting you with the option to consent to their use. This message will be displayed until such time as you agree to our site using cookies by clicking on the continue button. 

1.What is a cookie?

A cookie is a small amount of data, often including a unique identifier, sent to the browser of your computer or mobile phone (referred to here as a "device") from a website's computer. It is stored on your device's hard drive. Each website can send its own cookie to your browser if your browser's preferences allow it. To protect your privacy, your browser only permits a website to access the cookies it has already sent to you, and not the cookies sent to you by other websites. Many websites do this whenever a user visits them to track online traffic flows.

On the Channel Digital website, our cookies record information about your online preferences so we can tailor the site to your interests. You can set your device’s preferences to accept all cookies, notify you when a cookie is issued, or not receive cookies at all. Selecting the last option means you will not receive certain personalised features, which may result in you being unable to take full advantage of all the website's features. Each browser is different, so please check the "Help" menu of your browser to learn how to change your cookie preferences.

During the course of any visit to our website, every page you see, along with a cookie, is downloaded to your device. Many websites do this because cookies enable website publishers to do useful things like find out whether your device (and probably you) has visited the website before. On a repeat visit this is done by the website’s computer checking to see, and finding, the cookie left there on the last visit. 

2.How do we use cookies?

Information supplied by cookies can help us analyse the profile of our visitors, which helps us provide you with a better user experience. For example, if on a previous visit you went to our marketing pages, we might find this out from your cookie and highlight marketing information on subsequent visits.

Third party cookies on our pages

Please note that during your visits to our website you may notice some cookies which are unrelated to us. When you visit a page with content embedded from, for example, Twitter or YouTube, you may be presented with cookies from these websites. We do not control the dissemination of these cookies. You should check the third party websites for more information about these. 

3.Cookies used on our site

We only use cookies to help us continuously improve our website and maintain a nice browsing experience for our visitors. Here is a list of cookies used on this website:

  • Google Analytics - we use cookies to compile visitor statistics such as how many people have visited our website, what type of technology they are using (e.g. Mac or Windows which helps to identify when our site isn’t working as it should for particular technologies), how long they spend on the site, what page they look at etc.
  • Facebook, Twitter, LinkedIn, Google+  - social media share buttons that enable you to share our content
  • Session cookie - this is a standard cookie just to remember user preferences (like font size and saves you logging in every time you visit)

4.How to delete cookies or control them

This site will not use cookies to collect personally identifiable information about you. However, if you wish to restrict or block cookies set by this or any other website, you can do this through your browser settings. The Help function within your browser should tell you how.

Alternatively, you may wish to visit www.aboutcookies.org which contains comprehensive information on how to do this for a wide variety of browsers. You will also find details on how to clear cookies from your computer as well as more general information about cookies. For information on how to do this on your mobile phone’s browser, you will need to refer to your browser support information.

  • Library Hours
  • Strategic Plan
  • Giving to the Libraries
  • Jobs at the Libraries
  • Find Your Librarian
  • View All →
  • Google Scholar
  • Research Guides
  • Textbook/Reserves
  • Government Documents
  • Get It For Me
  • Print/Copy/Scan
  • Renew Materials
  • Study Rooms
  • Use a Computer
  • Borrow Tech Gear
  • Student Services
  • Faculty Services
  • Users with Disabilities
  • Visitors & Alumni
  • Special Collections
  • Find Information

Basics of Systematic Reviews

  • About Systematic Review

Types of Reviews

Literature review.

Collects key sources on a topic and discusses those sources in conversation with each other

  • Standard for research articles in most disciplines
  • Tells the reader what is known, or not known, about a particular issue, topic, or subject
  • Demonstrates knowledge and understanding of a topic
  • Establishes context or background for a case or argument
  • Helps develop the author’s ideas and perspective

Rapid Review

Thorough methodology but with process limitations in place to expeditethe completion of a review.

  • For questions that require timely answers
  • 3-4 months vs. 12-24 months
  • Limitations - scope, comprehensiveness bias, and quality of appraisal
  • Discusses potential effects that the limited methods may have had on results

Scoping Review

Determine the scope or coverage of a body of literature on a given topic and give clear indication of the volume of literature and studies available as well as an overview of its focus.

  • Identify types of available evidence in a given field
  • Clarify key concepts/definitions in the literature
  • Examine how research is conducted on a certain topic or field
  • Identify key factors related to a concept
  • Key difference is focus
  • Identify and analyze knowledge gaps

Systematic Review

Attempts to identify, appraise, and summarize all empirical evidence that fits pre-specified eligibility criteria to answer a specific research question.

  • clearly defined question with inclusion/exclusion criteria
  • rigorous and systematic search of the literature
  • thorough screening of results
  • data extraction and management
  • analysis and interpretation of results
  • risk of bias assessment of included studies

Meta-Analysis

Used to systematically synthesize or merge the findings of single, independent studies, using statistical methods to calculate an overall or ‘absolute’ effect.

  • Combines results from multiple empirical studies
  • Requires systematic review first
  • Use well recognized, systematic methods to account for differences in sample size, variability (heterogeneity) in study approach and findings (treatment effects)
  • Test how sensitive their results are to their own systematic review protocol

For additional types of reviews please see these articles:

  • Sutton, A., Clowes, M., Preston, L. and Booth, A. (2019), Meeting the review family: exploring review types and associated information retrieval requirements. Health Info Libr J, 36: 202-222. https://doi.org/10.1111/hir.12276
  • Grant, M.J. and Booth, A. (2009), A typology of reviews: an analysis of 14 review types and associated methodologies. Health Information & Libraries Journal, 26: 91-108. https://doi.org/10.1111/j.1471-1842.2009.00848.x
  • << Previous: About Systematic Review
  • Next: Sources >>
  • Last Updated: May 17, 2024 10:04 AM
  • URL: https://libguides.utsa.edu/systematicreview
  • Library Locations
  • Staff Directory
  • 508 Compliance
  • Site Search
  • © The University of Texas at San Antonio
  • Information: 210-458-4011
  • Campus Alerts
  • Required Links
  • UTSA Policies
  • Report Fraud
  • Open access
  • Published: 14 May 2024

Protocol for a scoping review study on learning plan use in undergraduate medical education

  • Anna Romanova   ORCID: orcid.org/0000-0003-1118-1604 1 ,
  • Claire Touchie 1 ,
  • Sydney Ruller 2 ,
  • Victoria Cole 3 &
  • Susan Humphrey-Murto 4  

Systematic Reviews volume  13 , Article number:  131 ( 2024 ) Cite this article

93 Accesses

Metrics details

The current paradigm of competency-based medical education and learner-centredness requires learners to take an active role in their training. However, deliberate and planned continual assessment and performance improvement is hindered by the fragmented nature of many medical training programs. Attempts to bridge this continuity gap between supervision and feedback through learner handover have been controversial. Learning plans are an alternate educational tool that helps trainees identify their learning needs and facilitate longitudinal assessment by providing supervisors with a roadmap of their goals. Informed by self-regulated learning theory, learning plans may be the answer to track trainees’ progress along their learning trajectory. The purpose of this study is to summarise the literature regarding learning plan use specifically in undergraduate medical education and explore the student’s role in all stages of learning plan development and implementation.

Following Arksey and O’Malley’s framework, a scoping review will be conducted to explore the use of learning plans in undergraduate medical education. Literature searches will be conducted using multiple databases by a librarian with expertise in scoping reviews. Through an iterative process, inclusion and exclusion criteria will be developed and a data extraction form refined. Data will be analysed using quantitative and qualitative content analyses.

By summarising the literature on learning plan use in undergraduate medical education, this study aims to better understand how to support self-regulated learning in undergraduate medical education. The results from this project will inform future scholarly work in competency-based medical education at the undergraduate level and have implications for improving feedback and supporting learners at all levels of competence.

Scoping review registration:

Open Science Framework osf.io/wvzbx.

Peer Review reports

Competency-based medical education (CBME) has transformed the approach to medical education to focus on demonstration of acquired competencies rather than time-based completion of rotations [ 1 ]. As a result, undergraduate and graduate medical training programs worldwide have adopted outcomes-based assessments in the form of entrustable professional activities (EPAs) comprised of competencies to be met [ 2 ]. These assessments are completed longitudinally by multiple different evaluators to generate an overall impression of a learner’s competency.

In CBME, trainees will progress along their learning trajectory at individual speeds and some may excel while others struggle to achieve the required knowledge, skills or attitudes. Therefore, deliberate and planned continual assessment and performance improvement is required. However, due to the fragmented nature of many medical training programs where learners rotate through different rotations and work with many supervisors, longitudinal observation is similarly fragmented. This makes it difficult to determine where trainees are on their learning trajectories and can affect the quality of feedback provided to them, which is a known major influencer of academic achievement [ 3 ]. As a result, struggling learners may not be identified until late in their training and the growth of high-performing learners may be stifled [ 4 , 5 , 6 ].

Bridging this continuity gap between supervision and feedback through some form of learner handover or forward feeding has been debated since the 1970s and continues to this day [ 5 , 7 , 8 , 9 , 10 , 11 ]. The goal of learner handover is to improve trainee assessment and feedback by sharing their performance and learning needs between supervisors or across rotations. However, several concerns have been raised about this approach including that it could inappropriately bias subsequent assessments of the learner’s abilities [ 9 , 11 , 12 ]. A different approach to keeping track of trainees’ learning goals and progress along their learning trajectories is required. Learning plans (LPs) informed by self-regulated learning (SRL) theory may be the answer.

SRL has been defined as a cyclical process where learners actively control their thoughts, actions and motivation to achieve their goals [ 13 ]. Several models of SRL exist but all entail that the trainee is responsible for setting, planning, executing, monitoring and reflecting on their learning goals [ 13 ]. According to Zimmerman’s SRL model, this process occurs in three stages: forethought phase before an activity, performance phase during an activity and self-reflection phase after an activity [ 13 ]. Since each trainee leads their own learning process and has an individual trajectory towards competence, this theory relates well to the CBME paradigm which is grounded in learner-centredness [ 1 ]. However, we know that medical students and residents have difficulty identifying their own learning goals and therefore need guidance to effectively partake in SRL [ 14 , 15 , 16 , 17 ]. Motivation has also emerged as a key component of SRL, and numerous studies have explored factors that influence student engagement in learning [ 18 , 19 ]. In addition to meeting their basic psychological needs of autonomy, relatedness and competence, perceived learning relevance through meaningful learning activities has been shown to increase trainee engagement in their learning [ 19 ].

LPs are a well-known tool across many educational fields including CBME that can provide trainees with meaningful learning activities since they help them direct their own learning goals in a guided fashion [ 20 ]. Also known as personal learning plans, learning contracts, personal action plans, personal development plans, and learning goals, LPs are documents that outline the learner’s roadmap to achieve their learning goals. They require the learner to self-identify what they need to learn and why, how they are going to do it, how they will know when they are finished, define the timeframe for goal achievement and assess the impact of their learning [ 20 ]. In so doing, LPs give more autonomy to the learner and facilitate objective and targeted feedback from supervisors. This approach has been described as “most congruent with the assumptions we make about adults as learners” [ 21 ].

LP use has been explored across various clinical settings and at all levels of medical education; however, most of the experience lies in postgraduate medical education [ 22 ]. Medical students are a unique learner population with learning needs that appear to be very well suited for using LPs for two main reasons. First, their education is often divided between classroom and clinical settings. During clinical training, students need to be more independent in setting learning goals to meet desired competencies as their education is no longer outlined for them in a detailed fashion by the medical school curriculum [ 23 ]. SRL in the workplace is also different than in the classroom due to additional complexities of clinical care that can impact students’ ability to self-regulate their learning [ 24 ]. Second, although most medical trainees have difficulty with goal setting, medical students in particular need more guidance compared to residents due to their relative lack of experience upon which they can build within the SRL framework [ 25 ]. LPs can therefore provide much-needed structure to their learning but should be guided by an experienced tutor to be effective [ 15 , 24 ].

LPs fit well within the learner-centred educational framework of CBME by helping trainees identify their learning needs and facilitating longitudinal assessment by providing supervisors with a roadmap of their goals. In so doing, they can address current issues with learner handover and identification as well as remediation of struggling learners. Moreover, they have the potential to help trainees develop lifelong skills with respect to continuing professional development after graduation which is required by many medical licensing bodies.

An initial search of the JBI Database, Cochrane Database, MEDLINE (PubMed) and Google Scholar conducted in July–August 2022 revealed a paucity of research on LP use in undergraduate medical education (UGME). A related systematic review by van Houten–Schat et al. [ 24 ] on SRL in the clinical setting identified three interventions used by medical students and residents in SRL—coaching, LPs and supportive tools. However, only a couple of the included studies looked specifically at medical students’ use of LPs, so this remains an area in need of more exploration. A scoping review would provide an excellent starting point to map the body of literature on this topic.

The objective of this scoping review will therefore be to explore LP use in UGME. In doing so, it will address a gap in knowledge and help determine additional areas for research.

This study will follow Arksey and O’Malley’s [ 26 ] five-step framework for scoping review methodology. It will not include the optional sixth step which entails stakeholder consultation as relevant stakeholders will be intentionally included in the research team (a member of UGME leadership, a medical student and a first-year resident).

Step 1—Identifying the research question

The overarching purpose of this study is to “explore the use of LPs in UGME”. More specifically we seek to achieve the following:

Summarise the literature regarding the use of LPs in UGME (including context, students targeted, frameworks used)

Explore the role of the student in all stages of the LP development and implementation

Determine existing research gaps

Step 2—Identifying relevant studies

An experienced health sciences librarian (VC) will conduct all searches and develop the initial search strategy. The preliminary search strategy is shown in Appendix A (see Additional file 2). Articles will be included if they meet the following criteria [ 27 ]:

Participants

Medical students enrolled at a medical school at the undergraduate level.

Any use of LPs by medical students. LPs are defined as a document, usually presented in a table format, that outlines the learner’s roadmap to achieve their learning goals [ 20 ].

Any stage of UGME in any geographic setting.

Types of evidence sources

We will search existing published and unpublished (grey) literature. This may include research studies, reviews, or expert opinion pieces.

Search strategy

With the assistance of an experienced librarian (VC), a pilot search will be conducted to inform the final search strategy. A search will be conducted in the following electronic databases: MEDLINE, Embase, Education Source, APA PsycInfo and Web of Science. The search terms will be developed in consultation with the research team and librarian. The search strategy will proceed according to the JBI Manual for Evidence Synthesis three-step search strategy for reviews [ 27 ]. First, we will conduct a limited search in two appropriate online databases and analyse text words from the title, abstracts and index terms of relevant papers. Next, we will conduct a second search using all identified key words in all databases. Third, we will review reference lists of all included studies to identify further relevant studies to include in the review. We will also contact the authors of relevant papers for further information if required. This will be an iterative process as the research team becomes more familiar with the literature and will be guided by the librarian. Any modifications to the search strategy as it evolves will be described in the scoping review report. As a measure of rigour, the search strategy will be peer-reviewed by another librarian using the PRESS checklist [ 28 ]. No language or date limits will be applied.

Step 3—Study selection

The screening process will consist of a two-step approach: screening titles/abstracts and, if they meet inclusion criteria, this will be followed by a full-text review. All screening will be done by two members of the research team and any disagreements will be resolved by an independent third member of the team. Based on preliminary inclusion criteria, the whole research team will first pilot the screening process by reviewing a random sample of 25 titles/abstracts. The search strategy, eligibility criteria and study objectives will be refined in an iterative process. We anticipate several meetings as the topic is not well described in the literature. A flowchart of the review process will be generated. Any modifications to the study selection process will be described in the scoping review report. The papers will be excluded if a full text is not available. The search results will be managed using Covidence software.

Step 4—Charting the data

A preliminary data extraction tool is shown in Appendix B (see Additional file 3 ). Data will be extracted into Excel and will include demographic information and specific details about the population, concept, context, study methods and outcomes as they relate to the scoping review objectives. The whole research team will pilot the data extraction tool on ten articles selected for full-text review. Through an iterative process, the final data extraction form will be refined. Subsequently, two members of the team will independently extract data from all articles included for full-text review using this tool. Charting disagreements will be resolved by the principal and senior investigators. Google Translate will be used for any included articles that are not in the English language.

Step 5—Collating, summarising and reporting the results

Quantitative and qualitative analyses will be used to summarise the results. Quantitative analysis will capture descriptive statistics with details about the population, concept, context, study methods and outcomes being examined in this scoping review. Qualitative content analysis will enable interpretation of text data through the systematic classification process of coding and identifying themes and patterns [ 29 ]. Several team meetings will be held to review potential themes to ensure an accurate representation of the data. The PRISMA Extension for Scoping Reviews (PRISMA-ScR) will be used to guide the reporting of review findings [ 30 ]. Data will be presented in tables and/or diagrams as applicable. A descriptive summary will explain the presented results and how they relate to the scoping review objectives.

By summarising the literature on LP use in UGME, this study will contribute to a better understanding of how to support SRL amongst medical students. The results from this project will also inform future scholarly work in CBME at the undergraduate level and have implications for improving feedback as well as supporting learners at all levels of competence. In doing so, this study may have practical applications by informing learning plan incorporation into CBME-based curricula.

We do not anticipate any practical or operational issues at this time. We assembled a team with the necessary expertise and tools to complete this project.

Availability of data and materials

All data generated or analysed during this study will be included in the published scoping review article.

Abbreviations

  • Competency-based medical education

Entrustable professional activity

  • Learning plan
  • Self-regulated learning
  • Undergraduate medical education

Frank JR, Snell LS, Cate OT, et al. Competency-based medical education: theory to practice. Med Teach. 2010;32(8):638–45.

Article   PubMed   Google Scholar  

Shorey S, Lau TC, Lau ST, Ang E. Entrustable professional activities in health care education: a scoping review. Med Educ. 2019;53(8):766–77.

Hattie J, Timperley H. The power of feedback. Rev Educ Res. 2007;77(1):81–112.

Article   Google Scholar  

Dudek NL, Marks MB, Regehr G. Failure to fail: the perspectives of clinical supervisors. Acad Med. 2005;80(10 Suppl):S84–7.

Warm EJ, Englander R, Pereira A, Barach P. Improving learner handovers in medical education. Acad Med. 2017;92(7):927–31.

Spooner M, Duane C, Uygur J, et al. Self-regulatory learning theory as a lens on how undergraduate and postgraduate learners respond to feedback: a BEME scoping review : BEME Guide No. 66. Med Teach. 2022;44(1):3–18.

Frellsen SL, Baker EA, Papp KK, Durning SJ. Medical school policies regarding struggling medical students during the internal medicine clerkships: results of a National Survey. Acad Med. 2008;83(9):876–81.

Humphrey-Murto S, LeBlanc A, Touchie C, et al. The influence of prior performance information on ratings of current performance and implications for learner handover: a scoping review. Acad Med. 2019;94(7):1050–7.

Morgan HK, Mejicano GC, Skochelak S, et al. A responsible educational handover: improving communication to improve learning. Acad Med. 2020;95(2):194–9.

Dory V, Danoff D, Plotnick LH, et al. Does educational handover influence subsequent assessment? Acad Med. 2021;96(1):118–25.

Humphrey-Murto S, Lingard L, Varpio L, et al. Learner handover: who is it really for? Acad Med. 2021;96(4):592–8.

Shaw T, Wood TJ, Touchie T, Pugh D, Humphrey-Murto S. How biased are you? The effect of prior performance information on attending physician ratings and implications for learner handover. Adv Health Sci Educ Theory Pract. 2021;26(1):199–214.

Artino AR, Brydges R, Gruppen LD. Chapter 14: Self-regulated learning in health professional education: theoretical perspectives and research methods. In: Cleland J, Duning SJ, editors. Researching Medical Education. 1st ed. John Wiley & Sons; 2015. p. 155–66.

Chapter   Google Scholar  

Cleland J, Arnold R, Chesser A. Failing finals is often a surprise for the student but not the teacher: identifying difficulties and supporting students with academic difficulties. Med Teach. 2005;27(6):504–8.

Reed S, Lockspeiser TM, Burke A, et al. Practical suggestions for the creation and use of meaningful learning goals in graduate medical education. Acad Pediatr. 2016;16(1):20–4.

Wolff M, Stojan J, Cranford J, et al. The impact of informed self-assessment on the development of medical students’ learning goals. Med Teach. 2018;40(3):296–301.

Sawatsky AP, Halvorsen AJ, Daniels PR, et al. Characteristics and quality of rotation-specific resident learning goals: a prospective study. Med Educ Online. 2020;25(1):1714198.

Article   PubMed   PubMed Central   Google Scholar  

Pintrich PR. Chapter 14: The role of goal orientation in self-regulated learning. In: Boekaerts M, Pintrich PR, Zeidner M, editors. Handbook of self-regulation. 1st ed. Academic Press; 2000. p. 451–502.

Kassab SE, El-Sayed W, Hamdy H. Student engagement in undergraduate medical education: a scoping review. Med Educ. 2022;56(7):703–15.

Challis M. AMEE medical education guide No. 19: Personal learning plans. Med Teach. 2000;22(3):225–36.

Knowles MS. Using learning contracts. 1 st ed. San Francisco: Jossey Bass; 1986.

Parsell G, Bligh J. Contract learning, clinical learning and clinicians. Postgrad Med J. 1996;72(847):284–9.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Teunissen PW, Scheele F, Scherpbier AJJA, et al. How residents learn: qualitative evidence for the pivotal role of clinical activities. Med Educ. 2007;41(8):763–70.

Article   CAS   PubMed   Google Scholar  

van Houten-Schat MA, Berkhout JJ, van Dijk N, Endedijk MD, Jaarsma ADC, Diemers AD. Self-regulated learning in the clinical context: a systematic review. Med Educ. 2018;52(10):1008–15.

Taylor DCM, Hamdy H. Adult learning theories: Implications for learning and teaching in medical education: AMEE Guide No. 83. Med Teach. 2013;35(11):e1561–72.

Arksey H, O’Malley L. Scoping studies: towards a methodological framework. Int J Soc Res Methodol. 2005;8(1):19–32.

Peters MDJ, Godfrey C, McInerney P, Munn Z, Tricco AC, Khalol H. Chapter 11: Scoping reviews. In: Aromataris E, Munn Z, eds. JBI Manual for Evidence Synthesis. JBI; 2020. https://synthesismanual.jbi.global. . Accessed 30 Aug 2022.

McGowan J, Sampson M, Salzwedel DM, Cogo E, Foerster V, Lefebvre C. PRESS Peer Review of Electronic Search Strategies: 2015 Guideline Statement. J Clin Epidemiol. 2016;75:40–6.

Hsieh HF, Shannon SE. Three approaches to qualitative content analysis. Qual Health Res. 2005;15(9):1277–88.

Tricco AC, Lillie E, Zarin W, et al. PRISMA extension for scoping reviews (PRISMA-ScR): checklist and explanation. Ann Intern Med. 2018;169(7):467–73.

Venables M, Larocque A, Sikora L, Archibald D, Grudniewicz A. Understanding indigenous health education and exploring indigenous anti-racism approaches in undergraduate medical education: a scoping review protocol. OSF; 2022. https://osf.io/umwgr/ . Accessed 26 Oct 2022.

Download references

Acknowledgements

Not applicable.

This study will be supported through grants from the Department of Medicine at the Ottawa Hospital and the University of Ottawa. The funding bodies had no role in the study design and will not have any role in the collection, analysis and interpretation of data or writing of the manuscript.

Author information

Authors and affiliations.

The Ottawa Hospital – General Campus, 501 Smyth Rd, PO Box 209, Ottawa, ON, K1H 8L6, Canada

Anna Romanova & Claire Touchie

The Ottawa Hospital Research Institute, Ottawa, Canada

Sydney Ruller

The University of Ottawa, Ottawa, Canada

Victoria Cole

The Ottawa Hospital – Riverside Campus, Ottawa, Canada

Susan Humphrey-Murto

You can also search for this author in PubMed   Google Scholar

Contributions

AR designed and drafted the protocol. CT and SH contributed to the refinement of the research question, study methods and editing of the manuscript. VC designed the initial search strategy. All authors reviewed the manuscript for final approval. The review guarantors are CT and SH. The corresponding author is AR.

Authors’ information

AR is a clinician teacher and Assistant Professor with the Division of General Internal Medicine at the University of Ottawa. She is also the Associate Director for the internal medicine clerkship rotation at the General campus of the Ottawa Hospital.

CT is a Professor of Medicine with the Divisions of General Internal Medicine and Infectious Diseases at the University of Ottawa. She is also a member of the UGME Competence Committee at the University of Ottawa and an advisor for the development of a new school of medicine at Toronto Metropolitan University.

SH is an Associate Professor with the Department of Medicine at the University of Ottawa and holds a Tier 2 Research Chair in Medical Education. She is also the Interim Director for the Research Support Unit within the Department of Innovation in Medical Education at the University of Ottawa.

CT and SH have extensive experience with medical education research and have numerous publications in this field.

SR is a Research Assistant with the Division of General Internal Medicine at the Ottawa Hospital Research Institute.

VC is a Health Sciences Research Librarian at the University of Ottawa.

SR and VC have extensive experience in systematic and scoping reviews.

Corresponding author

Correspondence to Anna Romanova .

Ethics declarations

Ethics approval and consent to participate, consent for publication, 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. prisma-p 2015 checklist., 13643_2024_2553_moesm2_esm.docx.

Additional file 2: Appendix A. Preliminary search strategy [ 31 ].

Additional file 3: Appendix B. Preliminary data extraction tool.

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

Cite this article.

Romanova, A., Touchie, C., Ruller, S. et al. Protocol for a scoping review study on learning plan use in undergraduate medical education. Syst Rev 13 , 131 (2024). https://doi.org/10.1186/s13643-024-02553-w

Download citation

Received : 29 November 2022

Accepted : 03 May 2024

Published : 14 May 2024

DOI : https://doi.org/10.1186/s13643-024-02553-w

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Systematic Reviews

ISSN: 2046-4053

  • Submission enquiries: Access here and click Contact Us
  • General enquiries: [email protected]

the purpose of a systematic review

  • Open access
  • Published: 21 May 2024

Combined systematic screening for malnutrition and dysphagia in hospitalized older adults: a scoping review

  • Susanne M. Javorszky 1 , 3 ,
  • Christoph Palli 2 ,
  • Susanne Domkar 3 &
  • Bernhard Iglseder 4  

BMC Geriatrics volume  24 , Article number:  445 ( 2024 ) Cite this article

Metrics details

Dysphagia affects about 40% of patients admitted to acute geriatric wards, as it is closely associated with diseases that rise in prevalence with advancing age, such as stroke, Parkinson’s disease, and dementia. Malnutrition is a highly associated predictive factor of dysphagia as well as one of the most common symptoms caused by dysphagia. Thus, the two conditions may exist simultaneously but also influence each other negatively and quickly cause functional decline especially in older adults. The purpose of this review was to determine whether institutions have established a protocol combining screenings for dysphagia and malnutrition on a global scale. If combined screening protocols have been implemented, the respective derived measures will be reported.

A scoping review was conducted. A systematic database search was carried out in January and February 2024. Studies were included that examined adult hospitalized patients who were systematically screened for dysphagia and malnutrition. The results were managed through the review software tool Covidence. The screening of titles and abstracts was handled independently by two reviewers; conflicts were discussed and resolved by consensus between three authors. This procedure was retained for full-text analysis and extraction. The extraction template was piloted and revised following feedback prior to extraction, which was carried out in February 2024.

A total of 2014 studies were found, 1075 of which were included for abstract screening, 80 for full text screening. In the end, 27 studies were extracted and reported following the reporting guideline PRISMA with the extension for Scoping Reviews.

Most of the studies considered the prevalence and association of dysphagia and malnutrition with varying outcomes such as nutritional status, pneumonia, oral nutrition, and swallowing function. Only two studies had implemented multi-professional nutrition teams.

Peer Review reports

Introduction

The World Health Organization (WHO) defines “older people” as 60–75, “old people” as 76–90, “very old people” as 91–100 and “oldest-old” or “centenarians” as people aged over 100 years [ 1 ]. Still, the definition of “older people” varies based on cultural, social, functional, and emotional factors. However, demographic changes contribute to an increasing proportion of individuals around and after retirement age. In the European Union, individuals aged 65 and over are considered “older people,” with a projected 28.5% share of the total EU population in 2050 [ 2 ]. This demographic shift poses significant challenges to the healthcare systems, given the association between aging and various health conditions such as diabetes, cardiovascular disease, Parkinson’s disease, stroke, and dementia. This challenge is compounded by the simultaneous retirement of baby boomers and a pronounced shortage of skilled workers in nursing and therapeutic care. Consequently, healthcare systems face the urgent need to formulate efficient, resource-saving, and high-quality care strategies for older adults in general and specifically for geriatric patients.

Aside from chronological aging, differences in functioning and subjective health lead to a need for a functional and multidimensional definition of “geriatric patients” [ 3 , 4 ]. Physiological aging affects cellular processes, which generally impacts nutrient metabolism as well as muscle function, thus changing the swallowing function physiologically, leading to an aged swallow called presbyphagia [ 5 , 6 ]. There is a high correlation between frailty, dysphagia and malnutrition, highlighting the importance of these conditions in the context of advancing age, accumulated burden of disease and frailty [ 7 , 8 ]. Advancing age raises the risk of malnutrition, either as a direct result of diseases as well as multimorbidity, or caused by challenges related to food intake itself, particularly issues with chewing and swallowing [ 9 , 10 ]. Insufficient supply of nutrients leads to a state of malnutrition, which in turn has a negative effect on all other health factors. It has been shown that frail older individuals benefit from individualized nutritional support. The achievement of energy and protein goals is essential for improved outcomes [ 11 ]. The use of fortified meals and oral nutritional supplements with > 400 kcal and at least 30 g of protein / day is recommended if the intake through meals is not sufficient [ 12 ]. Dysphagia (swallowing disorder), which impairs the ability of processing and swallowing food and liquids, is one of the most significant predictors of malnutrition and is in turn negatively influenced by it [ 13 , 14 , 15 ].

In 2016, the European Society of Swallowing Disorders (ESSD) declared dysphagia a geriatric syndrome that is multifactorial and has significant negative impact on the nutritional and health status of geriatric patients in general [ 16 ]. The prevalence of dysphagia in older adults ranges considerably, with reported rates between 10 and 90% in nursing homes and a pooled prevalence of 35,9% as reported in a recent meta-analysis by Tian et al. [ 17 ], while Doan et al. [ 18 ] report a pooled prevalence of 47%. As dysphagia is closely associated with frailty as well as neurodegenerative diseases, the prevalence will increase in institutions specialized in caring for older persons with advanced stages of diseases as well as higher age and higher rates of frailty [ 7 ]. The estimated prevalence in hospitalized older adults is 37% and in community dwelling older adults around 18% [ 18 ]. In addition to a loss of quality of life due to possible restrictions in food intake [ 19 , 20 , 21 ], one of the most serious consequences of dysphagia is malnutrition [ 8 ]. In individuals with a compromised immune system and poor oral health dysphagia can lead to the development ofaspiration pneumonia [ 22 , 23 ], which has a significantly higher mortality rate than pneumonia of other etiologies [ 24 , 25 ]. The diagnosis of dysphagia is carried out in several steps, usually starting with an initial screening to identify high-risk patients. This first screening can be completed by different professions [ 26 ]. If a patient is determined to be at risk, a referral is made to a clinical swallow examination carried out by a speech language therapist (SLT). This clinical assessment may result in the recommendation of an instrumental diagnosis of the swallowing disorder using FEES (flexible endoscopic evaluation of swallowing) or VFSS (videofluoroscopic swallowing study), which provides foundation for individualized treatment planning. Dysphagia treatment is carried out by means of speech language therapy. It may take place in a rehabilitative manner through targeted exercises, in a compensatory manner by developing strategies to optimize the swallowing function, and adaptively through the use of dietary modification and adaptation of tableware. These adaptations can include the recommendation ofspecific cups and cutlery as well as optimal positioning of patients when eating or drinking. Dysphagia management should also include dietetic and nutrition therapy [ 16 , 27 , 28 ].

Particularly for older people, malnutrition is a serious health problem with far-reaching effects on their quality of life and health [ 9 , 29 ]. Inadequate nutrient intake is associated with increased susceptibility to infection, extended recovery time, reduced muscle mass, a weakened immune system, and a higher risk of falls [ 30 ]. The diagnosis of malnutrition usually follows similar steps to those for diagnosing dysphagia, starting with rapidscreenings. A variety of screening and assessment methods are used in conjunction with blood parameters as well as measurements of body parameters such as weight, circumference of limbs or measuring skinfold thickness [ 31 ]. The treatment of malnutrition in view of determining needs and optimizing nutrient intake is carried out by dietitians.

Severe cases of both dysphagia and malnutrition can be treated with promising results using a combined treatment approach [ 32 ]. Numerous screening methods are endorsed internationally for early detection for both conditions. These methods are widely integrated into the healthcare of older individuals and explicitly recommended for use [ 33 , 34 , 35 ].

Due to the close association of the two disorders, assessing them in a combined screening procedure seems a promising strategy. Therefore, it might be feasible to detect both with one quick screening protocol. A positive screening result should lead to timely referrals to either speech language therapists (SLT) for dysphagia assessment and treatment or to registered dietitians (RD) for malnutrition assessmentand treatment. This consideration led to the decision to conduct a literature review with the aim of ascertaining whether internationally combined screenings for malnutrition and dysphagia already existed. In this case, follow-up questions were defined. Which specific screening tools were used by which profession? Which specific outcomes of an implemented combination screening were measured? The research question was structured according to the PICO scheme (Table  1 ). Since the question implies a heterogeneity of studies to be processed and possible outcome measures of the individual studies, the methodological approach of a scoping review according to the recommendations of the PRISMA-Scr statement was chosen. The large number of possible outcomes defined in the PICO question led to the decision to conduct a scoping review, which is particularly suitable for obtaining an initial overview of a topic and for broadly reflecting the state of research without narrowing the included results too much [ 36 , 37 ]. For guidance on the methodology, several sources were used [ 37 , 38 , 39 ].

An informal protocol was developed which included the defined search terms, research question, time schedule, task distribution amongst the authors, selected databases, selection criteria, and the search strand based on MeSH terms. This protocol was not published and merely facilitated orientation in the authors’ work process. Furthermore, the review protocol has not been registered in any official database.

The terms for the search strand were: presbyphagia, dysphagia, swallowing disorder, *nutrition*, sarcopenia*, weight loss, diet*, assessment, screen*, elderly, hospital admission, geriatric, nursing home, nursing facility, long term care. The asterisks stand as so-called “wildcard” symbols, which allow any stem ending of the word to be included. Using the completed search strands, the databases Science Direct, CINAHL, PubMed, Google Scholar, MedLine and Cochrane Library (Trials) were searched between 31.1.2024 and 21.2.2024. Previously published quantitative primary studies were included, which systematically recorded both parameters (dysphagia and malnutrition) in hospitalized adults. The exclusion criteria were an average age of less than 60 years, patients undergoing head and neck cancer surgery as well as persons in other healthcare settings. Furthermore, studies in which only one of the two parameters had been systematically recorded were excluded, as well as studies in which instrumental diagnostics were carried out instead of a screen. The decision to focus only on intramural care pathways was made due to the increasing heterogeneity of the professional groups and healthcare systems involved. In addition, studies that not published in German or English were excluded. There was no restriction on publication date.

For literature management, the web-based collaborative software platform tool Covidence was used to support the creation of literature reviews developed by Veritas Health Innovation [ 40 ]. Duplicates were automatically removed. Two authors rated the remaining studies for inclusion after screening all titles and abstracts with SMJ screening and CP and SD acting as second raters. Conflicting ratings were solved by consensus of SMJ, CP and SMD. In most cases, the main reason for different ratings was the reason for exclusion, as some studies met several of the exclusion criteria at the same time.

In the next step, a full-text review was carried out using Covidence following the same procedure as the title and abstract screening. Again, two authors had to give an independent evaluation; conflicts were solved by consensus between SMJ, CP and SD.

For data extraction, an extraction template was created in Covidence by SMJ, which was based on the PICO question. The template was piloted by CP and SD each in one study. Following feedback, it was revised and used for the extraction of all remaining studies as of February 2024 (Table  2 ). Extraction was carried out independently by two authors, either SMJ and CP or SMJ and SD. After extracting 13 studies, a meeting was held to avoid disparities in abbreviations (e.g. different use of commas and full stops leads to the software declaring a conflict) and formal details as differences were noticed when reporting studies details such as time frames by having been written as “January, 2015 – June, 2016” compared to “01/2015–06/2016” for example In another meeting following the extraction, all conflicts and open questions were discussed and solved based on consensus among the authors.

The heterogeneity of the study types processed and the methodological approach within the framework of a scoping review resulted in avoiding a critical assessment of the level of evidence and the bias risks of the individual studies [ 37 , 38 , 39 ]. The extracted data was exported to Microsoft Excel for an easier overview, comparison of the extracted information and to generate the tables.

As shown in Fig.  1 , a total of 2014 studies were retrieved from database search, 939 of which were duplicates that were removed. The remaining 1075 studies were independently evaluated by two authors based on title and abstract. 995 studies were excluded. 77 studies were assessed for inclusion based on their full text, which in turn was independently conducted by two authors. A total of 50 studies were excluded for the following reasons: language (4), study design (19), population (7), conference proceedings (1), instrumental diagnostics (5) or only 1 parameter screened systematically (14).

figure 1

PRISMA flowchart

Data was extracted from theremaining 27 articles using the piloted template. Table 3 gives an overview of the characteristics of all included studies. Only three of the included studies were older than 10 years, 12 studies were conducted between 2014 and 2019 and 12 studies were conducted since 2020 with the most recent dated 2023. The settings of the studies vary, but the vast majority was conducted in acute care hospitals (18), specifically in acute geriatrics (3). Three studies investigated patients in rehabilitation centers (3). Three studies were multicentric and examined populations from different settings (3). The total number of participants varied considerably, from small cohorts of 12 to large-scale investigations with over 1,000 participants.

All studies investigated the association between nutritional status and dysphagia in older people in cohort and cross-sectional studies. The studies include participants from Japan (6), Spain (7), New Zealand (2), Brazil (2), Denmark (1), France (1), Italy (1), Canada (1), Colombia (1), Mexico (1), Turkey (1), the United States (1), China (1) and Vietnam (1). The parameters studied vary from the prevalence of malnutrition and nutritional risks to the influence of speech therapy on dysphagia and associations between oral health, dysphagia, and malnutrition. The age groups range from over 65 years to over 70 or 75 years and older. The main characteristics of the described studies are shown in Table  3 .

Dysphagia screening

As per the inclusion and exclusion criteria, any bedside tool to detect swallowing difficulties was included under the broad term “dysphagia screening”. The term “screening” is generally understood to mean an easy to apply early detection protocol to identify individuals of risk of a specific disorder. A positive screening result, suggesting a possible risk for, in this case, a swallowing disorder, warrants further, more detailed assessment by specialized professionals [ 62 ]. Since dysphagia diagnostics can only be completed with an instrumental evaluation, any bedside assessment can be classified as a form of screening with differing sensitivity and specificity depending on the variables included and the amount of training needed to conduct the screening. Rommel & Hamdy describe the dysphagia detection process as a multi-step approach beginning with any screening. In case of a positive screening result, a clinical swallow assessment needs to be conducted by a dysphagia specialist, usually a speech language therapist (SLT). This assessment includes the patient history, a physical and oral motor exam as well as a food intake assessment [ 63 ]. This description illustrates the argument, that also protocols referred to as “assessments” like the Volume-Viscosity Swallowing Tool (V-VST) [ 64 ] can be defined as screening tools in the process of dysphagia detection. For this scoping review the authors decided to include the Swallowing-related Quality of Life Tool (Swal-QoL) [ 65 ] as well, as self-reported limitations in the quality of life because of swallowing disorders can be counted as proxy parameter for existing dysphagia.

The most frequently used screening tool for dysphagia was the self-reported Eating Assessment Tool (EAT-10) [ 66 ], which was used in eight studies as the sole tool and in three more in a combination of two or more tools. The second most commonly reported screening tool was the Volume-Viscosity Swallowing Test (V-VST) [ 64 ]. Table 4 gives an overview of all the 11 reported screening tools used to assess dysphagia in the 27 studies.

The professional groups carrying out the screenings were reported in only 6 studies (Nursing staff/study nurse: Cabre et al. 2010; Carrión et al. 2015; Mateos-Nozal et al. 2017; Shimazu et al. 2020; SLT: Crary et al. 2006; Moncayo-Hernandéz et al. 2023) [ 10 , 43 , 44 , 45 , 46 , 49 ]; 7 studies used exclusively self-reported tools (Manas-Martinez et al. 2018; Matsuo et al. 2017; Popman et al. 2016; Tran et al. 2021; Vidal Casariego et al. 2020; Wakabayashi & Matsuhima 2016; Wham et al. 2017) [ 41 , 47 , 50 , 57 , 58 , 60 , 67 ] and the remaining 14 did not specify the profession [ 13 , 42 , 48 , 51 , 52 , 53 , 54 , 55 , 56 , 59 , 61 , 68 , 69 , 70 ].

Malnutrition screening

Malnutrition screening was carried out by Mini Nutritional Assessment (MNA) [ 71 ] in the majority of studies having been reported 20 times, two times in the long form, 11 times in the short form and seven times without further specification. Two studies used the Geriatric Nutritional Risk Index (GNRI) [ 72 ] and the remaining two used the Subjective Global Assessment (SGA) [ 73 ].

Even in the case of malnutrition screening, information on the occupational group performing the tests was found in a minority of studies. Two studies reported registered dietitians as responsible [ 44 , 67 ], one named speech language therapists [ 10 ], one a multi-professional team [ 43 ], and two described nursing staff as being responsible with one of them explicitly mentioning a study nurse [ 45 , 46 ].

Interventions taken after screening

Only two of the reviewed studies provided clear information on the long-term implementation of the two screenings with Waza et al. using the FOIS and the GNRI [ 69 ] and Uno et al. using the FILS and the MNA-SF [ 68 ]. Both describe the screening process as part of routine patient care and report on specific measures derived from the screening results, such as a referral to swallowing of nutritional specialists or a recommendation on texture for oral nutrition. Five studies reported that screening was systematically repeated, either after an intervention, after a certain amount of time or right before discharge [ 51 , 55 , 56 , 68 , 69 ]. Table 5 provides an overview of the subsequent intervention following positive screening results.

While only three studies explicitly indicated nutritional therapy or referral to registered dietitians as an intervention, nutritional status is the most frequently reported outcome of the studies and was defined as such in all studies except Moncayo-Hernandéz et al. [ 49 ] and Omura et al. [ 54 ], i.e., a total of 25. The other two most common outcomes were sufficient oral intake and the occurrence of pneumonia. The full distribution of the different outcomes is shown in Table  6 .

Only two of the studies explicitly indicated the presence of a specialized nutrition team. Shimazu et al. report the team being led by a dietitian and consisting of doctors, nurses, pharmacists, dentists and SLTs [ 46 ]. Waza et al. describe the following professional groups as members of the nutrition team: SLT, registered dietitians, dental assistants, medicine, physiotherapists, occupational therapists, and nursing staff [ 69 ]. Furthermore, only Waza et al. report that nutritional counseling takes place systematically upon discharge.

Early detection of malnutrition as well as dysphagia can prevent a cascade of problems resulting from these conditions [ 15 , 30 ]. Being closely associated conditions, it might be beneficial to always screen for both, since having severe dysphagia will negatively impact nutritional status and vice versa. Guidelines for Geriatric Assessment state that both conditions should be assessed [ 34 , 35 , 74 ]. However, due to time and personnel limitations, this does not always seem to be possible. Looking at the Comprehensive Geriatric Assessment (CGA), for example, feeding and nutrition are not considered part of the core components but mentioned as additional components that may be included [ 35 ], hinting at an optional extended version. The British Geriatrics Society (BGS) specifically states in their CGA toolkit for primary care practitioners, that completing the CGA fully “may take up to two hours” but does not provide any specific screening tools for dysphagia aside from asking about swallowing problems, choking or “food getting stuck” [ 34 ]. To screen for malnutrition, the BGS recommends monitoring BMI or using the Malnutrition Universal Screening Tool (MUST) [ 74 ] or the MNA. In contrast, the living guideline by the German, Austrian and Swiss geriatric societies [ 75 ] do recommend three specific screening tools for dysphagia: the SSA (Standardized Swallowing Assessment) [ 76 ], the Daniels Water Test [ 77 ] and the DSTG (Deutsches Screening-Tool Geriatrie) [ 78 ]. Each of them uses solely the application of water in different amounts and with different scales. None of them were reported to be used in the studies included in this review, which in the case of the DSTG is not surprising as it is only available in German. To screen for malnutrition, the guideline recommends the MST (Malnutrition Screening Tool) [ 79 ], the MNA and the NRS, with the latter two having been used in several of the studies included in this review.

A screening protocol that combines both disorders and is feasible for different geriatric settings could improve prevention of complications resulting from either condition. As the MNA has excellent sensitivity and specificity (96% and 98% respectively) it might be, especially in its short form, a feasible tool to combine with a swallowing screen. The EAT-10, which has been reported as the sole dysphagia screening tool in eight studies in this review, has the advantage of being self-administered, thus being very feasible for routine use with sensitivity of 85% and specificity of 82% when using a cut-off value of three as recommended by the authors [ 66 , 80 ]. The downside of a self-administered tool is that language barriers, reduced reading skills, hearing loss and cognitive impairment might make it harder to conduct than simple water swallowing screens [ 81 ].

An easy to conduct and fast screening tool should bridge the gap between time and funding constraints and optimal care resulting from early detection of disorders that impact the overall well-being of older people and especially multimorbid geriatric patients. It should contain questions about swallowing ability as well as a quick orientation about nutritional status. The MNA-SF combined with the EAT-10 might offer such an overview. An alternative to the EAT-10 could be the 4QT (4-point questionnaire test), an even shorter questionnaire focusing on changes in swallowing and eating ability [ 82 ], which was designed specifically for older and frail people. A limiting factor is that it has not yet been validated against an instrumental examination. In older people who are not able to complete these self-reported questionnaires, the Minimal Eating Observation Form – version II (MEOF-II) could be used alternatively. It focuses on the whole process of eating rather than specifically on the risk of penetration or aspiration, but can be conducted without verbal language skills [ 83 ]. As an addition or alternative to the MNA-SF, the SNAQ (Simplified Nutritional Appetite Questionnaire) could be considered. The SNAQ consists of four items and shows a sensitivity of 82% as well a specifity of 85% for a 6-months weight loss of 5% [ 84 ].

This scoping review aimed to find out whether any screening protocol combining dysphagia and malnutrition is already in use internationally. Further investigations focus on which professions are involved and which screening tools are used in screening for malnutrition and dysphagia, and the possible outcomes as well as measures derived in the case of reported screening protocols.

The included studies vary widely in describing the use of the respective screenings and the measures taken from the screening results. Due to the large number of cross-sectional studies that assessed the prevalence and association between malnutrition and dysphagia rather than the impact of implementing a systematic screening, only the first part of the research question can be answered. The search results did not yield sufficient studies to answer the questions of how implementing such protocols impacted the outcomes described in the PICO question – namely pneumonia, falls, rehospitalization, length of stay, place of discharge, nutritional status, wound and bone healing, mobility, sufficient oral intake, oral medication, or bolus death.

The included studies are spread globally with the majority from Japan and Spain. It could be speculated that Japan has a high life expectancy for years [ 85 , 86 ], possibly leading to a stronger research focus on the elderly. However, the wide range of countries illustrates that the topic itself seems to be of worldwide interest. All included studies report the use of dysphagia screening as well as a malnutrition screening, but none have a single combined protocol. The settings of the studies vary, with most conducted in acute care hospitals. In terms of dysphagia screening, various instruments were used, such as the EAT-10, V-VST, FOIS, FILS, MASA, STAND, GUSS and MDADI, as well as water swallowing tests without further description (Table  4 ). The MNA, either in long or short form, was the most frequently used screening tool for malnutrition, with other instruments such as GNRI, NRS and SGA also being reported. Moreover, this review surveyed the performing professional groups, naming speech therapists, nursing staff and, in some cases, a multi-professional team, but most papers did not report on the professional group carrying out the screenings.

Only a few studies report repeated screening as part of routine care, either after an intervention, after a certain time or before discharge. Measures derived from abnormal screening results, such as referrals to speech therapy, decisions about oral feeding options, and dietary counseling, were documented in 10 of the studies, as shown in Table  5 . Overall, the measures reported in the studies vary broadly from simple texture modified diet prescriptions that have been internationally standardized by the International Dysphagia Diet Standardization Initiative (IDDSI) [ 87 ] to more complex interventions by nutritional teams. The mentioned nutritional teams comprised several professions and interventions such as speech therapy, counselling by a dietitian, positioning, promotion of oral health, nutritional therapy, respiratory therapy, and medication evaluation, as described in Waza et al. [ 69 ] and Shimazu et al. [ 46 ].

The most reported outcomes were nutritional status, occurrence of pneumonia, and sufficient oral intake. The distribution of the different outcomes varies, with nutritional status being by far the most frequent. While numerous outcomes were assessed in some studies, others were limited to one or two parameters. Only Waza et al. [ 69 ] and Shimazu et al. [ 46 ] described an interprofessional nutrition team of different professional groups. Systematic nutritional counseling at discharge was only described by one study [ 69 ].

The heterogeneity of the studies in terms of settings, screening instruments and performing professional groups makes direct comparisons between studies difficult. The issue of required training and possible user groups for specific screening tools might also be of interest, as well as the different settings in which the discussed studies were conducted. Examining the extent to which these differences affect the reported results might be intriguing. In addition, the implications of the screening results and the measures derived are not uniformly documented, making it difficult to draw clear conclusions regarding to the questions of this review.

It should be noted that by conducting a scoping review, a lot of different study designs were included in the selection process. Consequently, the research question can only be answered to a limited extent with the available results. The focus of the included studies estimating the prevalence and associations between malnutrition and dysphagia shows their interdependence. However, it is not possible to make assumptions about the impact of a systematically used combined screening for malnutrition and dysphagia on the originally described outcomes of this review. The mention of a nutrition team as well as systematic nutritional counseling at discharge is reported in only two studies. This does not necessarily mean that such procedures are not present, rather that they have not been systematically reported and evaluated. The prevalence and close association of the two investigated conditions strongly suggest a potential improvement of care through early detection of malnutrition and dysphagia.

Further research could focus on conducting implementation studies to determine the impact of a combined screening protocol on the care pathway of geriatric patients. Subsequently, it might be of interest to assess how and with which impact on health outcomes interdisciplinary nutrition teams can be effectively integrated into the care of older people. In future investigations, the validity of a joint screening tool, for example a combination of SNAQ and 4QT should be evaluated. If a combined screening tool proves to be feasible while maintaining high sensitivity and specificity, its integration into routine care should be evaluated considering the outcomes defined in this scoping review.

Dysphagia and malnutrition in the elderly have complex relationships that can affect not only the state of health, but also quality of life. A lot of different screening protocols are in use globally with different professions being involved in applying those tools. There was no reported combined screening tool, however both conditions are screened regularly in different geriatric inpatient setting globally. Systematic early detection and clearly defined care pathways potentially have a positive effect on nutritional status, sufficient oral intake, the ability to swallow safely and pneumonia rates. These outcomes were reported in the analyzed studies, however it was not possible to establish whether the implementation of screening protocols did influence those factors. Unfortunately, there is lack of research on the impact of systematically implemented screening protocols to ensure sound, evidence-based guidance on these important issues.

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Abbreviations

World Health Organization

Speech Language Therapist

Registered Dietitian

International Dysphagia Diet Standardization Initiative

Texture Modified Diet

Eating Assessment Tool

Functional Oral Intake Scale

Food Intake Level Scale

Gugging Swallowing Screen

Mann Assessment of Swallowing Ability

M.D. Anderson Dysphagia Inventory

Protocol of Risk Assessment for Dysphagia

Screening Tool for Neurological Dysphagia

Swallowing Related Quality of Life

Water Swallowing Test

Volume-Viscosity Swallowing Test

Mini Nutritional Assessment

Mini Nutritional Assessment – Short Form

Geriatric Nutritional Risk Index

Subjective Global Assessment

Activities of Daily Life

National Institute of Stroke Scale

Comprehensice Geriatric Assessment

4-Point Questionnaire Test

Simplified Nutritional Appetite Questionnaire

WHO. World report on ageing and health. Geneva: World Health Organization; 2015. Available from: URL: http://apps.who.int/iris/bitstream/10665/186463/1/9789240694811_eng.pdf?ua=1 .

European Commission. Ageing Europe: Looking at the lives of older people in the EU. 2019th ed. Luxembourg: Publications Office of the European Union; 2019. (Statistical books / Eurostat).

Google Scholar  

Pochobradsky E, Nemeth C. Prozesshandbuch Akutgeriatrie, Remobilisation. Wien: Gesundheit Österreich; 2013.

Hou Y, Dan X, Babbar M, Wei Y, Hasselbalch SG, Croteau DL, et al. Ageing as a risk factor for neurodegenerative disease. Nat Rev Neurol. 2019;15(10):565–81.

Article   PubMed   Google Scholar  

Campisi J, Kapahi P, Lithgow GJ, Melov S, Newman JC, Verdin E. From discoveries in ageing research to therapeutics for healthy ageing. Nature. 2019;571(7764):183–92.

Article   CAS   PubMed   PubMed Central   Google Scholar  

Jardine M, Miles A, Allen J. Self-reported swallowing and nutrition status in community-living older adults. Dysphagia. 2021;36(2):198–206.

Yang R-Y, Yang A-Y, Chen Y-C, Lee S-D, Lee S-H, Chen J-W. Association between dysphagia and frailty in older adults: a systematic review and meta-analysis. Nutrients 2022; 14(9).

Sire A de, Ferrillo M, Lippi L, Agostini F, Sire R de, Ferrara PE et al. Sarcopenic dysphagia, malnutrition, and oral frailty in elderly: a comprehensive review. Nutrients 2022;14(5):1–23.

Huppertz VA, Pilz W, Da Pilz Cunha G, de Groot LCPGM, van Helvoort A, Schols JMGA, et al. Malnutrition risk and oropharyngeal dysphagia in the chronic post-stroke phase. Front Neurol. 2022;13:939735.

Crary MA, Carnaby-Mann GD, Miller L, Antonios N, Silliman S. Dysphagia and nutritional status at the time of hospital admission for ischemic stroke. J Stroke Cerebrovasc Dis. 2006;15(4):164–71. Available from: https://www.sciencedirect.com/science/article/pii/S1052305706000796 .

Baumgartner A, Pachnis D, Parra L, Hersberger L, Bargetzi A, Bargetzi L, et al. The impact of nutritional support on malnourished inpatients with aging-related vulnerability. Nutrition. 2021;89:111279.

Article   CAS   PubMed   Google Scholar  

Volkert D, Beck AM, Cederholm T, Cruz-Jentoft A, Hooper L, Kiesswetter E, et al. ESPEN practical guideline: Clinical nutrition and hydration in geriatrics. Clin Nutr. 2022;41(4):958–89.

Donini LM, Dominguez LJ, Barbagallo M, Savina C, Castellaneta E, Cucinotta D, et al. Senile anorexia in different geriatric settings in Italy. J Nutr Health Aging. 2011;15(9):775–81.

Tran TP, Nguyen LT, Hirose K, Nguyen TH, Le HT, Shimura F, et al. Malnutrition is associated with dysphagia in Vietnamese older adult inpatients. Asia Pac J Clin Nutr. 2021;30(4):588–94.

PubMed   Google Scholar  

Sella-Weiss O. Association between swallowing function, malnutrition and frailty in community dwelling older people. Clin Nutr ESPEN. 2021;45:476–85.

Baijens LW, Clavé P, Cras P, Ekberg O, Forster A, Kolb GF, et al. European Society for Swallowing Disorders - European Union Geriatric Medicine Society white paper: oropharyngeal dysphagia as a geriatric syndrome. Clin Interv Aging. 2016;11:1403–28.

Article   PubMed   PubMed Central   Google Scholar  

Tian L, Hu Z, Yang L, Xiang Y. The prevalence of dysphagia at risk among older adults in nursing homes: a meta-analysis. Psychogeriatrics. 2024;24(1):127–37.

Doan T-N, Ho W-C, Wang L-H, Chang F-C, Nhu NT, Chou L-W. Prevalence and methods for assessment of oropharyngeal dysphagia in older adults: a systematic review and meta-analysis. J Clin Med 2022;11(9):1–14.

O’Keeffe M, Kelly M, O’Herlihy E, O’Toole PW, Kearney PM, Timmons S, et al. Potentially modifiable determinants of malnutrition in older adults: a systematic review. Clin Nutr. 2019;38(6):2477–98.

Hansen T, Beck AM, Kjaersgaard A, Poulsen I. Second update of a systematic review and evidence-based recommendations on texture modified foods and thickened liquids for adults (above 17 years) with oropharyngeal dysphagia. Clin Nutr ESPEN. 2022;49:551–5.

Flynn E, Smith CH, Walsh CD, Walshe M. Modifying the consistency of food and fluids for swallowing difficulties in dementia. Cochrane Database Syst Rev. 2018;9(9):CD011077.

Langmore SE, Terpenning MS, Schork A, Chen Y, Murray JT, Lopatin D, et al. Predictors of aspiration pneumonia: how important is dysphagia? Dysphagia. 1998;13(2):69–81.

Yoshimatsu Y, Melgaard D, Westergren A, Skrubbeltrang C, Smithard DG. The diagnosis of aspiration pneumonia in older persons: a systematic review. Eur Geriatr Med. 2022;13(5):1071–80.

Marik PE. Aspiration pneumonitis and aspiration pneumonia. N Engl J Med. 2001;344(9):665–71.

Wu XS, Miles A, Braakhuis A. Nutritional intake and meal composition of patients consuming texture modified diets and thickened fluids: a systematic review and meta-analysis. Healthcare (Basel) 2020;8(4):1–24.

Palli C, Fandler S, Doppelhofer K, Niederkorn K, Enzinger C, Vetta C, et al. Early dysphagia screening by trained nurses reduces pneumonia rate in stroke patients: a clinical intervention study. Stroke. 2017;48(9):2583–5.

Serra-Prat M, Palomera M, Gomez C, Sar-Shalom D, Saiz A, Montoya JG, et al. Oropharyngeal dysphagia as a risk factor for malnutrition and lower respiratory tract infection in independently living older persons: a population-based prospective study. Age Ageing. 2012;41(3):376–81.

Dziewas R, Pflug C, et al. Neurogene dysphagie: S1-Leitlinie. AWMF; 2020. 30/111 [cited 2023 Nov 20]. Available from: www.dgn.org/leitlinien .

Haldemann-Jenni E, Fierz K, Frei IA. Wollen und nicht können. Malnutritionsmanagement in medizinischen Kliniken eines schweizerischen Zentrumsspitals: Erleben und Sichtweisen von Patient(inn)en. Pflege. 2016;29(3):115–23.

Mizuno S, Wakabayashi H, Wada F. Rehabilitation nutrition for individuals with frailty, disability, sarcopenic dysphagia, or sarcopenic respiratory disability. Curr Opin Clin Nutr Metab Care. 2022;25(1):29–36.

Malone A, Mogensen KM. Key approaches to diagnosing malnutrition in adults. Nutr Clin Pract. 2022;37(1):23–34.

Yamada Y, Shamoto H, Maeda K, Wakabayashi H. Home-based Combined Therapy with Rehabilitation and Aggressive Nutrition Management for a Parkinson’s disease patient with sarcopenic dysphagia: a case report. Prog Rehabil Med. 2018;3:20180019.

Andrade PA, Santos CAD, Firmino HH, de Oliveira Barbosa CR. The importance of dysphagia screening and nutritional assessment in hospitalized patients. Einstein (Sao Paulo). 2018;16(2):eAO4189.

British Geriatrics Society. Comprehensive geriatric assessment toolkit for primary care practitioners: British Geriatrics Society; 2019. Available from: https://www.bgs.org.uk/cgatoolkit .

Ward KT, Reuben DB. Comprehensive geriatric assessment: Wolters Kleuwer; Available from: https://www.uptodate.com/contents/comprehensive-geriatric-assessment .

Tricco AC, Lillie E, Zarin W, O’Brien KK, Colquhoun H, Levac D, et al. PRISMA extension for scoping reviews (PRISMA-ScR): checklist and explanation. Ann Intern Med. 2018;169(7):467–73.

von Elm E, Schreiber G, Haupt CC. Methodische Anleitung für Scoping Reviews (JBI-Methodologie). Z Evid Fortbild Qual Gesundhwes. 2019;143:1–7. [cited 2024 Feb 1]. Available from: https://www.zefq-journal.com/action/showPdf?pii=S1865-9217%2819%2930066-2 .

Article   Google Scholar  

Ritschl V, Weigl R, Stamm TA, editors. Wissenschaftliches Arbeiten und Schreiben: Verstehen, Anwenden, Nutzen für die Praxis. 2. Auflage. Berlin, Heidelberg: Springer; 2023. (Studium Pflege, Therapie, Gesundheit).

Levac D, Colquhoun H, O’Brien KK. Scoping studies: advancing the methodology. Implement Sci. 2010;5:69.

Covidence: systematic review software. Melbourne: Veritas Health Innovation; 2023. Available from: www.covidence.org .

Wakabayashi H, Matsushima M. Dysphagia assessed by the 10-item eating assessment tool is associated with nutritional status and activities of daily living in elderly individuals requiring long-term care. J Nutr Health Aging. 2016;20(1):22–7.

Andrade PA, Santos CAD, Firmino HH, de Oliveira-Barbosa RC. The importance of dysphagia screening and nutritional assessment in hospitalized patients. Einstein (Sao Paulo, Brazil). 2018;16(2):eAO4189.

Cabre M, Serra-Prat M, Palomera E, Almirall J, Pallares R, Clavé P. Prevalence and prognostic implications of dysphagia in elderly patients with pneumonia. Age Ageing. 2010;39(1):39–45.

Carrión S, Cabré M, Monteis R, Roca M, Palomera E, Serra-Prat M, et al. Oropharyngeal dysphagia is a prevalent risk factor for malnutrition in a cohort of older patients admitted with an acute disease to a general hospital. Clin Nutr (Edinburgh, Scotland). 2015;34(3):436–42.

Mateos-Nozal J, Montero-Errasquín B, García ES, Rodríguez ER, Cruz-Jentoft AJ. High prevalence of oropharyngeal dysphagia in acutely hospitalized patients aged 80 years and older. J Am Med Dir Assoc. 2020;21(12):2008–11.

Shimazu S, Yoshimura Y, Kudo M, Nagano F, Bise T, Shiraishi A, et al. Frequent and personalized nutritional support leads to improved nutritional status, activities of daily living, and dysphagia after stroke. Nutrition (Burbank, Los Angeles County, Calif). 2021;83(beu, 8802712):111091.

Wham C, Fraser E, Buhs-Catterall J, Watkin R, Gammon C, Allen J. Malnutrition risk of older people across district health board community, hospital and residential care settings in New Zealand. Australas J Ageing. 2017;36(3):205–11.

Alvarez-Larruy M, Tomsen N, Guanyabens N, Palomeras E, Clavé P, Nascimento W. Spontaneous swallowing frequency in post-stroke patients with and without oropharyngeal dysphagia: an observational study. Dysphagia. 2023;38(1):200–10.

Moncayo-Hernández BA, Herrera-Guerrero JA, Vinazco S, Ocampo-Chaparro JM, Reyes-Ortiz CA. Sarcopenic dysphagia in institutionalised older adults. Endocrinologia, diabetes y nutricion 2021.

Mañas-Martínez AB, Bucar-Barjud M, Campos-Fernández J, Gimeno-Orna JA, Pérez-Calvo J, Ocón-Bretón J. Association of positive screening for dysphagia with nutritional status and long-term mortality in hospitalized elderly patients. Endocrinologia, diabetes y nutricion. 2018;65(7):402–8.

Drozdz D, Mancopes R, Silva AMT, Reppold C. Analysis of the level of Dysphagia, anxiety, and nutritional status before and after speech therapy in patients with stroke. Int Arch Otorhinolaryngol. 2014;18(2):172–7.

Namasivayam-MacDonald AM, Morrison JM, Steele CM, Keller H. How swallow pressures and dysphagia affect malnutrition and mealtime outcomes in long-term care. Dysphagia. 2017;32(6):785–96.

Olesen MD, Modlinski RM, Poulsen SH, Rosenvinge PM, Rasmussen HH, Holst M. Prevalence of signs of dysphagia and associated risk factors in geriatric patients admitted to an acute medical unit. Clin Nutr ESPEN. 2021;41:208–16. Available from:  https://www.sciencedirect.com/science/article/pii/S2405457720311268 .

Omura T, Matsuyama M, Nishioka S, Sagawa S, Seto M, Naoe M. Association between the swallowing reflex and the incidence of aspiration pneumonia in patients with dysphagia admitted to long-term care wards: a prospective cohort study of 60 days. Arch Phys Med Rehabil. 2021;102(11):2165–71. Available from:  https://www.sciencedirect.com/science/article/pii/S0003999321005013 .

Ortega Barrio MÁ, Valiñas Sieiro F, Almarza Fernández MT, Bravo Santamaría S, Moreno Maestro R. Effect of stroke on nutritional status and its relationship with dysphagia. Revista Científica de la Sociedad de Enfermería Neurológica (English ed). 2020;51:13–21. Available from:  https://www.sciencedirect.com/science/article/pii/S2530299X19300159 .

Poisson P, Laffond T, Campos S, Dupuis V, Bourdel-Marchasson I. Relationships between oral health, dysphagia and undernutrition in hospitalised elderly patients. Gerodontology. 2016;33(2):161–8.

Popman A, Richter M, Allen J, Wham C. High nutrition risk is associated with higher risk of dysphagia in advanced age adults newly admitted to hospital. Nutr Dietet. 2018;75(1):52–8.

Tran TP, Nguyen LT, Hirose K, Nguyen TH, Le HT, Shimura F, et al. Malnutrition is associated with dysphagia in Vietnamese older adult inpatients. Asia Pacific journal of clinical nutrition. 2021;30(4):588–94.

Ramos-Vázquez AG, Reyes-Torres CA, Castillo-Martínez L, Serralde Zúñiga AE. Body composition by bioelectrical impedance, muscle strength, and nutritional risk in oropharyngeal dysphagia patients. Nutr Hosp. 2021;38(2):315–20.

Vidal CA, Suárez SC, Rodríguez GL, Gutiérrez FP, Almeida SMC, Ramos MJR, et al. Utility of the EAT-10 in the detection of dysphagia in high-risk hospitalisation units at a university hospital: a cross-sectional study. Nutricion hospitalaria. 2020;37(6):1197–200.

Tanıgör G, Eyigör S. Evaluation of dysphagia in patients with sarcopenia in a rehabilitation setting: insights from the vicious cycle. Eur Geriatr Med. 2020;11(2):333–40.

Wald NJ. The definition of screening. J Med Screen. 2001;8(1):1. Available from: https://journals.sagepub.com/doi/full/10.1136/jms.8.1.1 .

Rommel N, Hamdy S. Oropharyngeal dysphagia: manifestations and diagnosis. Nat Rev Gastroenterol Hepatol. 2016;13(1):49–59.

Clavé P, Arreola V, Romea M, Medina L, Palomera E, Serra-Prat M. Accuracy of the volume-viscosity swallow test for clinical screening of oropharyngeal dysphagia and aspiration. Clin Nutr. 2008;27(6):806–15.

Chen AY, Frankowski R, Bishop-Leone J, Hebert T, Leyk S, Lewin J, et al. The development and validation of a dysphagia-specific quality-of-life questionnaire for patients with head and neck cancer: the M. D. Anderson dysphagia inventory. Arch Otolaryngol Head Neck Surg. 2001;127(7):870–6.

CAS   PubMed   Google Scholar  

Belafsky PC, Mouadeb DA, Rees CJ, Pryor JC, Postma GN, Allen J, et al. Validity and reliability of the Eating Assessment Tool (EAT-10). Ann Otol Rhinol Laryngol. 2008;117(12):919–24.

Matsuo H, Yoshimura Y, Ishizaki N, Ueno T. Dysphagia is associated with functional decline during acute-care hospitalization of older patients. Geriatr Gerontol Int. 2017;17(10):1610–6.

Uno C, Maeda K, Wakabayashi H, Nishioka S, Ogawa N, Okamoto T, et al. Nutritional status change and activities of daily living in elderly pneumonia patients admitted to acute care hospital: a retrospective cohort study from the Japan Rehabilitation Nutrition Database. Nutrition. 2020;71:110613. Available from:  https://www.sciencedirect.com/science/article/pii/S0899900719301960 .

Waza M, Maeda K, Katsuragawa C, Sugita A, Tanaka R, Ohtsuka A, et al. Comprehensive tool to assess oral feeding support for functional recovery in post-acute rehabilitation. J Am Med Dir Assoc. 2019;20(4):426–31.

Yu J, Zhu H, Zhang Y, Wang D, Guo H, Liu X, et al. The relationship between dysphagia and frailty among Chinese hospitalized older patients: a serial mediation model through self-perceived oral health and self-reported nutritional status. BMC Geriatr. 2024;24(1):110.

Guigoz Y, Lauque S, Vellas BJ. Identifying the elderly at risk for malnutrition. The mini nutritional assessment. Clin Geriatr Med. 2002;18(4):737–57.

Bouillanne O, Morineau G, Dupont C, Coulombel I, Vincent J-P, Nicolis I, et al. Geriatric nutritional risk index: a new index for evaluating at-risk elderly medical patients. Am J Clin Nutr. 2005;82(4):777–83.

Duerksen DR, Laporte M, Jeejeebhoy K. Evaluation of nutrition status using the subjective global assessment: malnutrition, cachexia, and sarcopenia. Nutr Clin Pract. 2021;36(5):942–56.

BAPEN. The ‘MUST’ Explanatory Booklet: A Guide to the ‘Malnutrition Universal Screening Tool’ (‘MUST’) for Adults; 2011. Available from: http://www.bapen.org.uk/pdfs/must/must_explan.pdf .

Deutsche Gesellschaft für Geriatrie. S1-Leitlinie Geriatrisches Assessment der Stufe 2: Living Guideline; AWMF-Register-Nr. 084–002LG. AWMF; 2022 Oct 15 3 [cited 2023 Nov 10]. Available from: https://register.awmf.org/de/leitlinien/detail/084-002LG .

Perry L. Screening swallowing function of patients with acute stroke. Part one: Identification, implementation and initial evaluation of a screening tool for use by nurses. J Clin Nurs. 2001;10(4):463–73.

Daniels SK, McAdam CP, Brailey K, Foundas AL. Clinical assessment of swallowing and prediction of dysphagia severity. Am J Speech Lang Pathol. 1997;6(4):17–24.

Thiem U, Jäger M, Stege H, Wirth R. Diagnostic accuracy of the “Dysphagia Screening Tool for Geriatric Patients” (DSTG) compared to Flexible Endoscopic Evaluation of Swallowing (FEES) for assessing dysphagia in hospitalized geriatric patients - a diagnostic study. BMC Geriatr. 2023;23(1):856.

Ferguson M, Capra S, Bauer J, Banks M. Development of a valid and reliable malnutrition screening tool for adult acute hospital patients. Nutrition. 1999;15(6):458–64.

Zhang P-P, Yuan Y, Lu D-Z, Li T-T, Zhang H, Wang H-Y, et al. Diagnostic accuracy of the Eating Assessment Tool-10 (EAT-10) in screening dysphagia: a systematic review and meta-analysis. Dysphagia. 2023;38(1):145–58.

Wirth R, Dziewas R, Beck AM, Clavé P, Hamdy S, Heppner HJ, et al. Oropharyngeal dysphagia in older persons - from pathophysiology to adequate intervention: a review and summary of an international expert meeting. Clin Interv Aging. 2016;11:189–208.

Tsang K, Lau ES, Shazra M, Eyres R, Hansjee D, Smithard DG. A new simple screening tool-4QT: can it identify those with swallowing problems? A pilot study. Geriatrics (Basel) 2020;5(1):1–9.

Westergren A, Smithard D, Westergaard M, Norup A, Riis J, Krarup A, et al. Convergent and discriminant validity of the Minimal Eating Observation Form - version II: a cross-sectional study. BMC Geriatr. 2024;24(1):27.

Wilson M-MG, Thomas DR, Rubenstein LZ, Chibnall JT, Anderson S, Baxi A, et al. Appetite assessment: simple appetite questionnaire predicts weight loss in community-dwelling adults and nursing home residents. Am J Clin Nutr. 2005;82(5):1074–81.

Leroi I, Kitagawa K, Vatter S, Sugihara T. Dementia in “super-aged” Japan: challenges and solutions. Neurodegener Dis Manag. 2018;8(4):257–66.

Minagawa Y, Saito Y. Changes in health expectancy for older adults in Japan: results from two longitudinal surveys. Gerontology. 2022;68(10):1166–73.

Cichero JAY, Lam P, Steele CM, Hanson B, Chen J, Dantas RO, et al. Development of international terminology and definitions for texture-modified foods and thickened fluids used in dysphagia management: the IDDSI framework. Dysphagia. 2017;32(2):293–314.

Download references

Acknowledgements

Not applicable.

The current study did not receive funding as it is part of SMJs dissertation.

Author information

Authors and affiliations.

Institute of Nursing Science and Research, Paracelsus Medical University, Strubergasse 21, 5020, Salzburg, Austria

Susanne M. Javorszky

FH Joanneum, Institute of Health and Nursing, Alte Post Straße 149, 8020, Graz, Austria

Christoph Palli

FH Campus Wien, Department of Health Sciences, Favoritenstraße, 226, 1100, Vienna, Austria

Susanne M. Javorszky & Susanne Domkar

Department of Geriatric Medicine, Christian-Doppler-Klinik, Paracelsus Medical University, Ignaz-Harrer-Straße 79, 5020, Salzburg, Austria

Bernhard Iglseder

You can also search for this author in PubMed   Google Scholar

Contributions

S.M.J. planned the study. Data was collected and analyzed by S.M.J., C.P. and S.D. B.I. was a major contributor in writing the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Susanne M. Javorszky .

Ethics declarations

Ethics approval and consent to participate, consent for publication, competing interests.

The authors declare no competing interests.

Additional information

Publisher’s note.

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

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

Cite this article.

Javorszky, S.M., Palli, C., Domkar, S. et al. Combined systematic screening for malnutrition and dysphagia in hospitalized older adults: a scoping review. BMC Geriatr 24 , 445 (2024). https://doi.org/10.1186/s12877-024-05070-6

Download citation

Received : 12 March 2024

Accepted : 13 May 2024

Published : 21 May 2024

DOI : https://doi.org/10.1186/s12877-024-05070-6

Share this article

Anyone you share the following link with will be able to read this content:

Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

  • Presbyphagia
  • Malnutrition

BMC Geriatrics

ISSN: 1471-2318

the purpose of a systematic review

COMMENTS

  1. Introduction to systematic review and meta-analysis

    A systematic review collects all possible studies related to a given topic and design, and reviews and analyzes their results [ 1 ]. During the systematic review process, the quality of studies is evaluated, and a statistical meta-analysis of the study results is conducted on the basis of their quality. A meta-analysis is a valid, objective ...

  2. Systematic Review

    A systematic review is a type of review that uses repeatable methods to find, select, and synthesize all available evidence. It answers a clearly formulated research question and explicitly states the methods used to arrive at the answer. Example: Systematic review. In 2008, Dr. Robert Boyle and his colleagues published a systematic review in ...

  3. Why systematic reviews matter

    A brief history of systematic reviews. The first example of a systematic review was conducted in 1753 by James Lind, who published a paper that aimed to provide a concise and unbiased summary of evidence on scurvy 8 9. However, it was not until the 1970's and 1980's when more attention was paid to the growing need to improve the state of ...

  4. Systematic review

    A systematic review is a scholarly synthesis of the evidence on a clearly presented topic using critical methods to identify, define and assess research on the topic. A systematic review extracts and interprets data from published studies on the topic (in the scientific literature), then analyzes, describes, critically appraises and summarizes interpretations into a refined evidence-based ...

  5. What is a systematic review?

    A high-quality systematic review is described as the most reliable source of evidence to guide clinical practice. The purpose of a systematic review is to deliver a meticulous summary of all the available primary research in response to a research question. A systematic review uses all the existing research and is sometime called 'secondary research' (research on research).

  6. How to do a systematic review

    A systematic review aims to bring evidence together to answer a pre-defined research question. This involves the identification of all primary research relevant to the defined review question, the critical appraisal of this research, and the synthesis of the findings.13 Systematic reviews may combine data from different.

  7. Introduction to Systematic Reviews

    A systematic review identifies and synthesizes all relevant studies that fit prespecified criteria to answer a research question. Systematic review methods can be used to answer many types of research questions. ... The purpose of the qualitative synthesis is "to develop and convey a deeper understanding of the diversity of questions ...

  8. What are systematic reviews?

    What are systematic reviews? Watch on. Cochrane evidence, including our systematic reviews, provides a powerful tool to enhance your healthcare knowledge and decision making. This video from Cochrane Sweden explains a bit about how we create health evidence and what Cochrane does. About Cochrane.

  9. What are systematic reviews?

    A systematic review requires a considerable amount of time and resources, and is one type of literature review. If the purpose of a review is to make justifiable evidence claims, then it should be systematic, as a systematic review uses rigorous explicit methods. The methods used can depend on the purpose of the review, and the time and ...

  10. Systematic reviews: Structure, form and content

    A systematic review collects secondary data, and is a synthesis of all available, relevant evidence which brings together all existing primary studies for review (Cochrane 2016). A systematic review differs from other types of literature review in several major ways. It requires a transparent, reproducible methodology which indicates how ...

  11. An introduction to overviews of reviews: planning ...

    To summarise evidence from more than one systematic review of the same intervention for the same condition or problem where different outcomes are addressed in different systematic reviews ... et al. Toward a comprehensive evidence map of overview of systematic review methods: paper 1—purpose, eligibility, search and data extraction ...

  12. Guidance on Conducting a Systematic Literature Review

    Ideally, a systematic review should be conducted before empirical research, and a subset of the literature from the systematic review that is closely related to the empirical work can be used as background review. ... Based on the purpose of the review (column 1 in Table 1) and the type of literature (column 2), researchers can narrow down to ...

  13. About Cochrane Reviews

    A systematic review attempts to identify, appraise and synthesize all the empirical evidence that meets pre-specified eligibility criteria to answer a specific research question. Researchers conducting systematic reviews use explicit, systematic methods that are selected with a view aimed at minimizing bias, to produce more reliable findings to ...

  14. Easy guide to conducting a systematic review

    A systematic review is a type of study that synthesises research that has been conducted on a particular topic. Systematic reviews are considered to provide the highest level of evidence on the hierarchy of evidence pyramid. Systematic reviews are conducted following rigorous research methodology. To minimise bias, systematic reviews utilise a ...

  15. Systematic reviews: Brief overview of methods, limitations, and

    CONCLUSION. Siddaway 16 noted that, "The best reviews synthesize studies to draw broad theoretical conclusions about what the literature means, linking theory to evidence and evidence to theory" (p. 747). To that end, high quality systematic reviews are explicit, rigorous, and reproducible. It is these three criteria that should guide authors seeking to write a systematic review or editors ...

  16. The PRISMA 2020 statement: an updated guideline for reporting ...

    The Preferred Reporting Items for Systematic reviews and Meta-Analyses (PRISMA) statement, published in 2009, was designed to help systematic reviewers transparently report why the review was done, what the authors did, and what they found. Over the past decade, advances in systematic review methodology and terminology have necessitated an update to the guideline. The PRISMA 2020 statement ...

  17. Systematic reviews and meta‐analyses; and real‐world evidence

    This chapter discusses systematic reviews of randomised controlled trials (RCTs), including stages of a systematic review, meta-analysis, and considerations when evaluating a systematic review. RCTs are the most reliable way to assess the efficacy and safety of a new intervention. Real-world data (RWD) and real-world evidence (RWE) are meant to ...

  18. What is a systematic review?

    A systematic review is an academic research paper, also called a report, that uses a method called 'evidence synthesis' to look for answers to a pre-defined question. The purpose of a systematic review is to sum up the best available research on that specific question. This is done by synthesizing the results of several studies. A systematic review uses transparent procedures to find, evaluate ...

  19. LibGuides: Basics of Systematic Reviews: Types of Reviews

    Systematic Review. Attempts to identify, appraise, and summarize all empirical evidence that fits pre-specified eligibility criteria to answer a specific research question. clearly defined question with inclusion/exclusion criteria. rigorous and systematic search of the literature. thorough screening of results. data extraction and management.

  20. Protocol for a scoping review study on learning plan use in

    The purpose of this study is to summarise the literature regarding learning plan use specifically in undergraduate medical education and explore the student's role in all stages of learning plan development and implementation. ... (UGME). A related systematic review by van Houten-Schat et al. on SRL in the clinical setting identified three ...

  21. Combined systematic screening for malnutrition and dysphagia in

    The purpose of this review was to determine whether institutions have established a protocol combining screenings for dysphagia and malnutrition on a global scale. If combined screening protocols have been implemented, the respective derived measures will be reported. ... A scoping review was conducted. A systematic database search was carried ...

  22. Enhancing Foreign Language Learning Approaches to ...

    The main purpose of this study is to investigate the best approaches to teaching a foreign language to older people to help them achieve the desired results and explore their benefits. This review strictly follows the PRISMA methodology for systematic reviews and meta-analyses to identify the core experimental studies that deal with the topic of foreign language learning approaches among the ...

  23. ERIC

    The purpose of this systematic review and meta-analysis is twofold: (a) to understand how the three key student engagement dimensions (i.e., affective, behavioral, and cognitive) have been conceptualized, operationalized, and measured by researchers in the field and (b) to examine the extent to which the construct, its dimensions, and subtypes are associated with academic achievement and ...

  24. Return to Work After Anterior Cruciate Ligament Reconstruction: A

    In our systematic review, no studies reported RTW timing for ACLRs performed with BTB and all-soft tissue QT autografts. In contrast, the times to RTW for bone-block QT autograft (45.8 ± 42 days) and the LARS device (48.2 ± 20.3 days) were reported by Horstmann et al 11 and Kentel et al, 14 respectively. The duration of RTW in patients using ...