Category: 3. Business

  • Prompt versus delayed triple therapy in COPD: Solutions to time-relate

    Prompt versus delayed triple therapy in COPD: Solutions to time-relate

    Introduction

    Maintenance therapy is recommended for patients diagnosed with chronic obstructive pulmonary disease (COPD). These treatments include long-acting muscarinic antagonists (LAMAs) and long-acting beta2-agonists (LABAs), with an inhaled corticosteroid (ICS) added according to the frequency of exacerbations.1 Currently, several single-inhaler triple combinations of these treatment classes are available.

    Besides the general recommendations for which inhaler combination to use, the question of when to initiate maintenance therapy with these inhalers has been put forward. Recent observational studies have investigated the comparative effectiveness of prompt versus delayed timing of initiating single-inhaler triple inhaler therapy after a COPD exacerbation.2–6 These studies found that prompt initiation of single-inhaler triple therapy was associated with significant reductions in the rates of moderate and severe exacerbations, compared with delayed initiation. Such observational studies present major methodological challenges related to the definition of timing of initiation in relation to the timing of the outcome events, that can result in time-related biases.

    We review these studies and discuss methodological aspects of their study design that can introduce bias in the results. We illustrate the biases using a general practice clinical database and present results of the analysis using an approach that avoids these biases.

    The Published Studies

    As the published observational studies used a similar design, we describe the first one in detail to explain the approach.2 The Mannino study evaluated the impact of prompt versus delayed initiation of single-inhaler triple therapy (SITT) with fluticasone furoate, umeclidinium, and vilanterol, following a COPD exacerbation, using a US claims database. Patients with a COPD exacerbation between September 2017 and September 2019 were identified, with the first exacerbation occurring in that period taken as the index exacerbation. The index date was taken as the date of discharge for exacerbations requiring hospitalisation and the date of the physician visit for moderate exacerbations. The study cohort was formed exclusively from those who initiated a SITT within 6 months after the index date, with SITT timing classified as prompt (initiation within 30 days after the index date; N = 529) or delayed (initiation 31–180 days after the index date; N = 1,375). Patients were aged 40 years or more at the index date, had at least 12 months of continuous health insurance coverage before index (baseline), no exacerbation and no SITT prescription during this baseline period. The subjects needed at least 6 months of coverage after the index. Subjects were followed from the index date until the end of the observation period for the occurrence of COPD exacerbations and other outcomes. The technique of inverse probability of treatment weighting was used to adjust the rate ratio of these outcomes for differences in baseline characteristics between the prompt and delayed groups. Patients in the prompt initiation group had a 21% lower rate of COPD exacerbation (rate ratio 0.79; 95% CI: 0.65–0.94) and a 28% lower incidence of a first exacerbation (hazard ratio 0.72; 95% CI: 0.62–0.83) compared with delayed initiators.

    Methodological Issues

    A randomized trial of this question would enroll patients at the index exacerbation and randomly allocate them to either the prompt or delayed treatment strategy. The allocation of the two groups is thus known at the time of randomization (index date) with outcome events counted as of this time, and which can thus occur prior to treatment initiation. The published observational studies, on the other hand, had to peek into “future” to define the two treatment groups, such as treatment initiation within 30 days after the index (prompt) or at 31–180 days after the index (delayed). This use of “future” time creates several methodological challenges and can lead to potential biases in observational studies. We use the Mannino study described in detail above as an example to explain the methodological issues.2

    The first methodological issue with the observational studies involves the outcome (exacerbations) allowed to occur prior to the initiation of triple therapy, which can introduce protopathic bias.7 Indeed, some physicians may wait for a second exacerbation, namely the first during follow-up, as an indication to initiate triple therapy, as per guidelines.1 Thus, it may not be the treatment that led to the exacerbation, but the reverse. In particular, patients with early exacerbations will likely receive their triple inhaler after day 30, when the exacerbation ended, and thus more likely to be classified in the “delayed” treatment group, a bias compounded by the longer duration of this delayed period. This could explain the observational study’s reported median times to the first COPD exacerbation of 367 versus 200 days for the prompt and delayed initiation groups, respectively.2 Moreover, the corresponding Kaplan–Meier curve shows that around 10% of the delayed group had their first exacerbation in the first 30 days, the period defining “prompt” treatment, and that 48% of that group had their first exacerbation before 6 months, the end of the “delayed” treatment period.

    The second methodological issue relates to the cohort selection that imposes a period of continuous health insurance coverage after the index date, resulting in potential selection bias from immortal time.8,9 Indeed, the cohort included only patients with at least 6 months of coverage after the index date, thus excluding patients who die in this 6-month period, even if they had initiated triple therapy and had exacerbations before death. By this criterion, patients must survive to initiate treatment, whether prompt or delayed, even if they had exacerbations before death. This imposition of 6 months of coverage after an index that inherently excludes deaths can result in selection bias that could favor one group over the other. The Mannino study reports that 25% of the original 668,011 subjects were excluded because they had less than 6 months of eligibility, with no information on mortality.2

    Study Design to Avoid Bias

    The study design that avoids these biases must attempt to emulate the randomized trial when using these observational data, while not looking into the future. The key with the randomized trial is that the timing of inhaler initiation, namely allocation to a prompt or delayed treatment regimen, is known at the time of randomisation, so that the two groups can be properly compared on the rate of exacerbation during a prespecified follow-up period after the index exacerbation. In this case, exacerbation events can precede the inhaler initiation but will follow the random treatment group assignment time. The difference in the rates between the two groups will provide the effect of prompt versus delayed treatment.

    The observational study must thus also seek to allocate “exposure” (prompt or delayed inhaler initiation) at the index date to avoid the methodological biases raised above when this exposure allocation is based on looking in the future. To accomplish this, it is important to recognize that a subject who has not yet received the inhaler at the index date could, in fact, belong to both the prompt and delayed group at that time point and up until inhaler therapy is initiated. To resolve this dilemma, the concept of “cloning” is used in observational research to assign each patient to the two possible treatment strategies (prompt and delayed inhaler initiation) that the patient could belong to at the index date.10–12 Thus, the patient is “cloned” so that their data are included twice in the analysis, though the follow-up will be censored according to the timing of the inhaler initiation and the outcome events, which could be counted in one, two, or none of the groups depending on where the clones are censored.

    As an illustration for the outcome of time to first exacerbation, consider, for example, a patient who initiates triple therapy on day 15, thus within 30 days (Figure 1, subject 1). This patient will generate two clones, a “prompt” clone that will be considered as exposed to the prompt treatment strategy for the entire follow-up, whose follow-up will end at the time of the event, and a “delayed” clone that will be considered as exposed to the delayed treatment strategy and censored at 14 days, the day at which we still did not know the membership of its parent. The second example (subject 2) is a patient who initiates triple therapy on day 60 (during the 31–180-day period). This patient will generate two clones, the first is a “prompt” clone that will be censored at 30 days, the time they can no longer be prompt, and a “delayed” clone that will be classified as “delayed” exposure for the entire follow-up, whose follow-up will end at the time of the event (Figure 1, subject 2). The third example (subject 3) does not initiate triple therapy at all during follow-up. The “prompt” clone will be censored at 30 days and the delayed clone follow-up will end at the time of the event within the 31–180 period (Figure 1, subject 3). The fourth example (subject 4) is a patient who initiates triple therapy on day 35 (during the 31–180-day period), who has their first exacerbation on day 20. The two generated clones, one prompt and one delayed, will both have an outcome event at day 20, at which point their follow-up stops (Figure 1, subject 4). Figure 2 displays the corresponding cloning patterns illustrating the situation where the outcome involves the frequency of exacerbations over time.

    Figure 1 Illustration of cloning approach for the analysis of time to first exacerbation.

    Figure 2 Illustration of cloning approach for the analysis of the frequency of exacerbations.

    This cloning approach addresses the bias resulting from peeking into the future to define exposure. Nonetheless, simply computing the corresponding cumulative incidences or rates of exacerbation for the two cloned treatment strategies will still produce bias from giving equal weights to the clones. This is addressed by accounting for the artificial censoring of the clones at specific times, which can be done using inverse probability of censoring weights (ICPW).11

    Illustration

    We formed a cohort of patients with COPD from the Clinical Practice Research Datalink (CPRD), a primary care database from the United Kingdom (UK) that contains primary care medical records for over 50 million people enrolled in more than 1800 general practices. These data have shown to be of high quality, including for studies of COPD.13–17

    The study cohort included all patients with a diagnosis of COPD, treated with maintenance therapy, at or after age 40 who had a moderate or severe exacerbation of COPD after 15 September 2017, the year single-inhaler triple therapy became available in the UK. A moderate exacerbation was defined by a new prescription for prednisolone, while a severe exacerbation was defined as a hospitalization for COPD (ICD-10: J41, J42, J43, J44). The first such exacerbation defined the index date. All subjects had to have at least one year of medical history prior to the index date (baseline period). Patients receiving triple therapy, either in a single inhaler or multiple inhalers, in the year before the exacerbation defining cohort entry, were excluded. All subjects were followed for up to one year after the index date, with follow-up ending at death, 31 March 2021, or the end of the patient’s registration in the practice, whichever occurred first.

    The covariates measured at baseline included age, sex, body mass index (BMI), smoking status and alcohol abuse. The severity of COPD was measured using the type of index exacerbation (moderate, severe), the number of COPD hospitalisations and the use of other respiratory drugs (SABA, SAMA, theophylline), during the one-year baseline period, as well as by the percent predicted FEV1. A prescription for prednisolone, LABA, LAMA, ICS, and respiratory antibiotics in the month prior to the index date were also considered. Baseline co-morbidity in the one-year baseline period was measured using clinical diagnoses, hospitalizations, and prescriptions (Table 1).

    Table 1 Baseline Characteristics of the Overall Study Cohort of 91,958 Subjects with an Index Exacerbation, Generating 91,958 Clones of Prompt and 91,958 Clones of Delayed Initiation of Single-Inhaler Triple Therapy, After Weighing by Inverse Probability of Censoring

    Each cohort subject was cloned and assigned to both the prompt initiators arm (initiation within 30 days of the index date) and to the delayed initiators arm (initiation 31–180 days after the index date). Within each treatment group, clones were artificially censored at the time that the treatment they received was no longer compatible with their group membership. To account for the bias introduced by this artificial censoring mechanism, IPCW was estimated by pooled logistic regression, separately for prompt initiators at day 30, for delayed initiators at index date, between day 1 and day 30 and at day 180, as a function of the baseline covariates. IPCW estimation for prompt initiators, delayed initiators at index date and at day 180, was based on time updated values of the baseline covariates and the type of initial exacerbation (moderate or severe). For the delayed initiators, estimation of IPCW between day 1 and day 30 also included the time since cohort entry (linear, quadratic and cubic terms). Only prompt initiators starting treatment between day 16 and 30 were upweighted to replace clones censored on day 30. Similarly, only delayed initiators between day 152 and 180 were upweighted to replace clones censored on day 180. In addition to the artificial censoring associated with the cloning process, patients were also censored when they initiated triple therapy in multiple inhalers (same day prescription for LABA, LAMA and ICS). To account for this type of censoring another IPCW was estimated in the full cohort and before cloning, using pooled logistic regression and with the same set of variables. Final time-varying weights for each section of person-time were obtained from the cumulative product of all IPCWs.

    For data analysis, we used the Cox proportional hazards model to compare the incidence of an exacerbation during the one-year follow-up, weighted for the inverse of the probability of censoring. The corresponding 95% confidence intervals (CI) for the hazard ratios were obtained using the non-parametric bootstrap method based on 1000 random samples. Similarly, weighted cumulative incidence curves were estimated over the one-year follow-up, as well as differences and ratios of cumulative incidence at 3-month time points during follow-up.

    We also replicated the approach used in the Mannino study, as described above.2 Briefly, the study cohort included exclusively the subjects who had a prompt (within 30 days after the index date) or delayed (31–180 days after the index date classification) treatment initiation, restricted to those who had no exacerbation prior to the index date and at least 6 months of coverage after the index date. Inverse probability of treatment weighting, with stabilised weights, was used to adjust the hazard ratio of exacerbation, using the bootstrap method based on 1000 random samples outcomes to estimate the corresponding confidence interval. Since our study cohort on which the cloning analysis was based included subjects with exacerbations prior to the index date, we repeated the analysis, but not restricted to those who had no exacerbation prior to the index date and at least 6 months of coverage after the index date. The study protocol was approved by CPRD’s Research Data Governance Committee (protocol # 23_002846) and the Research Ethics Board of the Jewish General Hospital (protocol # JGH-2024-3847), Montreal Canada.

    Results

    The overall study cohort included 91,958 eligible subjects who had an exacerbation after September 2017. There were 4,876 new-users of single-inhaler triple therapy within 180 days after the index COPD exacerbation. Of these, 1,394 were prompt initiators and 3,482 delayed initiators of single-inhaler triple therapy, with 87,082 who either received it after 180 days or not at all. The prompt initiators appear more severe, with lower FEV1 percent predicted and more likely to have a hospitalised exacerbation as the index event (Table S1).

    The cloning of these subjects generated 91,958 prompt initiator clones and the same number of delayed initiator clones, including 426 clones who initiated triple therapy in multiple inhalers on the index date and 419 clones who initiated single-inhaler triple therapy on the index date (Table 1).

    Of the 91,532 clones assigned to the prompt initiation strategy, 18,748 had a moderate or severe exacerbation after the index date, during 86,431 person-months of follow-up, resulting in an unadjusted incidence rate of a first exacerbation of 0.217 per patient per month (Table 2). The corresponding incidence rate for clones assigned to the delayed initiation strategy was 0.130 per patient per month. After weighing for censoring, the incidence rates are 0.101 and 0.103 per person per month, respectively, for the clones assigned to the prompt and delayed initiation regimens (HR 0.98; 95% CI: 0.80–1.19). The adjusted cumulative incidence curves of a moderate or severe exacerbation, weighted for censoring, are displayed in Figure 3. For severe exacerbations, the weighted incidence rates are 0.011 and 0.008 per person per month for the clones assigned to the prompt and delayed initiation regimens, respectively (HR 1.26; 95% CI: 0.81–1.96), with the corresponding adjusted cumulative incidence curves displayed in Figure 4. For the cumulative incidence curves given in Figures 3 and 4 the estimates of the differences and ratios of these cumulative incidences between the prompt and delayed initiators at different time points in follow-up are displayed in Table 3.

    Table 2 Hazard Ratio of a Moderate or Severe Exacerbation for Prompt versus Delayed Initiation of Single-Inhaler Triple Therapy, Estimated Using Cloning to Define the Treatment Strategy Over Time, Weighed by Inverse Probability of Censoring

    Table 3 Difference and Ratio of the Cumulative Incidence of Exacerbation Over Follow-up Time Comparing Prompt versus Delayed Initiation of Single-Inhaler Triple Therapy, Estimated Using Cloning to Define the Treatment Strategy Over Time, Weighted by Inverse Probability of Censoring

    Figure 3 Cumulative incidence of the first moderate or severe exacerbation, for the prompt and delayed initiators, using the cloning approach, weighted by inverse probability of censoring.

    Figure 4 Cumulative incidence of the first severe exacerbation, for the prompt and delayed initiators, using the cloning approach, weighted by inverse probability of censoring.

    For the replication of the Mannino study, the analysis was restricted to the 2,650 new-users of single-inhaler triple therapy within 6 months after the index COPD exacerbation, with no prior exacerbations and at least 6 months of follow-up. Of these, 809 were prompt initiators of single-inhaler triple therapy and 1,841 were delayed initiators, who were similar in terms of baseline clinical characteristics after weighing (Table 4). The hazard ratio of a first moderate or severe exacerbation was 0.73 (95% CI: 0.65–0.81), comparing prompt with delayed initiation, while it was 0.58 (95% CI: 0.46–0.74) for a severe exacerbation (Table 5). These results were similar after removing the restrictions of 6 months of coverage and no prior exacerbations (Table 5).

    Table 4 Baseline Characteristics of the 809 Prompt and 1,841 Delayed Initiators of Single-Inhaler Triple Therapy, Identified from the Cohort of 91,958 Subjects with an Index Exacerbation, Crude and Weighted by Inverse Probability of Treatment, Used to Replicate the Approach of Mannino

    Table 5 Hazard Ratio of Moderate or Severe Exacerbation for Prompt versus Delayed Initiation of Single-Inhaler Triple Therapy, Used to Replicate the Approach of Mannino, Estimated Using the Cox Proportional Hazards Model, Adjusted by Inverse Probability of Treatment Weighing

    Discussion

    In this large-scale real-world study, we found that prompt treatment with single-inhaler triple therapy after a COPD exacerbation was not more effective than delayed treatment on reducing the incidence of a subsequent exacerbation. We showed that the methods used by previous studies that suggested significant effectiveness with prompt therapy, were affected by major time-related biases that favored the prompt treatment group.2,3,6 For example, our illustration showed that, using the corrected approach, the hazard ratio of a COPD exacerbation was 0.98 (95% CI: 0.80–1.19) with prompt versus delayed treatment, while the corresponding HR with the time-related biased method employed by the previous studies was a significant 0.73 (95% CI: 0.65–0.81).

    The time-related biases affecting the previous studies first involved “peeking into the future” to define prompt and delayed treatment, with a return to time zero to start follow-up for outcome exacerbation events. This approach thus allowed treatment initiation occurring after outcome events, which introduces protopathic bias.7 Indeed, multiple exacerbations, the study outcome, are an indication to initiate triple therapy, as recommended by the GOLD guidelines.1 The other source of bias, namely selection bias from immortal time, resulting from imposing 6 months of coverage after the index date, was present in two of the studies to date.2,6 This criterion excludes subjects who died during this period, when they could have initiated triple therapy and had exacerbations, which could be differential in the two treatment groups. While other studies did not impose this 6-month condition, they did introduce protopathic bias.3–5 Our bias analysis showed that this 6-month imposition did not affect the findings, implying that the major time-related bias in these studies is protopathic bias, a bias present in all studies.

    This approach has also been used in several other observational studies have investigated the comparative effectiveness of prompt versus delayed initiation of triple therapy, though including triple therapy in multiple inhalers after a COPD exacerbation.18–22 These studies also found that prompt initiation of triple therapy was associated with significant reductions in the rates of exacerbations, and related costs, compared with delayed initiation. These findings are thus also affected by the same protopathic bias.

    The “cloning” approach that we used is specifically designed to avoid the protopathic bias resulting from peeking into the future to define the treatment strategy. This cloning approach emulates a randomized trial by allocating the treatment strategy, prompt or delayed initiation of triple therapy, as of the index date, thus not looking in the future. Cloning involves creating data replicates of each patient, one for each of the study treatment strategies (in this case, prompt and delayed initiation) that the patient could belong to at the index date.10–12 The patient’s data are thus included twice in the analysis, censored by the timing of exposure and outcome events. While the same outcome events can be counted in multiple regimens in this approach, a censor-weighted data analysis and the bootstrap can account for the replicated data.

    Our study has some limitations typical to observational studies. The inhaler information is based on written prescriptions and can thus introduce some exposure misclassification, including on the timing of the actual treatment initiation, which can lag behind the prescription date. Thus, the 30- and 180-day thresholds used to define prompt and delayed treatment will necessarily be affected by this misclassification, which should differentially affect the shorter prompt treatment period. Indeed, we can assume that some subjects with a prescription date just prior to 30 days will be misclassified as prompt initiators if they in fact initiate their inhaler after 30 days. The use of censoring weights, however, because they are calculated using the observed exposure timing, does not account for this misclassification. To account for such exposure measurement error would require validating exposure in a subset of the study population and incorporating sensitivity analyses alongside censoring weights. Also, the outcome of a moderate exacerbation is defined only based on a prescription for prednisolone which, while a common practice in the UK, could introduce some misclassification. Our study also has strengths, besides the cloning approach that avoids the time-related biases of previous studies. Indeed, this approach is not affected by confounding as the cloning results in the same patients being replicated, making the two comparison groups identical on all subject characteristics.

    In conclusion, this large-scale real-world study found that prompt treatment with single-inhaler triple therapy after a COPD exacerbation was not more effective than delayed treatment on reducing the incidence of a subsequent exacerbation. We showed that the methods used by previous studies that suggested significant effectiveness with prompt therapy, were affected by time-related biases that favored the prompt treatment group. For example, using the corrected approach, we found no reduction in the risk of a COPD exacerbation with prompt versus delayed treatment, while the time-related biased method used in previous studies suggested a significant 27% reduction in the outcome event.

    Data Sharing Statement

    This study is based in part on data from the Clinical Practice Research Datalink obtained under license from the UK Medicines and Healthcare products Regulatory Agency. The data are provided by patients and collected by the UK National Health Service as part of their care and support. Because electronic health records are classified as “sensitive data” by the UK Data Protection Act, information governance restrictions (to protect patient confidentiality) prevent data sharing via public deposition. Data are available with approval through the individual constituent entities controlling access to the data. Specifically, the primary care data can be requested via application to the Clinical Practice Research Datalink (https://www.cprd.com).

    Author Contributions

    All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

    Funding

    This research was funded in part by grants from the Canadian Institutes of Health Research (CIHR) and the Canadian Foundation for Innovation (CFI). Pr. Suissa is the recipient of the Distinguished James McGill Professorship award.

    Disclosure

    SS attended, in the last three years, scientific advisory committee meetings or received speaking fees from AstraZeneca, Boehringer-Ingelheim, Novartis, and Panalgo. The authors report no other conflicts of interest in this work.

    References

    1. Agustí A, Celli BR, Criner GJ, et al. Global initiative for chronic obstructive lung disease 2023 report: GOLD executive summary. Eur Respir J. 2023;61:2300239. doi:10.1183/13993003.00239-2023

    2. Mannino D, Bogart M, Germain G, et al. Benefit of prompt versus delayed use of single-inhaler Fluticasone Furoate/Umeclidinium/Vilanterol (FF/UMEC/VI) following a COPD exacerbation. Int J Chron Obstruct Pulmon Dis. 2022;17:491–504. doi:10.2147/COPD.S337668

    3. Ismaila AS, Rothnie KJ, Wood RP, et al. Benefit of prompt initiation of single-inhaler fluticasone furoate, umeclidinium, and vilanterol (FF/UMEC/VI) in patients with COPD in England following an exacerbation: a retrospective cohort study. Respir Res. 2023;24(1):229. doi:10.1186/s12931-023-02523-1

    4. Strange C, Tkacz J, Schinkel J, et al. Exacerbations and real-world outcomes after single-inhaler triple therapy of Budesonide/Glycopyrrolate/Formoterol Fumarate, among patients with COPD: results from the EROS (US) Study. Int J Chron Obstruct Pulmon Dis. 2023;18:2245–2256. doi:10.2147/COPD.S432963

    5. Czira A, Akiyama S, Ishii T, et al. Benefit of prompt vs delayed initiation of triple therapy following an exacerbation in patients with COPD in Japan: a retrospective cohort study. Int J Chron Obstruct Pulmon Dis. 2023;18:2933–2953. doi:10.2147/COPD.S419119

    6. Mannino D, DiRocco K, Germain G, et al. Fluticasone Furoate/Umeclidinium/Vilanterol initiation following a COPD exacerbation: benefits of prompt initiation on COPD outcomes. Adv Ther. 2024;41(12):4557–4580. doi:10.1007/s12325-024-02999-3

    7. Horwitz RI, Feinstein AR. The problem of “protopathic bias” in case-control studies. Am J Med. 1980;68(2):255–258. doi:10.1016/0002-9343(80)90363-0

    8. Suissa S. Effectiveness of inhaled corticosteroids in chronic obstructive pulmonary disease: immortal time bias in observational studies. Am J Respir Crit Care Med. 2003;168(1):49–53. doi:10.1164/rccm.200210-1231OC

    9. Suissa S. Immortal time bias in pharmacoepidemiology. Am J Epidemiol. 2008;167(4):492–499. doi:10.1093/aje/kwm324

    10. Cain LE, Robins JM, Lanoy E, et al. When to start treatment? A systematic approach to the comparison of dynamic regimes using observational data. Int J Biostat. 2010;6(2). doi:10.2202/1557-4679.1212

    11. Hernán MA. How to estimate the effect of treatment duration on survival outcomes using observational data. BMJ. 2018;360:k182. doi:10.1136/bmj.k182

    12. Gaber CE, Ghazarian AA, Strassle PD, et al. De-mystifying the clone-censor-weight method for causal research using observational data: a primer for cancer researchers. Cancer Med. 2024;13(23):e70461. doi:10.1002/cam4.70461

    13. Quint JK, Mullerova H, DiSantostefano RL, et al. Validation of chronic obstructive pulmonary disease recording in the clinical practice research datalink (CPRD-GOLD). BMJ Open. 2014;4(7):e005540. doi:10.1136/bmjopen-2014-005540

    14. Rothnie KJ, Müllerová H, Thomas SL, et al. Recording of hospitalizations for acute exacerbations of COPD in UK electronic health care records. Clin Epidemiol. 2016;8:771–782. doi:10.2147/CLEP.S117867

    15. Suissa S, Dell’Aniello S, Ernst P. Comparative effectiveness of LABA-ICS versus LAMA as initial treatment in COPD targeted by blood eosinophils: a population-based cohort study. Lancet Respir Med. 2018;6(11):855–862. doi:10.1016/S2213-2600(18)30368-0

    16. Herrett E, Thomas SL, Schoonen WM, Smeeth L, Hall AJ. Validation and validity of diagnoses in the general practice research database: a systematic review. Br J Clin Pharmacol. 2010;69(1):4–14. doi:10.1111/j.1365-2125.2009.03537.x

    17. Herrett E, Gallagher AM, Bhaskaran K, et al. Data resource profile: clinical practice research datalink (CPRD). Int J Epidemiol. 2015;44(3):827–836. doi:10.1093/ije/dyv098

    18. Bogart M, Glassberg MB, Reinsch T, Stanford RH. Impact of prompt versus delayed initiation of triple therapy post COPD exacerbation in a US-managed care setting. Respir Med. 2018;145:138–144. doi:10.1016/j.rmed.2018.10.013

    19. Sicras Mainar A, Huerta A, Navarro Artieda R, et al. Economic impact of delaying initiation with multiple-inhaler maintenance triple therapy in Spanish patients with chronic obstructive pulmonary disease. Int J Chron Obstruct Pulmon Dis. 2019;14:2121–2129. doi:10.2147/COPD.S211854

    20. Sansbury LB, Wood RP, Anley GA, Nam Y, Ismaila AS. Quantifying the economic impact of delayed multiple-inhaler triple therapy initiation in patients with COPD: a retrospective cohort study of linked electronic medical record and hospital administrative data in England. Int J Chron Obstruct Pulmon Dis. 2021;16:2795–2808. doi:10.2147/COPD.S312853

    21. Tkacz J, Evans KA, Touchette DR, et al. PRIMUS – prompt initiation of maintenance therapy in the US: a real-world analysis of clinical and economic outcomes among patients initiating triple therapy following a COPD exacerbation. Int J Chron Obstruct Pulmon Dis. 2022;17:329–342. doi:10.2147/COPD.S347735

    22. Evans KA, Pollack M, Portillo E, et al. Prompt initiation of triple therapy following hospitalization for a chronic obstructive pulmonary disease exacerbation in the United States: an analysis of the PRIMUS study. J Manage Care Specialty Pharm. 2022;28(12):1366–1377. doi:10.18553/jmcp.2022.28.12.1366

    Continue Reading

  • Buy now, pay later loans will now affect US credit scores – what does that mean for consumers? | Buy now, pay later

    Buy now, pay later loans will now affect US credit scores – what does that mean for consumers? | Buy now, pay later

    A new change to buy now, pay later loans means borrowers’ credit scores may see a change, which has worried some users of the loans.

    “I have a feeling that I’m just not going to have as much access to spending power and zero or really low APR rates,” said Nicole Nitta, a 31-year-old Las Vegas resident, who uses BNPL and shared that she already does not have great credit.

    Fico, the credit scoring company used by most US lenders, announced on 23 June that they would include BNPL loans, which play “an increasingly important role in consumers’ financial lives”, to help lenders more “accurately evaluate credit readiness”.

    For users of companies like Affirm, Afterpay and Klarna, the new calculation could benefit them because it allows them to build their credit – if, of course, they pay back the loans on time, experts say.

    Nitta first used BNPL for essentials in 2021, like non-perishable food items. She was out of work and “basically living off of savings”, she said.

    Now, working as an office manager for a private therapy practice and studying marriage and family therapy, Nitta is more stable financially but has significant student loan debt. She has since used BNPL for Christmas gifts and dishware when she moved into a new apartment, but said she always makes her payments on time.

    Ted Rossman, a senior industry analyst at the financial site Bankrate, says: “if you’re using buy now, pay later responsibly,” like Nitta, “I would argue [the change] should help you as a steppingstone to improve your credit, and maybe it helps you get your first credit card or car loan.

    “The main downside is if it dings you because you’re paying late or racking up too much debt. I would say that’s a fair consequence, because that is what happens on credit cards and other products,” he added.

    Companies like Affirm, Afterpay and Klarna were founded more than a decade ago, but their usage expanded significantly during the Covid-19 pandemic. These companies provided $180m in loans totaling more than $24bn in 2021, an almost tenfold increase from 2019, according to the Consumer Financial Protection Bureau.

    Fumiko Hayashi, a vice-president at the Federal Reserve Bank of Kansas City who conducts economic research on payments, noted that the change was due to a shift from purchasing in stores to buying online – in addition to an economic downturn during the pandemic.

    A typical BNPL loan allows consumers to divide a $50 to $1,000 purchase into four interest-free instalments. If a borrower does not make the required payments, then the lender charges them a late fee. Lenders also charge transaction fees to merchants.

    BNPL is most popular among people ages 33 and under, who represented 70% of borrowers of such loans in 2022, according to the financial protection bureau.

    Hayashi noted that a downside for the younger users is “if they keep using BNPL only and they don’t use a credit card at all, they cannot build credit history”. With Fico’s change, using BNPL responsibly could be beneficial for some younger users with no previous credit history – but less so for those not as responsible.

    For most users, Fico and Affirm say, including BNPL data in credit reports produced higher scores or no score changes – a finding in a year-long report released in February that looked at 500,000 consumers.

    Still, there are people who could be hurt by Fico using BNPL data. People with sub-prime or deep sub-prime credit scores obtained more than 60% of new BNPL loans from 2021 to 2022, according to the financial protection bureau.

    And 24% of BNPL borrowers were late making a payment in 2024, a 6% increase from the prior year, according to the Federal Reserve. Among people who make $25,000 or less, the rate increased from 31% to 40%.

    Notably, BNPL access “significantly reduces the sensitivity of spending relative to income”, according to a Harvard Business School report.

    “This effect is concentrated among individuals likely to be liquidity constrained, specifically, lower-income users and users without credit cards,” the report notes.

    Becca, a 26-year-old tech worker in New York who declined to use her last name, said she used BNPL for things like pricier beauty products – including a Chanel perfume. She said she might “spend like 80 bucks this month on it and make two separate $40 payments, and then next month, I pay off the rest”.

    While the payment option has helped her, she is concerned about companies like DoorDash offering BNPL for minor purchases like a pizza delivery.

    “It’s just encouraging poor spending behavior from young people,” Becca said. “All these items build up because you’re using it again and again and again. You don’t feel like you’re spending a lot of money.”

    It may be some time before the economy feels the impact of the new credit score calculation, Rossman said. While Fico stated that it would make the new scores available in fall 2025, most lenders continue to use a credit score model from 2009, (despite Fico since releasing new versions).

    “Change comes relatively slowly in the credit scoring world, so even if this becomes available in the fall, that doesn’t mean everybody is going to be using it right away,” said Rossman.

    “It’s kind of like your phone. For instance, Apple has the iPhone 16, but a lot of people are still using the 15 or the 14 or even older models. Credit scoring works the same way.”

    Continue Reading

  • Salanersen Shows Promise in Phase 1, MESA Open-Label Data Released, Fenfluramine Meets End Points in Phase 3 GEMZ Trial

    Salanersen Shows Promise in Phase 1, MESA Open-Label Data Released, Fenfluramine Meets End Points in Phase 3 GEMZ Trial

    WATCH TIME: 4 minutes

    Welcome to this special edition of Neurology News Network. I’m Marco Meglio.

    Newly announced interim data from a phase 1 study showed that treatment with salanersen (Biogen), an investigational antisense oligonucleotide administered once a year, was safe and led to slowing of neurodegeneration in patients with spinal muscular atrophy (SMA) previously on gene therapy. Based on these findings, Biogen plans to test the therapy in phase 3 studies, the design of which is being discussed with the FDA. Building on the mechanism of action of nusinersen (Spinraza; Biogen), salanersen is designed to enhance potency and allow for once-yearly dosing, offering potential improvements in convenience and efficacy. Mechanistically, salanersen targets the SMN2 gene, modulating its splicing to increase production of functional survival motor neuron (SMN) protein, which is deficient in SMA.

    Edgewise Therapeutics has announced positive topline data from the MESA open-label extension study, with results showing that treatment with investigational sevasemten led to notable improvements in North Star Ambulatory Assessment (NSAA) scores over an 18-month period in patients with Becker muscular dystrophy (BMD). In addition, the company shared encouraging data from its phase 2 LYNX and FOX trials of Duchenne muscular dystrophy (DMD), as well as completion of a Type C meeting with the FDA, paving a path for sevasemten to become the first approved therapy for BMD. MESA, an open-label extension, featured 99% of eligible participants from the previously completed ARCH, CANYON, GRAND CANYON, or DUNE trials. In the latest data update, results showed a 0.8-point increase in NSAA scores among participants from CANYON, the major phase 2 trial of sevasemten, after 18 months of treatment. More notably, those who switched from placebo to sevasemten demonstrated a 0.2-point improvement since their crossover.

    In a new announcement from UCB, fenfluramine (Fintepla), an FDA-approved antiseizure medication, met its primary and secondary end points in the phase 3 GEMZ trial of patients with CDKL5 deficiency disorder (CDD). Based on these data, the company plans to submit an application for fenfluramine to become a potential treatment option for patients living with CDD. Between baseline and the titration plus maintenance phase, fenfluramine demonstrated statistically significant changes relative to placebo on the primary end point of percent change in countable motor seizure frequency. In this phase 3, double-blind, placebo-controlled, fixed-dose study, fenfluramine continued to show a safety profile that was consistent with its previous indications in Dravet syndrome (DS) and Lennox-Gastaut syndrome (LGS).

    For more direct access to expert insight, head to NeurologyLive.com. This has been Neurology News Network. Thanks for watching.

    Continue Reading

  • Dealmakers hit pause on M&A as caution rules the boardroom

    Dealmakers hit pause on M&A as caution rules the boardroom

    Unlock the Editor’s Digest for free

    Dealmaking slumped in the second quarter to the lowest level in a decade, excluding the early months of the pandemic, as Donald Trump’s “liberation day” tariffs extended a run of uncertainty that has forced dealmakers to pull back from all but the largest takeovers.

    The total number of deals announced in the three months to June 30 fell to about 10,900, according to data from the London Stock Exchange Group. Excluding the second quarter of 2022, when Covid-19 lockdowns upended global markets and just 10,600 deals were unveiled, the figure was the lowest since the start of 2015.

    Dealmakers had initially expected that a more conservative White House would pull back on regulation and unleash a wave of takeovers.

    Instead, companies and investors have had to navigate a more perilous geopolitical backdrop than anticipated, with the announcement of wide-ranging tariffs by the US on April 2 and conflicts in the Middle East driving volatility in markets.

    “Following the initial exuberance of the first month or two, the attitude in the boardroom has been cautious,” said Lorenzo Corte, global head of transactions at the law firm Skadden. 

    Despite the escalation of trade tensions since the start of the quarter in April, the LSEG data show that the value of transactions held steady from the first quarter of the year at $969bn, propped up by a handful of strategic megadeals.

    Deals worth more than $10bn have risen by three-quarters this year, with top transactions in the second quarter including Cox’s $35bn takeover of Charter Communications, a $33bn take-private of Toyota Motor’s biggest subsidiary, and a consortium led by Abu Dhabi National Oil Company’s $24bn acquisition of Australia’s Santos.

    “There’s pent up demand to do large strategic transactions,” said Jim Langston, partner at law firm Paul Weiss. “If companies are going to make a bet on M&A, they want it to be something that moves the needle, that the reward is worth the risk. ”

    The uncertain outlook for economic growth, inflation and the dollar have also acted as a particular drag on the private equity industry, making it more difficult to value assets.

    Global private-equity backed acquisitions slowed sharply between the first and second quarters of the year, from about 2,500 in the first three months to closer to 1,850 in the second. There were 1,250 fewer private equity deals struck in the first half of this year compared with the same period in 2024.

    Dealmakers have focused on public company takeovers and the sale or carve-out of assets regarded as no longer core, according to Jens Welter, Citi’s head of North America investment banking coverage.

    Such transactions include KKR’s £4.7bn acquisition of London-listed industrial group Spectris, and BP’s exploration of a sale for its lubricants arm Castrol. Welter said that dealmakers were adding in terms to contracts to help agree transactions in choppier markets.

    “While we expect the take-private and corporate carve-out volumes to remain at record levels, transactions are highly structured involving rollovers and deferred mechanisms,” Welter said.

    Some advisers remained optimistic that a stabilising geopolitical outlook would lead to a pick-up in activity in the second half of the year.

    Oliver Smith, co-head of Davis Polk’s M&A practice, said the build-up in demand felt like the early days of the Covid-19 pandemic.

    “People realised then that the sky wasn’t falling in and things picked up for a while,” said Smith. “It feels like that moment in time is coming once companies get used to the uncertainty.”

    Continue Reading

  • China’s first Legoland opens to visitors in Shanghai

    China’s first Legoland opens to visitors in Shanghai

    SHANGHAI — A giant 26-meter (85-foot) Lego figure named Dada welcomed visitors to the new Legoland resort in Shanghai.

    The resort, which opened Saturday, is the first in China. It is one of 11 parks across the world and was built with 85 million Lego bricks.

    Among the main attractions is Miniland, which replicates well-known sights from across the world using Lego bricks. It features landmarks across China like Beijing’s Temple of Heaven and Shanghai’s Bund waterfront. There’s also a boat tour through a historic Chinese water town built with Lego bricks.

    “My first impression is it is a good recreation, like a real fairyland of Lego,” said Ji Yujia, a Lego fan who was there on opening day.

    The resort was developed in conjunction with the Shanghai government by Merlin Entertainments and the LEGO Group.

    Visitors were greeted by performances featuring Legoland characters. Tickets range from $44 (319 yuan) to $84 (599 yuan).

    —-

    Corrects to say that Legoland in Shanghai is not the largest in the world.

    Continue Reading

  • Role of Artificial Intelligence in Minimizing Missed and Undiagnosed F

    Role of Artificial Intelligence in Minimizing Missed and Undiagnosed F

    Introduction

    Fractures take place in people of all age groups. The episode depends on the type of trauma, location, and associated injuries. The incidence of fractures ranges between 733 and 4017 per 100000 patient-years.1–3 Traumatic fractures are the major cause of morbidity and mortality, and in one study, 23,917 individuals sustained 27,169 fractures, with 64.5% of the fractures occurring in women.1 The epidemiological data for fractures and dislocations in Saudi Arabia are not available.4,5 It is expected that the number of fractures and dislocations will increase due to population growth.

    Figure 1 PRISMA flowchart Showing the Final Selection of Analyzed studies.

    Figure 2 Comparison between AI Model and Clinicians for Accuracy, Sensitivity and Specificity.

    The reported incidence of missed diagnosis of fractures or dislocations by plain radiographs ranges between 3% and 10%,6–8 and this inversely affects the final outcome of the recovery. The majority of the errors take place in the emergency room, where the radiographs are wrongly elucidated as some injuries might be tenuous, and in the majority, conspicuous injuries are missed due to improper training with sub-standard techniques employed in radiological evaluation.9 This could be more common in the junior residents under training in the emergency room and orthopedics and traumatology. Unfortunately, this is not uncommon in trained radiologists as well. In the USA, radiologists were at the 6th position in malpractice claims,10–14 even though they make up about 3.1% of the 892 million physicians.15 It becomes mandatory to find ways to reduce this discrepancy at both fronts at the training levels and the trained level, and one such tenet is to bring the utilization of AI in the field of diagnosis of fractures and dislocations.

    AI, which is part of computer science, can perform tasks that are usually performed by humans to humans. AI requires a high level of input from different images and then can use different algorithms using machine learning, deep learning, and convolutional neural networks to extricate high-level information from the input of images.16 Recent studies have suggested convincing accuracy of diagnosis of fractures and dislocations using AI algorithms, and with the objective to assess the accuracy, sensitivity, and specificity of AI algorithms in the diagnosis of fractures using plain radiographs, this review was carried out.

    Methods

    We searched all related electronic databases for English language literature between January 2015 and July 2023, Pub Med, Scopus, Web of Science, Cochrane Central Ovid Medline, Ovid Embase, EBSCO Cumulative Index to Allied Health Literature, Web of Science, and Cochrane Central with keywords of Artificial Intelligence, fractures, dislocations, X-rays, radiographs, missed diagnosis. All articles that fulfilled the following inclusion criteria: primary research using validated AI algorithms for fracture detection and Only studies with a comparative study between AI algorithms and clinicians were included in the analysis. Only studies with a comparative study between AI algorithms and clinicians were included in the analysis. All other publications and data were excluded, including reports by letter to the editor, conference presentations, and systematic reviews. EndNoteTM 39 was used to tabulate the references and delete any duplicates.

    Data Extraction

    We extracted available information from included studies fitting our inclusion criteria. The data extracted included a number of patients/images studied, site of fractures analyzed, algorithms used, the accuracy of the report based on the algorithm, sensitivity and specificity, area under the curve (AUC), comparison between the algorithm, junior orthopedic resident, emergency physicians, and board certified radiologists.

    Statistical Analysis

    The diagnostic prediction of the fractures of different algorithms was analyzed using contingency tables for validation. Regression analysis was performed between the different sites of fractures and the influence of the algorithms. A p-value of <0.05 was accepted as statistically significant at a 95% confidence interval (CI). SPSS (Statistical Package for Social Sciences) Inc., which is a statistical software developed by IBM for data management, advanced analytics, multivariate analysis, and business intelligence version 29, was used.

    Results

    We identified 2049 studies retrieved in which 347 were duplicates, and 1651 publications were excluded due to inclusion and exclusion criteria. Fifty-one studies were reviewed in depth as they nearly fulfilled the inclusion criteria, and only 27 publications fulfilled our objectives to be analyzed in detail and were included in this study (Figure 1). Eighty-eight thousand, nine hundred and ninety-six images were analyzed for fractures (Table 1), which showed that the overall accuracy of the correct diagnosis was 90.35±6.88 (73.59–98) percent, sensitivity 90.08±8.2 (73.8–99) percent, specificity 90.16±7 (72–100) and AUC was 0.931±0.06 (0.72–0.994). The fractures analyzed were common fractures from the wrist, upper and lower limbs, and spine. All studies had internally and externally validated algorithms for Diffusion-convolutional neural networks (DCNN). The majority of the studies limited their analysis for diagnoses based on a single view of the radiograph.

    Table 1 Characteristics of Studies, Number of Images Analyzed, Site of Fractures, Algorithms Used, Accuracy, Sensitivity, Specificity and Area Under Curve

    Table 2 shows the analysis of 214950 images where a comparison was made between the AI algorithm versus a junior resident in training. The accuracy of the AI model was 94.24±4.19, and that of orthopedic resident was 85.18±7.01 (P value of <0.0001), with sensitivity 92.15±7.12 versus 86.38±7.6 (P<0.0001) and specificity of 93.77±4.03 versus 87.05±12.9 (P<0.0001). Yamada et al (2020) 40 compared the AI model versus orthopedic residents and board-certified radiologists and found the accuracy to be 98% versus 87% and 92% (P value of <0.0001). Figure 2 shows the comparison between the AI model and the clinician for accuracy, sensitivity, and specificity.

    Table 2 Comparative Data Between the AI Models and Clinicians

    Discussion

    This review shows that accuracy in the diagnosis of fractures using AI algorithms surpasses that of the trained and trainee residents. Secondly, the use of AI helped the trainees and trained radiologists in improving the accuracy, sensitivity, and specificity of fracture diagnosis. In this study, the AI with different models showed that the overall accuracy of the correct diagnosis was 90.35±6.88%, sensitivity 90.08±8.2%, specificity 90.16±7 and AUC was 0.931±0.06. These results were based on plain radiographs and included all limb and vertebral fractures.

    In the recent past, there has been a consequential increase in different AI models, particularly CNNs, in the arena of trauma and orthopedics. Individual models have conclusively shown that AI models are accurate in the diagnosis of fractures, which are better than junior residents and, if not better, but at par with the senior radiologist. One aspect that needs to be questioned is that most of the reported data comes from retrospective testing, and few only are based prospectively on clinical practice. The accuracy of diagnosis of fractures varied at different sites of fractures. Murphy et al (2022)44 reported an analysis of hip fractures, comparing the AI model with two trained and expert clinicians, and found that the AI model was 19% more accurate than the physicians. Another report suggested that the sensitivity of the correct diagnosis increases by over 10%. Lindsey et al (2018)33 reported that the physician’s average sensitivity in the diagnosis of fractures improved from 80.8% to 91.5% (95% CI, 89.3–92.9%), and specificity was 87.5% to 93.9% (95% CI, 92.9–94.9%) when they were aided with Deep convolutional neural network and added to this the physicians experienced a reduction in misreading around 47.0%. Duron et al (2021)42 further concurred after their review that emergency room physicians improved their results after AI assistance from 61.3% to 74.3% (up 13.0%), and the trained radiologists enhanced their diagnosis from 80.2% to 84.6% (up 4.3%). Distal radius fractures, which amount to over 20% of all fractures, were studied using an ensemble model of AI between three groups: AI, orthopedic surgeons, and radiologists, and it was reported statistically significant between the three groups. The accuracy, sensitivity, and specificity between the attending orthopedic surgeons and radiologists showed significant differences: 93.69%, 91.94%, and 95.44% compared to 92.53%, 90.44%, and 94.62%. When the physician’s groups were compared to the AI ensemble tool, it was a highly significant score of 97.75%, 97.13%, and 98.37% by the AI tool.43

    Missed extremity fracture diagnosis in trauma practice has always been an issue and is the second most injuries to be misdiagnosed.45 The most common malpractice claims against radiologists involve inaccuracies in the reporting of extremity fractures.10,46,47 Orthopaedic residents are not immune to making misinterpretations of radiographs in extremity fractures. One such study from the United Kingdom highlights that Senior Orthopaedic Residents on plain radiographs missed 4% of fractures, 7.8% made a wrong diagnosis, and 12.6%, a fracture was diagnosed when there was none.48 Report indicates that over the years, the number of claims against orthopadicians has increased, but complaints have remained comparatively the same.49 In the present belligerent and litigation-oriented society, it is imperative that junior orthopedic residents have all the help in making a correct fracture diagnosis and not miss even a meager injury. AI and its algorithms can never replace human doctors but can unquestionably enhance and complement in improving the accuracy of fracture diagnosis.37 Moreover, adequate and timely training of trainee residents in radiographic interpretation is paramount. It was reported that junior residents till 3rd of training level are more vulnerable to making errors in radiographic interpretation.9

    Our review has limitations due to the number of studies we have included in the analysis, as there are a number of publications that are increasing by the day, and it is possible that we have not included the most recent literature. Secondly, we could not add the data of comparative accuracy between the unaided and aided AI tools in the fracture diagnosis. Lastly, we are basing the conclusion on the retrospective studies, and there were no prospective studies to compare with. The strength of the study is we have compared a large dataset, which suggests that the different AI models are more accurate than the physicians.

    In conclusion, this review highlights with unbiased evaluations recommend that the use of AI models can definitely help residents in training by increasing the accuracy of fracture diagnosis and reducing the errors in diagnosis of fractures. AI has developed cutting edge tools, which need to be further evaluated so that procurement authorities in hospitals could integrate AI into healthcare and help physicians at all levels to improve correctness in fracture diagnosis, to prevent complications of delayed diagnosis.

    Disclosure

    The authors report no conflicts of interest in this work.

    References

    1. Bergh C, Wennergren D, Möller M, Brisby H. Fracture incidence in adults in relation to age and gender: a study of 27,169 fractures in the Swedish fracture register in a well-defined catchment area. PLoS One. 2020;15(12):e0244291. doi:10.1371/journal.pone.0244291

    2. Amin S, Achenbach SJ, Atkinson EJ, Khosla S, Melton LJ. Trends in fracture incidence: a population-based study over 20 years. J Bone Miner Res. 2014;29(3):581–589. doi:10.1002/jbmr.2072

    3. Curtis EM, van der Velde R, Moon RJ, et al. Epidemiology of fractures in the United Kingdom 1988-2012: variation with age, sex, geography, ethnicity and socioeconomic status. Bone. 2016;87:19–26. doi:10.1016/j.bone.2016.03.006

    4. Sadat-Ali M, Ahlberg A. Fractured neck of the femur in young adults. Injury. 1992;23:311–313. doi:10.1016/0020-1383(92)90176-S

    5. Sadat-Ali M, AlOmran AS, Azam MQ, et al. Epidemiology of fractures and dislocations among urban communities of Eastern Saudi Arabia. Saudi J Med Med Sci. 2015;3:54–57. doi:10.4103/1658-631X.149682

    6. Wei CJ, Tsai WC, Tiu CM, Wu HT, Chiou HJ, Chang CY. Systematic analysis of missed extremity fractures in emergency radiology. Acta Radiol. 2006;47(7):710–717. doi:10.1080/02841850600806340

    7. Williams SM, Connelly DJ, Wadsworth S, Wilson DJ. Radiological review of accident and emergency radiographs: a 1-year audit. Clin Radiol. 2000;55(11):861–865. doi:10.1053/crad.2000.0548

    8. Hallas P, Ellingsen T. Errors in fracture diagnoses in the emergency department characteristics of patients and diurnal variation. BMC Emerg Med. 2006;6(1):4. doi:10.1186/1471-227X-6-4

    9. Pinto A, Berritto D, Russo A, et al. Traumatic fractures in adults: missed diagnosis on plain radiographs in the Emergency Department. Acta Biomed. 2018;89(1–S):111–123.

    10. Whang JS, Baker SR, Patel R, Luk L, Castro A. The causes of medical malpractice suits against radiologists in the United States. Radiology. 2013;266:548–554. doi:10.1148/radiol.12111119

    11. De Filippo M, Pesce A, Barile A, et al. Imaging of postoperative shoulder instability. Musculoskeletal Surg. 2017;101:15–22. doi:10.1007/s12306-017-0461-4

    12. Splendiani A, Bruno F, Patriarca L, et al. Thoracic spine trauma: advanced imaging modality. Radiol Med. 2016;121:780–792. doi:10.1007/s11547-016-0657-y

    13. de Filippo M, Azzali E, Pesce A, et al. CT arthrography for evaluation of autologous chondrocyte and chondral-inductor scaffold implantation in the osteochondral lesions of the talus. Acta Biomed. 2016;87:51–56.

    14. Splendiani A, Perri M, Grattacaso G, et al. Magnetic resonance imaging (MRI) of the lumbar spine with dedicated G-scan machine in the upright position: a retrospective study and our experience in 10 years with 4305 patients. Radiol Med. 2016;121:38–44. doi:10.1007/s11547-015-0570-9

    15. Available from: https://www.aamc.org/data-reports/workforce/data/number-people-active-physician-specialty-2021. Assessed July 25, 2024.

    16. Bizzo BC, Almeida RR, Michalski MH, Alkasab TK. Artificial intelligence and clinical decision support for radiologists and referring providers. J Am Coll Radiol. 2019;16(9 Pt B):1351–1356. (). doi:10.1016/j.jacr.2019.06.010

    17. Kim DH, MacKinnon T. Artificial intelligence in fracture detection: transfer learning from deep convolutional neural networks. Clin Radiol. 2018;73(5):439–445. doi:10.1016/j.crad.2017.11.015

    18. Adams M, Chen W, Holcdorf D, McCusker MW, Howe PD, Gail- Lard F. Computer vs human: deep learning versus perceptual training for the detection of neck of femur fractures. J Med Imaging Radiat Oncol. 2019;63(1):27–32. doi:10.1111/1754-9485.12828

    19. Cheng CT, Ho TY, Lee TY, et al. Application of a deep learning algorithm for detection and visualization of hip fractures on plain pelvic radiographs. Eur Radiol. 2019;29(10):5469–5477. doi:10.1007/s00330-019-06167-y

    20. Derkatch S, Kirby C, Kimelman D, Jozani MJ, Davidson JM, Leslie WD. Identification of vertebral fractures by convolutional neural networks to predict nonvertebral and hip fractures: a registry-based cohort study of dual X-ray absorptiometry. Radiology. 2019;293(2):405–411. doi:10.1148/radiol.2019190201

    21. Rayan JC, Reddy N, Kan JH, Zhang W, Annapragada A. Binomial classification of. pediatric elbow fractures using a deep learning multi-view approach emulating radiologist decision making. Radiol Artif Intell. 2019;1(1):e180015. doi:10.1148/ryai.2019180015

    22. Starosolski ZA, Kan H, Annapragada AV. CNN-based radiographic acute tibial fracture detection in the setting of open growth plates. bioRxiv pre- print bioRxiv:506154. Available from: https://www.biorxiv.org/content/10.1101/506154. Accessed July 25, 2024.

    23. Choi JW, Cho YJ, Lee S, et al. Using a dual-input convolutional neural network for automated detection of pediatric supracondylar fracture on conventional radiography. Invest Radiol. 2020;55(2):101–110. doi:10.1097/RLI.0000000000000615

    24. Jiménez-Sánchez A, Kazi A, Albarqouni S, et al. Precise proximal femur fracture classification for interactive training and surgical planning. Int J CARS. 2020;15(5):847–857. doi:10.1007/s11548-020-02150-x

    25. Mawatari T, Hayashida Y, Katsuragawa S, et al. The effect of deep convolutional neural networks on radiologists’ performance in the detection of hip fractures on digital pelvic radiographs. Eur J Radiol. 2020;130:109188. doi:10.1016/j.ejrad.2020.109188

    26. Chen HY, Hsu BW, Yin YK, et al. A human-algorithm integration system for hip fracture detection on plain radiography: system development and validation study. JMIR Med Inform. 2020;8(11):e19416. doi:10.2196/19416

    27. Cheng CT, Wang Y, Chen HW, et al. A scalable physician-level deep learning algorithm detects universal trauma on pelvic radiographs. Nat Commun. 2021;12(1):1066. doi:10.1038/s41467-021-21311-3

    28. Raisuddin AM, Vaattovaara E, Nevalainen M, et al. Critical evaluation of deep neural networks for wrist fracture detection. Sci Rep. 2021;11(1):6006. doi:10.1038/s41598-021-85570-2

    29. Yoon AP, Lee YL, Kane RL, Kuo CF, Lin C, Chung KC. Development and validation of a deep learning model using convolutional neural net- works to identify scaphoid fractures in radiographs. JAMA Network Open. 2021;4(5):e216096. doi:10.1001/jamanetworkopen.2021.6096

    30. Grauhan NF, Niehues SM, Gaudin RA, et al. Deep learning for accurately recognizing common causes of shoulder pain on radiographs. Skeletal Radiol. 2022;51(2):355–362. doi:10.1007/s00256-021-03740-9

    31. Ozkaya E, Topal FE, Bulut T, Gursoy M, Ozuysal M, Karakaya Z. Evaluation of an artificial intelligence system for diagnosing scaphoid fracture on direct radiography. Eur J Trauma Emerg Surg. 2022;48(1):585–592. doi:10.1007/s00068-020-01468-0

    32. Chung SW, Han SS, Lee JW, et al. Automated detection and classification of the proximal humerus fracture by using deep learning algorithm. Acta Orthop. 2018;89(4):468–473. doi:10.1080/17453674.2018.1453714

    33. Lindsey R, Daluiski A, Chopra S, Lachapelle A, Mozer M. Sicular S et al. Deep neural network improves fracture detection by clinicians. Proc Natl Acad Sci U S A. 2018;115(45):11591–11596. doi:10.1073/pnas.1806905115

    34. Wang Y, Lu L, Cheng CT, et al. Weakly supervised universal fracture detection in pelvic x-rays. In: Shen D, Liu T, Peters TM, et al. editors. Medical Image Computing and Computer Assisted Intervention – MICCAI 2019. MICCAI 2019. Lecture Notes in Computer Science. Vol. 11769, Cham, Switzerland: Springer; 2019:459–467.

    35. Blüthgen C, Becker AS, Vittoria de Martini I, Meier A, Martini K, Frauen-felder T. Detection and localization of distal radius fractures: deep learning system versus radiologists. Eur J Radiol. 2020;126:108925. doi:10.1016/j.ejrad.2020.108925

    36. Chen HY, Hsu BW, Yin YK, et al. Application of deep learning algorithm to detect and visualize vertebral fractures on plain frontal radiographs. PLoS One. 2021;16(1):e0245992. doi:10.1371/journal.pone.0245992

    37. Cheng CT, Chen CC, Cheng FJ, et al. Human-algorithm integration system for hip fracture detection on plain radiography: system development and validation study. JMIR Med Inform. 2020;8(11):e19416.

    38. Krogue JD, Cheng KV, Hwang KM, et al. Automatic Hip fracture identification and functional subclassification with deep learning. Radiol Artif Intell. 2020;2(2):e190023. doi:10.1148/ryai.2020190023

    39. Murata K, Endo K, Aihara T, et al. Artificial intelligence for the detection of vertebral fractures on plain spinal radiography. Sci Rep. 2020;10(1):20031. doi:10.1038/s41598-020-76866-w

    40. Yamada Y, Maki S, Kishida S, et al. Automated classification of hip fractures using deep convolutional neural networks with orthopedic surgeon- level accuracy: ensemble decision-making with antero-posterior and lateral radiographs. Acta Orthop. 2020;91(6):699–704. doi:10.1080/17453674.2020.1803664

    41. Yu JS, Yu SM, Erdal BS, et al. Detection and localisation of hip fractures on anteroposterior radiographs with artificial intelligence: proof of concept. Clin Radiol. 2020;75(3):237.e1–237e9. doi:10.1016/j.crad.2019.10.022

    42. Duron L, Ducarouge A, Gillibert A, et al. Assessment of an AI aid in detection of adult appendicular skeletal fractures by emergency physicians and radiologists: a multicenter cross-sectional diagnostic study. Radiology. 2021;300(1):120–129. doi:10.1148/radiol.2021203886

    43. Zhang J, Li Z, Li H, et al. Deep learning assisted diagnosis system: improving the diagnostic accuracy of distal radius fractures. Front Med. 2023;10:1224489. PMID: 37663656; PMCID: PMC10471443. doi:10.3389/fmed.2023.1224489

    44. Murphy EA, Ehrhardt B, Gregson CL, et al. Machine learning outperforms clinical experts in classification of Hip fractures. Sci Rep. 2022;12(1):2058. PMID: 35136091; PMCID: PMC8825848. doi:10.1038/s41598-022-06018-9

    45. Porrino JA, Maloney E, Scherer K, Mulcahy H, Ha AS, Allan C. Fracture of the distal radius: epidemiology and premanagement radiographic characterization. Am J Roentgenol. 2014;203:551–559. doi:10.2214/AJR.13.12140

    46. Guly HR. Diagnostic errors in an accident and emergency department. Emerg Med J. 2001;18:263–269. doi:10.1136/emj.18.4.263

    47. Festekjian A, Kwan KY, Chang TP, Lai H, Fahit M, Liberman DB. Radiologic discrepancies in children with special healthcare needs in a pediatric emergency department. Am J Emerg Med. 2018;36:1356–1362. doi:10.1016/j.ajem.2017.12.041

    48. Sharma H, Bhagat S, Gaine WJ. Reducing diagnostic errors in musculoskeletal trauma by reviewing non-admission orthopaedic referrals in the next-day trauma meeting. Ann R Coll Surg Engl. 2007;89(7):692–695. doi:10.1308/003588407X205305

    49. Khan IH, Jamil W, Lynn SM, Khan OH, Markland K, Giddins G. Analysis of NHSLA claims in orthopedic surgery. Orthopedics. 2012;35:726–731. doi:10.3928/01477447-20120426-28

    Continue Reading

  • PSX soars: KSE-100 gains 7,570 points in first week of fiscal year

    PSX soars: KSE-100 gains 7,570 points in first week of fiscal year





    PSX soars: KSE-100 gains 7,570 points in first week of fiscal year – Daily Times


































    Continue Reading

  • Lee ECZ, Anand VV, Razavi AC, Alebna PL, Muthiah MD, Siddiqui MS et al. The global epidemic of metabolic fatty liver disease. Curr Cardiol Rep. 2024;26(4).

  • Liu J, Ayada I, Zhang X, Wang L, Li Y, Wen T et al. Estimating global prevalence of metabolic Dysfunction-Associated fatty liver disease in overweight or obese adults. Clin Gastroenterol Hepatol. 2022;20(3).

  • Younossi ZM, Golabi P, Paik J, Owrangi S, Yilmaz Y, El-Kassas M et al. Prevalence of metabolic dysfunction-associated steatotic liver disease in the middle East and North Africa. Liver Int. 2024;44(4).

  • Byrne CD, Targher G. NAFLD: A multisystem disease. J Hepatol. 2015;62(S1):S47–64.

    Article 
    PubMed 

    Google Scholar 

  • Ha J, Hong OK, Han K, Kwon HS. Metabolic dysfunction-associated fatty liver disease increases the risk of type 2 diabetes mellitus in young Korean adults. Diabetes Res Clin Pract. 2024;212.

  • Ma YB, Cheng N, Lu YB, Li HY, Li JS, Ding J et al. [Association between fatty liver and type 2 diabetes in the baseline population of Jinchang cohort]. Zhonghua Liu Xing Bing Xue Za Zhi. 2018;39(6).

  • Yu Y, Yu Y, Wang Y, Chen Y, Wang N, Wang B et al. Nonalcoholic fatty liver disease and type 2 diabetes: an observational and Mendelian randomization study. Front Endocrinol (Lausanne). 2023;14.

  • Kim KS, Hong S, Han K, Park CY. Association of non-alcoholic fatty liver disease with cardiovascular disease and all cause death in patients with type 2 diabetes mellitus: nationwide population based study. BMJ. 2024;384.

  • Targher G, Corey KE, Byrne CD, Roden M. The complex link between NAFLD and type 2 diabetes mellitus — mechanisms and treatments. Volume 18. Nature Reviews Gastroenterology and Hepatology; 2021.

  • Poulos JE, Kalogerinis PT, Milanov V, Kalogerinis CT, Poulos EJ. The effects of vitamin E, Silymarin and carnitine on the metabolic abnormalities associated with nonalcoholic liver disease. J Diet Suppl. 2022;19(3).

  • Li S, Hong M, Tan HY, Wang N, Feng Y. Insights into the Role and Interdependence of Oxidative Stress and Inflammation in Liver Diseases. Vol. 2016, Oxidative Medicine and Cellular Longevity. 2016.

  • Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Volume 152. Free Radical Biology and Medicine 2020.

  • Reyes-Gordillo K, Shah R, Muriel P. Oxidative Stress and Inflammation in Hepatic Diseases: Current and Future Therapy. Vol. 2017, Oxidative Medicine and Cellular Longevity. 2017.

  • Cichoz-Lach H, Michalak A. Oxidative stress as a crucial factor in liver diseases. 20, World J Gastroenterol. 2014.

  • Heeba GH, Morsy MA. Fucoidan ameliorates steatohepatitis and insulin resistance by suppressing oxidative stress and inflammatory cytokines in experimental non-alcoholic fatty liver disease. Environ Toxicol Pharmacol. 2015;40(3):907–14.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Rahman MM, Islam MR, Shohag S, Hossain ME, Rahaman MS, Islam F, et al. The multifunctional role of herbal products in the management of diabetes and obesity: a comprehensive review. Molecules. 2022;27(5):1713.

    Article 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 

  • Zhang CY, Liu S, Yang M. Antioxidant and anti-inflammatory agents in chronic liver diseases: molecular mechanisms and therapy. World J Hepatol. 2023;15(2):180.

    Article 
    PubMed 
    PubMed Central 

    Google Scholar 

  • Unuofin JO, Lebelo SL. Antioxidant effects and mechanisms of medicinal plants and their bioactive compounds for the prevention and treatment of type 2 diabetes: an updated review. Oxid Med Cell Longev. 2020;2020.

  • Inzaugarat ME, De Matteo E, Baz P, Lucero D, García CC, Ballerga EG et al. New evidence for the therapeutic potential of Curcumin to treat nonalcoholic fatty liver disease in humans. PLoS ONE. 2017;12(3).

  • Aslam F, Iqbal S, Imran M, Anjum AA, Gilani SA, Farooq Z, et al. Characterization of white Sesame seed oil and its bioactive components. J Microbiol Biotechnol Food Sci. 2021;10(6):1–4.

    Article 

    Google Scholar 

  • Bigoniya P, Nishad R, Singh CS. Preventive effect of Sesame seed cake on hyperglycemia and obesity against high fructose-diet induced type 2 diabetes in rats. Food Chem. 2012;133(4).

  • Atefi M, Ghavami A, Hadi A, Askari G. The effect of barberry (Berberis vulgaris L.) supplementation on blood pressure: A systematic review and meta-analysis of the randomized controlled trials. Complement Ther Med. 2021;56:102608.

    Article 
    PubMed 

    Google Scholar 

  • Aslam F, Iqbal S, Nasir M, Anjum AA. White Sesame seed oil mitigates blood glucose level, reduces oxidative stress, and improves biomarkers of hepatic and renal function in participants with type 2 diabetes mellitus. J Am Coll Nutr. 2019;38(3):235–46.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Oboulbiga EB, Douamba Z, Compaoré-Sérémé D, Semporé JN, Dabo R, Semde Z et al. Physicochemical, potential nutritional, antioxidant and health properties of Sesame seed oil: a review. 10, Frontiers in Nutrition. 2023.

  • Huang SM, Chuang CH, Rejano CJF, Tayo LL, Hsieh CY, Huang SKH et al. Sesamin: A promising therapeutic agent for ameliorating symptoms of diabetes. Molecules. 2023;28(21).

  • Hong L, Yi W, Liangliang C, Juncheng H, Qin W, Xiaoxiang Z. Hypoglycaemic and hypolipidaemic activities of Sesamin from Sesame meal and its ability to ameliorate insulin resistance in KK-Ay mice. J Sci Food Agric. 2013;93(8):1833–8.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Breslow JL. n-3 fatty acids and cardiovascular disease. Am J Clin Nutr. 2006;83(6):S1477–82.

    Article 

    Google Scholar 

  • Simopoulos a P. The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular disease and other chronic diseases. Exp Biol Med (Maywood). 2008;233(6):674–88.

    Article 
    PubMed 

    Google Scholar 

  • George ES, Forsyth A, Itsiopoulos C, Nicoll AJ, Ryan M, Sood S, et al. Practical dietary recommendations for the prevention andmanagement of nonalcoholic fatty liver disease in adults. Adv Nutr. 2018;9(1):30–40.

    Article 
    PubMed 
    PubMed Central 

    Google Scholar 

  • Tian A, Sun Z, Zhang M, Li J, Pan X, Chen P. Associations between dietary fatty acid patterns and non-alcoholic fatty liver disease in typical dietary population: A UK biobank study. Front Nutr. 2023;10.

  • Wacal C, Ogata N, Basalirwa D, Sasagawa D, Kato M, Handa T et al. Fatty acid composition of Sesame (Sesamum indicum L.) seeds in relation to yield and soil chemical properties on continuously monocropped upland fields converted from paddy fields. Agronomy. 2019;9(12).

  • Hamilton JS, Klett EL. Linoleic acid and the regulation of glucose homeostasis: A review of the evidence. Volume 175. Prostaglandins Leukotrienes and Essential Fatty Acids 2021.

  • Tuo Y, Wang D, Li S, Chen C. Long-term exposure of INS-1 rat Insulinoma cells to Linoleic acid and glucose in vitro affects cell viability and function through mitochondrial-mediated pathways. Endocrine. 2011;39(2).

  • Raeisi-Dehkordi H, Amiri M, Zimorovat A, Moghtaderi F, Zarei S, Forbes SC, et al. Canola oil compared with Sesame and Sesame-canola oil on glycaemic control and liver function in patients with type 2 diabetes: A three-way randomized triple-blind cross-over trial. Diabetes Metab Res Rev. 2021;37(5):e3399.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Pan JH, Kim MJ, Kim JH, Cho YJ, Shin HS, Sung JS, et al. Inhibition of the lipogenesis in liver and adipose tissue of diet-induced obese C57BL/6 mice by feeding oleic acid-rich Sesame oil. Food Sci Biotechnol. 2015;24(3):1115–21.

    Article 
    CAS 

    Google Scholar 

  • Rehman K, Haider K, Jabeen K, Akash MSH. Current perspectives of oleic acid: regulation of molecular pathways in mitochondrial and endothelial functioning against insulin resistance and diabetes. 21, Reviews in Endocrine and Metabolic Disorders. 2020.

  • Granado-Casas M, Mauricio D. Oleic acid in the diet and what it does: implications for diabetes and its complications. In: Bioactive Food as Dietary Interventions for Diabetes. 2019.

  • Haeiwa H, Fujita T, Saitoh Y, Miwa N. Oleic acid promotes adaptability against oxidative stress in 3T3-L1 cells through lipohormesis. Mol Cell Biochem. 2014;386(1–2).

  • Belury MA. Linoleic acid, an omega-6 fatty acid that reduces risk for cardiometabolic diseases: premise, promise and practical implications. 26, Current Opinion in Clinical Nutrition and Metabolic Care. 2023.

  • Chavali SR, Zhong WW, Forse RA. Dietary α-linolenic acid increases TNF-α, and decreases IL-6, IL-10 in response to LPS: effects of Sesamin on the ∆-5 desaturation of ω6 and ω3 fatty acids in mice. Prostaglandins Leukot Essent Fat Acids. 1998;58(3):185–91.

    Article 
    CAS 

    Google Scholar 

  • Afroz M, Zihad SMNK, Uddin SJ, Rouf R, Rahman MS, Islam MT et al. A systematic review on antioxidant and antiinflammatory activity of Sesame (Sesamum indicum L.) oil and further confirmation of antiinflammatory activity by chemical profiling and molecular Docking. Phytother Res. 2019;33.

  • Abdul-Lattif RF. Effect of Sesame oil on lipid profile and liver enzymes in male albino rats treated with Carbone tetrachloride (CCl4). Ibn AL- Haitham. J Pure Appl Sci. 2018;41.

  • Periasamy S, Hsu DZ, Chang PC, Liu MY. Sesame oil attenuates nutritional fibrosing steatohepatitis by modulating matrix metalloproteinases-2, 9 and PPAR-γ. J Nutr Biochem. 2014;25(3):337–44.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Atefi M, Entezari MH, Vahedi H, Hassanzadeh A. Sesame Oil Ameliorates Alanine Aminotransferase, Aspartate Aminotransferase, and Fatty Liver Grade in Women with Nonalcoholic Fatty Liver Disease Undergoing Low-Calorie Diet: A Randomized Double-Blind Controlled Trial. Int J Clin Pract. 2022;2022.

  • Moher D, Hopewell S, Schulz KF, Montori V, Gøtzsche PC, Devereaux PJ et al. CONSORT 2010 explanation and elaboration: updated guidelines for reporting parallel group randomised trials. BMJ. 2010;340.

  • Jaceldo-Siegl K, Haddad E, Knutsen S, Fan J, Lloren J, Bellinger D, Nutrition et al. Metabolism Cardiovasc Dis. 2018;28(8).

  • Carter RE, Sonne SC, Brady KT. Practical considerations for estimating clinical trial accrual periods: application to a multi-center effectiveness study. BMC Med Res Methodol. 2005;5.

  • Sohouli MH, Haghshenas N, Hernández-Ruiz Á, Shidfar F. Consumption of Sesame seeds and Sesame products has favorable effects on blood glucose levels but not on insulin resistance: A systematic review and meta‐analysis of controlled clinical trials. Phytother Res. 2022;36(3):1126–34.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Atefi M, Entezari MH, Vahedi H, Hassanzadeh A. The effects of Sesame oil on metabolic biomarkers: a systematic review and meta-analysis of clinical trials. 21, J Diabetes Metab Disord. 2022.

  • Aalipanah E, Berizi E, Khamoushi A, Zare M, Sohrabi Z. Edible oils consumption and the fatty acids profile of the most common consumed brands in shiraz, Iran. Indian J Forensic Med Toxicol. 2021;15(1).

  • Mifflin MD, St Jeor ST, Hill LA, Scott BJ, Daugherty SA, Koh YO. A new predictive equation for resting energy expenditure in healthy individuals. Am J Clin Nutr. 1990;51(2):241–7.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Gasparini A, Ferrentino G, Angeli L, Morozova K, Zatelli D, Scampicchio M. Ultrasound assisted extraction of oils from Apple seeds: A comparative study with supercritical fluid and conventional solvent extraction. Innovative Food Sci Emerg Technol. 2023;86.

  • Gutch M, Kumar S, Razi SM, Gupta K, Gupta A. Assessment of insulin sensitivity/resistance. Indian J Endocrinol Metab. 2015;19(1):160–4.

    Article 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 

  • Pisprasert V, Ingram KH, Lopez-Davila MF, Munoz AJ, Garvey WT. Limitations in the use of indices using glucose and insulin levels to predict insulin sensitivity: impact of race and gender and superiority of the indices derived from oral glucose tolerance test in African Americans. Diabetes Care. 2013;36(4):845–53.

    Article 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 

  • Chow SC, Shao J, Wang H, Lokhnygina Y. Sample size calculations in clinical research, third edition. Sample Size Calculations in Clinical Research, Third Edition. Chapman and Hall/CRC 2017;1–511.

  • Rezaei S, Akhlaghi M, Sasani MR, Barati Boldaji R. Olive oil lessened fatty liver severity independent of cardiometabolic correction in patients with non-alcoholic fatty liver disease: A randomized clinical trial. Nutrition. 2019;57.

  • Bo T, Gao L, Yao Z, Shao S, Wang X, Proud CG, et al. Hepatic selective insulin resistance at the intersection of insulin signaling and metabolic dysfunction-associated steatotic liver disease. Cell Metab. 2024;36(5):947–68.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Mondal P, Prasad A, Girdhar K. Interventions to improve β-cell mass and function. Annales d’endocrinologie. Elsevier 2017;469–77.

  • Rariden C. Prediabetes: early identification and management. Nurse Pract. 2018;43(12).

  • Gast KB, Tjeerdema N, Stijnen T, Smit JWA, Dekkers OM. Insulin resistance and risk of incident cardiovascular events in adults without diabetes: Meta-Analysis. PLoS ONE. 2012;7(12).

  • Farajbakhsh A, Mazloomi SM, Mazidi M, Rezaie P, Akbarzadeh M, Ahmad SP, et al. Sesame oil and vitamin E co-administration May improve cardiometabolic risk factors in patients with metabolic syndrome: a randomized clinical trial. Eur J Clin Nutr. 2019;73(10):1403–11.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Mitra A. Study on the benefits of Sesame oil over coconut oil in patients of insulin resistance syndrome, notably type 2 diabetes and dyslipidaemia. J Hum Ecol. 2007;22(1):61–6.

    Article 

    Google Scholar 

  • Imamura F, Micha R, Wu JHY, de Oliveira Otto MC, Otite FO, Abioye AI, et al. Effects of saturated fat, polyunsaturated fat, monounsaturated fat, and carbohydrate on glucose-insulin homeostasis: A systematic review and meta-analysis of randomised controlled feeding trials. PLoS Med. 2016;13(7):e1002087.

    Article 
    PubMed 
    PubMed Central 

    Google Scholar 

  • Sutanto K, Sunardi D, Felicia A. Metabolic effects of monounsaturated fatty acid– enriched diets on glycaemic control in patient with type 2 diabetes: an evidence-based case report. World Nutr J. 2023;6(2).

  • Paniagua JA, de la Sacristana AG, Sánchez E, Romero I, Peréz-Martinez P, López-Miranda J, et al. A MUFA-rich diet improves posprandial glucose, lipid and GLP-1 responses in insulin-resistant subjects. J Am Coll Nutr. 2007;26(5):434–44.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Yang ZH, Miyahara H, Mori T, Doisaki N, Hatanaka A. Beneficial effects of dietary fish-oil-derived monounsaturated fatty acids on metabolic syndrome risk factors and insulin resistance in mice. J Agric Food Chem. 2011;59(13):7482–9.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Rocca AS, Lagreca J, Kalitsky J, Brubaker PL. Monounsaturated fatty acid diets improve glycemic tolerance through increased secretion of glucagon-like peptide-1. Endocrinology. 2001;142(3):1148–55.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Yargholi A, Najafi MH, Zareian MA, Hawkins J, Shirbeigi L, Ayati MH. The effects of Sesame consumption on glycemic control in adults: A systematic review and meta-analysis of randomized clinical trial. Evidence-based Complementary and Alternative Medicine. 2021;2021.

  • Nakai M, Harada M, Nakahara K, Akimoto K, Shibata H, Miki W, et al. Novel antioxidative metabolites in rat liver with ingested Sesamin. J Agric Food Chem. 2003;51(6):1666–70.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Namayandeh SM, Kaseb F, Lesan S. Olive and Sesame oil effect on lipid profile in hypercholesterolemic patients, which better? Int J Prev Med. 2013;4(9):1059–62.

    PubMed 
    PubMed Central 

    Google Scholar 

  • Atefi M, Pishdad GR, Faghih S. The effects of Canola and Olive oils on insulin resistance, inflammation and oxidative stress in women with type 2 diabetes: a randomized and controlled trial. J Diabetes Metab Disord. 2018;17(2).

  • Raeisi-Dehkordi H, Amiri M, Moghtaderi F, Zimorovat A, Rahmanian M, Mozaffari-Khosravi H et al. Effects of sesame, Canola and sesame-canola oils on body weight and composition in adults with type 2 diabetes mellitus: a randomized, triple-blind, cross-over clinical trial. J Sci Food Agric. 2021;101(14).

  • Burmeister A, Assi LK, Ferro CJ, Hughes RG, Barnett AH, Bellary S, et al. The relationship between high-sensitivity CRP and polyclonal free light chains as markers of inflammation in chronic disease. Int J Lab Hematol. 2014;36(4):415–24.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Halliwell B, Whiteman M. Measuring reactive species and oxidative damage in vivo and in cell culture: how should you do it and what do the results mean? Br J Pharmacol. 2004;142(2):231–55.

    Article 
    CAS 
    PubMed 
    PubMed Central 

    Google Scholar 

  • Periasamy S, Chien SP, Chang PC, Hsu DZ, Liu MY. Sesame oil mitigates nutritional steatohepatitis via Attenuation of oxidative stress and inflammation: A Tale of two-hit hypothesis. J Nutr Biochem. 2014;25(2):232–40.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Burgos-Morón E, Abad-Jiménez Z, de Marañón AM, Iannantuoni F, Escribano-López I, López-Domènech S, et al. Relationship between oxidative stress, ER stress, and inflammation in type 2 diabetes: the battle continues. J Clin Med. 2019;8(9):1385.

    Article 
    PubMed 
    PubMed Central 

    Google Scholar 

  • De Gouveia AV, Cardoso L, De Oliveira CA, Rosa GMM, Moreira G. Effects of the intake of Sesame seeds (Sesamum indicum L.) and derivatives on oxidative stress: A systematic review. J Med Food. 2016;19(4):337–45.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Hsu E, Parthasarathy S. Anti-inflammatory and antioxidant effects of Sesame oil on atherosclerosis: A descriptive literature review. Cureus. 2017;9(7).

  • Rafiee S, Faryabi R, Yargholi A, Zareian MA, Hawkins J, Shivappa N et al. Effects of Sesame consumption on inflammatory biomarkers in humans: A systematic review and meta-analysis of randomized controlled trials. Evidence-based Complementary and Alternative Medicine. 2021;2021.

  • Sankar D, Sambandam G, Ramakrishna Rao M, Pugalendi KV. Modulation of blood pressure, lipid profiles and redox status in hypertensive patients taking different edible oils. Clin Chim Acta. 2005;355(1–2):97–104.

    Article 
    CAS 
    PubMed 

    Google Scholar 

  • Sankar D, Ramakrishna Rao M, Sambandam G, Pugalendi KV. Effect of Sesame oil on diuretics or ß-blockers in the modulation of blood pressure, anthropometry, lipid profile, and redox status. Yale J Biology Med. 2006;79(1):19–26.

    CAS 

    Google Scholar 

  • Khoubnasabjafari M, Ansarin K, Jouyban A. Reliability of malondialdehyde as a biomarker of oxidative stress in psychological disorders. BioImpacts. 2015;5(3).

  • Scott JL, Vijayakumar A, Woodside JV, Neville CE. Feasibility of wearable camera use to improve the accuracy of dietary assessment among adults. J Nutr Sci. 2022;11.

  • Illner AK, Freisling H, Boeing H, Huybrechts I, Crispim SP, Slimani N. Review and evaluation of innovative technologies for measuring diet in nutritional epidemiology. Int J Epidemiol. 2012;41.

Continue Reading

  • A novel presurgical risk prediction model for chronic post-surgical pa

    A novel presurgical risk prediction model for chronic post-surgical pa

    Introduction

    Chronic post-surgical pain (CPSP) is known as a debilitating disease that significantly reduces quality of life and carries substantial biopsychosocial and economic consequences for both patients and society. Furthermore, low return-to-work rates and increased school absenteeism further contribute to the high socio-economic burden of chronic pain.1 Given that over 230 million people undergo surgery globally each year, with CPSP developing in 10% of surgical procedures and reaching up to 85% in certain outliers,2,3 a vast potential for CPSP is represented.4,5 Additionally, the number of surgical procedures is expected to expand with increasing obesity, inflammatory diseases and increased life expectancy. After recognizing the problem of CPSP in the 1990s, the definition underwent several modifications in recent years and many different definitions of CPSP are maintained in the literature.6–8 Recently, the definition of CPSP became more standardized after inclusion in the International Classification of Disease (ICD-11). However, since this ICD-11 definition is relatively new, its implementation is still in progress.3 The clinical success of (preventive) CPSP management remains unfortunately often unsatisfactory with persistent pain complaints and accompanying anxiety and depressive symptoms.9 In the last two decades, research on risk factors in the development of CPSP has grown significantly.2,5 Nevertheless, this has not yet led to improved postsurgical patient outcomes.9 A presurgical CPSP prediction model, suitable for daily use across a large surgical population, which has the potential to allocate high risk patients to the appropriate type of care, is urgently needed.

    Up to now, different models have been developed estimating postsurgical acute and chronic pain.10–13 However, current predictive models for CPSP often face limitations due to a narrow selection of surgical procedures, which restricts their generalizability. They also struggle with incorporating parameters from the postoperative period and rely on data that are challenging to collect in routine practice. Furthermore, many models lack robust validation, reducing their clinical utility in identifying high-risk patients across diverse surgical contexts. Additionally, the increasing adoption of the International Association for the Study of Pain (IASP) definition of CPSP raises concerns about the clinical use in cases where patients experience ongoing pain after surgery, either due to pre-existing complex pain unrelated to surgery, mixed pain conditions or minimal reductions in pain intensity post-surgically, reflecting even more its complexity.13

    Despite the complex biopsychosocial interplay of chronic pain, various surgery– and patient-related risk factors consistently emerge in the likelihood of CPSP occurrence.9 Yet, many of these factors are not easily modifiable, and others are too labor-intensive to assess effectively in a typical preoperative clinical setting. However, estimating probability of CPSP occurrence using a generalizable preoperative model could not only improve patient care and surgical outcomes by facilitating early pain management but also guide further research in the treatment of CPSP such as evaluation of pharmacological strategies in identified high-risk individuals. Moreover, CPSP prediction can have economic benefits by enhanced recovery post-surgery with early pain management and a more fluent reintegration including return to work.

    Therefore, the aim of this study was to develop a presurgical CPSP risk prediction model with good discriminative power, clinical applicability, and possible generalization to a broad group of adults undergoing different types of surgery.

    Materials and Methods

    Participants

    After approval by the Ethics Committee (BUN B3002022000112, September 2022), this single center observational pragmatic pilot study, called PERISCOPE trial, was conducted at the Antwerp University Hospital (UZA), Belgium, in accordance with the Helsinki Declaration and GCP guidelines. The protocol, including the design, of this observational pragmatic pilot study (ClinicalTrials.gov NCT05526976) has been published previously.14 Between December 2022 and September 2023, 660 Dutch-speaking adults scheduled for any type of surgery were recruited preoperatively at the tertiary Antwerp University Hospital. A written informed consent was provided by all participants prior to participation. Patients were excluded if one of the following was present: age below 18 years, not able to complete questionnaires, (diagnostic) procedures without scheduled intervention (such as bronchoscopy, hysteroscopy, gastroscopy, and colonoscopy), or informed consent refusal. Included patients received an analgesic regimen prescribed for postoperative pain by the attending anesthesiologist according to the surgery-specific anesthesia guidelines as performed in our center.

    Study Design, Data Collection and Outcome Variable

    During the study duration, there were no deviations from the standard of care nor additional interventions were executed. This study followed the Transparent Reporting of a multivariable prediction model for Individual Prognosis or Diagnosis (TRIPOD) guidelines for prediction model development.15,16

    Patient‐reported data acquired at screening visit, post-surgery day 1, month 1 and month 3 were collected electronically (REDCAP®, Research Electronic Data Capture, Version 13.6.1, Vanderbilt University, Nashville, Tennessee, USA). During the screening visit, the socio-demographic characteristics (age, sex, level of education and BMI) of all participants, as well as their medical history, preoperative analgesic usage and surgical details were recorded and verified through the electronic medical record. Additionally, participants were instructed in the pain assessment that would be conducted throughout the study trajectory. Participants were asked to electronically complete the following patient-reported outcome measurements (PROMs) at three different timepoints (before surgery, 1 month and 3 months after surgery): surgical-site pain intensity (11-level numeric rating scale (NRS)),17 health-related quality of life (EQ-5D-5L),18 patient experienced level of depression and anxiety (Hospital Anxiety and Depression Scale (HADS) and Spielberger’s Trait Anxiety Inventory (STAI-trait).19–21 Herewith, experienced concerns about the surgery were assessed considering its prevalence in daily practice and suggested opportunity for future intervention.9,21–23 When, pain intensity was scored above two on the 11-level NRS, pain assessment was extended with a screening for neuropathic pain characteristics (Douleur Neuropathique questionnaire (questionnaire part of the DN4)24 and a validated self-report of pain impact on life (Multidimensional Pain Inventory (MPI) part 125). Additionally, a modified version of Kalkman and Althaus index, which previously showed good predictive properties, was assessed presurgical.26,27

    Acute postoperative pain intensity (NRS) on post-surgery day 1 was registered as an additional timepoint. A condensed version of the study design is illustrated in Figure 1. To minimize follow-up non-compliance, up to five reminders were sent via Email and telephone. During follow-up contacts changes in medication, diagnosed surgical complications and visits to the general practitioner/psychologist/surgeon/pain physician were logged.

    Figure 1 Study design. Perioperative patients flow with indicated times when questionnaires were asked to be filled in.

    The objective of this study was to develop a presurgical CPSP risk prediction model useful for clinical practice, with a generalizability to a variety of surgical procedures. The outcome parameter of interest for CPSP was defined as the pain intensity localized at the surgical field of ≥3 on NRS, three months post-surgery.

    Candidate Predictors

    Based on clinical knowledge and a review of the literature, we identified 33 candidate predictors (comprehensive list available in Appendix 1). Within this predictors group, we considered sociodemographic characteristics including sex, age, BMI and educational level (low (no secondary education)/intermediate (secondary education)/high (higher education)), presurgical analgesic consumption (Yes/No, opioids and antineuropathics), surgical procedure and the above mentioned PROMs (modified version of Kalkman and Althaus, NRS, EQ-5D-5L, HADS, STAI).18,19,26–28

    The 44 different executed surgical procedures across 11 disciplines were categorized by 3 independent pain physicians into six categories according to the Kalkman classification: Ophthalmology, Laparoscopy, Ear-nose throat (ENT) surgery, Orthopedic surgery, Laparotomy and Other surgeries.26 Furthermore, the procedures were classified into the following categories: small procedures with high risk, large procedures with high risk, and other, as outlined in the protocol.14 Additionally, these surgical procedures were divided into a categorization using 7 categories (Surgical-7 categorization) as an alternative to the Kalkman classification.

    Sample Size Calculation

    A logistic regression model has been developed to predict the probability of CPSP 3 months post-surgery. The estimated probability from this model was then used to construct a ROC curve (receiver operating characteristic) to discriminate between CPSP and non-CPSP and determine the optimal cut-off risk value regarding maximal sensitivity and specificity.

    The sample size calculation was based on constructing a 95% confidence interval for the area under the ROC curve (AUC). A width of 0.2 and assuming an AUC of 0.7 showed the need for at least a group of 56 CPSP-patients.29 Based on the available scientific evidence at the time of study design, considering a possible mixed CPSP incidence of 10%, we needed a minimum inclusion of 560 patients scheduled for surgery. Taking subject withdrawal, incomplete data or lost to follow-up into account, a total of 660 patients were recruited.

    Statistical Analysis

    Numeric variables are summarized with mean and standard deviation or median and interquartile range and categorical variables with observed frequency and percentage. An initial logistic regression model was fitted with Kalkman score and modified Althaus as predictors and CPSP as outcome. Given a multivariable model with all predictors was not possible (only 80 cases), a stepwise forward approach was followed to build the prediction model. Starting with univariable models evaluating all 33 candidate predictors, the predictor resulting in the best model in terms of highest AUC and significance was then retained. In the next step, multivariable models with 2 predictors are considered, keeping in each model the best univariable candidate and adding one of the other candidate predictors. Overlapping candidates were left out (eg, different instances of pain, different instances or surgery). These steps are repeated for multivariable models with 3 (keeping the 2 best candidates from the previous model fixed and adding one extra candidate) and 4 predictors leading to a final model. The ROC curve of the final model is then compared with the ROC curve of the initial model using a DeLong’s test for correlated ROC curves. Bootstrap techniques were used to evaluate the model’s performance in similar future patients. Random bootstrap samples were drawn with replacement (100 replications) from the data set consisting of all patients who filled in the Kalkman and modified Althaus questionnaire preoperatively and the NRS score at month 3 (n = 415). Forward selection of the candidates was repeated within each bootstrap sample. This allowed us to adjust the estimated model performance and regression coefficients for overoptimism or overfitting. A calibration plot was constructed to examine the agreement between the predicted probabilities and the observed frequencies and calibration measures (Expected/Observed) E/O ratio, calibration slope and calibration in the large (CITL) are reported.

    A complete case analysis on n = 415 patients was used per considered model (if a patient has missings in a variable not included in the model, the patient contributed to the considered model) (Figure 2). No single candidate predictor had missing values >5%. As the outcome was missing in 16% of 496 patients, multiple imputation was considered with 50 imputed datasets and model selection and bootstrap validation were repeated on the imputed datasets.

    Figure 2 Flowchart of sample cohort for analysis.

    All analyses were performed with the statistical software R version 4.3.1. except the bootstrap validation of the final model which was done in Stata version 18.5. Multiple imputation was done with the R-packages MICE and psfmi was used for model estimation, pooling and validation after imputation.

    Results

    Patient Demographics and Characteristics

    In this pilot study, 660 patients were included. Of the 660 recruited subjects, 164 were excluded from analysis due to preoperative factors (24.8%). Of the 496 subjects having preoperative data, 81 were excluded due to postoperative factors (16.3%). Figure 2 summarizes the study sample cohort for analysis. Table 1 provides descriptive statistics for the 415 patients at the screening visit. These 415 patients underwent 44 different operations in 11 disciplines. Three months post-surgery 19.3% of the surveyed subjects reported a pain NRS score ≥3 at the surgical site (Figure 3).

    Table 1 Descriptive Statistics at Screening Visit

    Figure 3 Overview of the distribution of pain intensity (NRS) at the surgical site area three months post-surgery.

    Development of a New Predictive Model

    The initial model with Kalkman score (p < 0.0001) and Althaus risk index (p = 0.074) as predictors for CPSP, results in an AUC of 0.72 (95% CI [0.66,0.78]). From the univariable models with each of the 33 predictors, several predictors are significant where highest AUCs are obtained for the preoperative pain questions (NRS q1 (p < 0.001), q2 (p < 0.001) and Kalkman preoperative pain (p < 0.001)), respectively, 0.76, 0.77 and 0.76). Next, the 30 predictors (leaving out NRS q2 and Kalkman preoperative pain as they are also pain variables) were now added to the model with NRS q1. This model with two predictors led to the highest AUC of 0.80 when including Kalkman surgery (p = 0.001) besides NRS q1. As the category Ophthalmology is very small the Kalkman surgery is from now on recategorized into five categories with Ophthalmology and Other as one category. In the next step evaluating a model with three predictors including the concern question “I am worried about the procedure” (p = 0.032) is retained with an AUC of 0.81. Finally, a model with four predictors adding Education (p = 0.047) as fourth variable gives an AUC of 0.81 (95% CI [0.76,0.87]). Comparing the final model with the four predictors NRS q1, surgery, concern question and education to the initial model with Kalkman score and Althaus risk index gives a p-value of 0.0003 (DeLong’s test) showing a significant improvement (higher AUC) of the final model to this initial model. Together, these four questions form the Persistent Post-surgical Pain Prediction (P4)-Prevoque™ questionnaire (Table 2).

    Table 2 Final Model for Presurgical CPSP Prediction: PrevoqueTM Questionnaire

    Bootstrap was used to adjust for overfitting and the AUC of the final model was 0.76 (overoptimism 0.05). The odds (adjusted for overfitting) on CPSP were 27% higher when the pre-operative pain score goes up 1 unit on the 11-level scale. ENT surgery has the smallest odds on CPSP, and abdominal surgery has more than 8 times higher odds on CPSP compared to ENT, other and ophthalmic surgery 6 times higher odds compared to ENT, orthopedic surgery more than 4 times and laparoscopic surgery 2.5 times higher compared to ENT. The odds on CPSP were 19% higher when the answer on “I worry about the operation” goes up with one unit. The patients with low education level have an almost 3 times higher odds on CPSP compared to intermediate and high education level. The predicted probabilities using the regression coefficients adjusted for overfitting in the final model were then calculated. According to the Youden index the ideal cutoff on these predicted probabilities is 23.9% resulting in a sensitivity of 69.7% and specificity of 82.0%. Using the closest in the top left corner method, a cutoff of 20.58% is chosen resulting in a sensitivity of 73.7% and a specificity of 77.0%. Figure 4 displays the calibration plot of the observed outcomes versus the predicted outcomes with the performance measures E/O ratio, calibration slope and calibration in the large where we see a slight tendency that estimates are a bit too high for individuals at high risk, and too low for those at low risk but overall calibration measures are fair.

    Figure 4 Calibration plot for validation of the proposed prediction model.

    Pooling and selecting the logistic regression models of the 50 imputed datasets revealed a similar model with the same 4 predictors. Internal validation across the imputed datasets with bootstrapping resulted in an optimism corrected AUC of 0.76 (95% CI [0.67,0.82]). Optimism corrections were larger for calibration results.

    Discussion

    This study presents the development of a presurgical CPSP prediction model for adults undergoing a wide range of surgical procedures. Prediction models are being developed to help healthcare providers estimate the likelihood of a particular event occurring so that they can adjust their decisions accordingly.30 So far, various models have been created in recent years to predict postsurgical pain. However, to date, no generalizable CPSP risk stratification model independent for type of surgery is extensively applied. Our multivariable developed model, P4-Prevoque™ questionnaire, can presurgical classify adults undergoing a scheduled surgical procedure, forecasting an individual likelihood of CPSP based on four pre-operative patients’ characteristics: preoperatively pain score at the surgical area (0 to 10 on NRS) [1], the type of surgery (in 5 categories) [2], education level (in 3 levels) [3] and concerns reported about the planned surgical procedure (in 6 levels) [4]. Model performance is good in terms of discriminative power and calibration meaning the model is a useful tool for detecting CPSP.31

    In our single center study cohort, 19.3% of the included patients reported a pain intensity of more than 2 to 10, three months after surgery. Although this finding falls within the broad spectrum of reported CPSP incidence, this average may still be considered comparatively high.3,5,9,32,33 Type of surgery is a known contributing factor to the substantial incidence variation.9 Moreover, this high mean incidence in our study cohort may be indicative of the tertiary hospital surgical procedures and population. Additionally, the inconsistent application of CPSP-definitions may serve as a confounding factor. Since the ICD-11 definition is relatively recent and implementation is ongoing, it is important to note that many different definitions are still maintained in the literature as described by Glare et al.34 In a supplementary analysis, the most recent definition of CPSP as outlined in the ICD-11 defining CPSP as an increase in NRS score of 1 or more at 3 months post-surgery compared to preoperative values was considered for the subjects who had an increase in pain intensity at month 3 compared to pre-surgery.3 This resulted in a group of 72 CPSP patients according to ICD-11 definition. Of those 72 patients, 29 had CPSP both according to the ICD-11 definition and the primary outcome variable. Furthermore, a second predictive model analysis for this subsequent outcome variable was executed. This included presurgical pain intensity at the surgical area (11-level NRS), type of surgery (Surgery-7 classification) and STAI trait and is as such similar to the proposed primary model. Despite the necessity for uniformity in definition, we acknowledge the concerns raised in the recent publication by Papadomanolakis-Pakis et al regarding the ICD-11 definition. Specifically, patients with stable or reduced pain levels are not classified as CPSP by the ICD-11 definition, highlighting the need for pain assessment in educated patients or clinical confirmation in the diagnosis of CPSP.13 Furthermore, patients exhibiting the maximum indicated pain score preoperatively are unable to report a higher score on the NRS post-surgery, thereby rendering them ineligible for the diagnosis of CPSP under this definition. This CPSP definition, in response to the need for a scientifically rigorous and practically applicable framework, is likely to evolve over the next few years. It may also incorporate considerations regarding the impact of analgesic use and changes in pain type within its diagnostic criteria.

    Presurgical pain is found to be a strong predictor in our proposed model. This finding is in line with literature that the presence of persistent nociceptive stimulation may cause pain physiology changes leading to a sensitized nervous system.35,36 Also, the predictive role of surgical type is in accordance with previous literature as surgical tissue trauma, surgery duration and neuronal damage are important contributing factors.9 Furthermore, a recent meta-analysis by Giusti evaluated psychosocial predictors for CPSP and concluded that a heterogeneous group of psychological predictors are significantly associated with CPSP.22 In this study, concerns about the surgery, and anxiety and depression as identified psychosocial predictors were assessed. Worrying about the planned surgical procedure as a single question answer appears to be more predictive than anxiety or depressive states in this study cohort.

    As mentioned, during recent years, a handful of prognostic models have been developed on postoperative pain intensity.10,13,32,37 Two existing prediction models identified as potentially useful in daily practice were incorporated in this research and compared with our prediction model.26,27 However, the Althaus risk index was specifically developed to predict pain 6 months post-surgery and included post-surgical acute pain as a predictor (although a version of the model without this variable is also presented in their publication).27 Similarly, the Kalkman score focused on the presence of severe postoperative pain within the first hour following surgery as the outcome.26 Consequently, these models differ in their outcomes and are therefore not entirely comparable. Notwithstanding, the modified version of Kalkman and Althaus risk index (without the post-surgical acute pain item) was assessed preoperatively, and logistic model evaluation resulted in an AUC of 0.72. Comparatively, the developed prediction model resulted in an AUC of 0.81. Both ROC curves were then compared with a DeLong’s test for correlated ROC curves leading to a p-value of 0.0003 showing the significant improvement of the AUC of our model. Our findings suggests that both the Kalkman score and the Althaus index are effective presurgical prediction models for CPSP, although they were not used in this study for the intended outcome or at the appropriate timepoint. However, the P4-Prevoque™ model, as designed, demonstrates greater accuracy and significantly enhanced predictive power compared to the two previously mentioned models.

    A comparison with even more recently developed prediction models11,12,38 is not feasible due to substantial differences in the study populations and outcome variables including varying interpretations and applications of CPSP definition, timing and the selection of predictors. Beyond the similarities and differences between the P4-Prevoque™ model and the few existing alternatives, the proposed P4-Prevoque™ model distinguishes itself by its inclusive nature, enhancing its generalizability. Our objective was to design a risk model for CPSP that is both clinically relevant and suitable for widespread implementation, by reducing the number of questionnaires and categorizing the responses in a manner conducive to preoperative screening visits, telephone consultations, and digital preoperative care pathways. The P4-Prevoque™ questionnaire facilitates a rapid assessment of the risk for developing CPSP or transitioning to a more severe pain condition. It offers multiple applications for vulnerable patients scheduled for surgery, enhancing patient allocation in research trials, informing tailored management strategies, and ultimately improving their comprehensive postoperative outcomes.

    The potential benefit in reducing the incidence of CPSP using a prediction model, to date, still remains unclear.9–13 Nevertheless, CPSP as a complex biopsychosocial phenomenon with an often challenging treatment approach could benefit from an early, presurgical patient-centered care.9,22,34,39 Early identification of individuals at risk for developing CPSP is essential to ensure prompt assignment to the appropriate care. Subsequently, it will have to be investigated whether early non-pharmacological and pharmacological approaches in at-risk subjects planned for various types of surgery could result in CPSP incidence reduction.9,40,41

    Thereafter, decision analysis methods can be used to assess whether a prediction model should be used in practice by incorporating and quantifying its clinical impact, considering the anticipated benefits, risks, and costs. Furthermore, this study focuses on CPSP three months after surgery. Between three and six months postoperatively, pain complaints may fluctuate in terms of prevalence, intensity, and clinical relevance. As a result, prediction models targeting outcomes at three and six months may differ. Yet, by targeting the three-month outcome, we aim to identify patients early, when there is a meaningful opportunity for intervention.

    In addition to the described strengths, our study has several limitations. First, only 415 out of 660 patients completed questionnaires, resulting in a considerable amount of missing data. Comparisons between completers and non-completers on sex, age, BMI, education and surgery type only showed a significant difference in education level. Thirty percent of non-completers had a high education level compared to 45% of completers. This is an important group missing in the analysis, already known for previously identified patient-related risk factors. Besides that, the small sample size also prohibited a backward selection procedure in the model building step. Another potential limitation is the probability of misclassification of the endpoint CPSP. Similar to others, in this pragmatic study, postoperative in-person follow-up visits were not conducted.11,13 Participants underwent remote pain assessments, after education during the screening visit. Diagnosed surgical complications and readmissions were verified via telephone and cross-checked with the medical records. However, the lack of a physical examination to thoroughly assess the characteristics of CPSP might be point of discussion as reported in recent research.33 Finally, no detailed assessment of the presurgical pain complaints was performed. This could affect the excitability of a nervous system such as in preexisting nociplastic pain syndromes as described by Fitzcharles et al.42

    In recent years, more research has addressed machine learning (ML) models. Langford and colleagues43 reviewed the use of ML to predict postoperative pain and opioid use, highlighting the growing potential of these methods to improve early risk identification. They emphasized the need for rigorous methodological standards and validation to ensure clinical applicability. These findings support the relevance of our approach in developing a robust and interpretable CPSP prediction model.

    Conclusion

    In conclusion, using the designed model, the occurrence of CPSP can be presurgical estimated in adults scheduled for surgery with a sensitivity of 74% and specificity of 77% in the studied population. The P4-Prevoque™ model, composed of four questions, can be easily obtained and has the potential to seamlessly integrate into preoperative workflows through digital tools such as online forms and mobile apps, as well as during in-person visits via kiosks in waiting areas or at healthcare providers’ offices, supporting both modern and traditional care approaches. Future research should prioritize the external validation of the prediction model using an independent dataset, its evaluation in non-university hospital surgical settings, and subsequently its implementation and valorization. If CPSP-at risk subjects can be identified early, preventive pharmacological and non-pharmacological antinociceptive interventions may be reconsidered. Given the relative immutability of surgery type and educational level, we argue that research and prevention efforts should concentrate not only on pain but also on the psychological aspects, such as patient fear, anxiety, and concerns about the surgical procedure. Following prediction model validation, it is important to evaluate its impact on patient-reported outcome measures and patient-reported experience measures. Ultimately, it remains to be determined whether and which interventions targeting high-risk individuals will lead to a reduction in the burden of CPSP.

    Data Sharing Statement

    Requests for (de-identified) raw data used in this clinical trial can be directed to the corresponding author.

    Acknowledgments

    This research has been conducted with screening at the preoperative screening anesthesiology department. We would like to thank the staff, especially Dr H Vandervelde, for their contributions.

    Author Contributions

    All authors made a significant contribution to the work reported, whether that is in the conception, study design, execution, acquisition of data, analysis and interpretation, or in all these areas; took part in drafting, revising or critically reviewing the article; gave final approval of the version to be published; have agreed on the journal to which the article has been submitted; and agree to be accountable for all aspects of the work.

    Funding

    No funding was obtained for this research project.

    Disclosure

    The authors declare that they have no conflicts of interest in this work.

    References

    1. Turk DC, Wilson HD, Cahana A. Pain 2 treatment of chronic non-cancer pain. Lancet. 2011;377(9784):2226–2235. doi:10.1016/S0140-6736(11)60402-9

    2. Fletcher D, Stamer UM, Pogatzki-Zahn E, et al. Chronic postsurgical pain in Europe: an observational study. Eur J Anaesthesiol. 2015;32(10):725–734. doi:10.1097/EJA.0000000000000319

    3. Schug SA, Lavand’Homme P, Barke A, Korwisi B, Rief W, Treede RD. The IASP classification of chronic pain for ICD-11: chronic postsurgical or posttraumatic pain. Pain. 2019;160(1):45–52. doi:10.1097/j.pain.0000000000001413

    4. Weiser TG, Haynes AB, Molina G, et al. Surgical services: access and coverage estimate of the global volume of surgery in 2012: an assessment supporting improved health outcomes. Available from: www.thelancet.com. Accessed July 1, 2025.

    5. Kehlet H, Jensen TS, Woolf CJ. Persistent postsurgical pain: risk factors and prevention. Lancet. 2006;367(9522):1618–1625. doi:10.1016/S0140-6736(06)68700-X

    6. Macrae WA. Chronic post-surgical pain: 10 years on. Br J Anaesth. 2008;101(1):77–86. doi:10.1093/bja/aen099

    7. Macrae WA. Chronic pain after surgery. Br J Anaesth. 2001;87(1):88–98. doi:10.1093/bja/87.1.88

    8. Werner MU, Kongsgaard UE. Defining persistent post-surgical pain: is an update required? Br J Anaesth. 2014;113(1):1–4. doi:10.1093/bja/aeu012

    9. Rosenberger DC, Pogatzki-Zahn EM. Chronic post-surgical pain – update on incidence, risk factors and preventive treatment options. BJA Educ. 2022;22(5):190–196. doi:10.1016/j.bjae.2021.11.008

    10. Papadomanolakis-Pakis N, Uhrbrand P, Haroutounian S, Nikolajsen L. Prognostic prediction models for chronic postsurgical pain in adults: a systematic review. Pain. 2021;162(11):2644–2657. doi:10.1097/j.pain.0000000000002261

    11. van Driel MEC, van Dijk JFM, Baart SJ, Meissner W, Huygen FJPM, Rijsdijk M. Development and validation of a multivariable prediction model for early prediction of chronic postsurgical pain in adults: a prospective cohort study. Br J Anaesth. 2022;129(3):407–415. doi:10.1016/j.bja.2022.04.030

    12. Sluka KA, Wager TD, Sutherland SP, et al. Predicting chronic postsurgical pain: current evidence and a novel program to develop predictive biomarker signatures. Pain. 2023;164(9):1912–1926. doi:10.1097/j.pain.0000000000002938

    13. Papadomanolakis-Pakis N, Haroutounian S, Sørensen JK, et al. Development and internal validation of a clinical risk tool to predict chronic postsurgical pain in adults: a prospective multicentre cohort study. Pain. 2022. doi:10.1097/j.pain.0000000000003405

    14. Wildemeersch D, Meeus I, Wauters E, et al. Evaluating the predictive value of a short preoperative holistic risk factor screening questionnaire in preventing persistent pain in elective adult surgery: study protocol for a prospective observational pragmatic trial [PERISCOPE]. J Pain Res. 2023;16:4281–4287. doi:10.2147/JPR.S439824

    15. Collins GS, Reitsma JB, Altman DG, Moons KGM. Transparent reporting of a multivariable prediction model for individual prognosis or diagnosis (TRIPOD): the TRIPOD statement. BMC Med. 2015;13(1):1. doi:10.1186/s12916-014-0241-z

    16. Collins GS, Moons KGM, Dhiman P, et al. TRIPOD+AI statement: updated guidance for reporting clinical prediction models that use regression or machine learning methods. BMJ. 2024. doi:10.1136/bmj-2023-078378

    17. Downie WW, Leatham PA, Rhind VM, Wright V, Branco JA, Anderson JA. Studies with pain rating scales. Ann Rheum Dis. 1978;37(4):378–381. doi:10.1136/ard.37.4.378

    18. Devlin N, Pickard S, Busschbach J. The development of the EQ-5D-5L and its value sets. In: Value Sets for EQ-5D-5L. Springer International Publishing; 2022:1–12. doi:10.1007/978-3-030-89289-0_1

    19. Zigmond AS, Snaith RP. The hospital anxiety and depression scale. Acta Psychiatr Scand. 1983;67(6):361–370. doi:10.1111/j.1600-0447.1983.tb09716.x

    20. Marteau TM, Bekker H. The development of a six‐item short‐form of the state scale of the Spielberger State—Trait Anxiety Inventory (STAI). Br J Clin Psychol. 1992;31(3):301–306. doi:10.1111/j.2044-8260.1992.tb00997.x

    21. Moerman N, van Dam FSAM, Muller MJ, Oosting H. The Amsterdam Preoperative Anxiety and Information Scale (APAIS). Anesth Analg. 1996;82(3):445–451. doi:10.1097/00000539-199603000-00002

    22. Giusti EM, Lacerenza M, Manzoni GM, Castelnuovo G. Psychological and psychosocial predictors of chronic postsurgical pain: a systematic review and meta-analysis. Pain. 2021;162(1):10–30. doi:10.1097/j.pain.0000000000001999

    23. Weinrib AZ, Azam MA, Birnie KA, Burns LC, Clarke H, Katz J. The psychology of chronic post-surgical pain: new frontiers in risk factor identification, prevention and management. Br J Pain. 2017;11(4):169–177. doi:10.1177/2049463717720636

    24. Bouhassira D, Attal N, Alchaar H, et al. Comparison of pain syndromes associated with nervous or somatic lesions and development of a new neuropathic pain diagnostic questionnaire (DN4). Pain. 2005;114(1):29–36. doi:10.1016/j.pain.2004.12.010

    25. Lousberg R, Van Breukelen GJP, Groenman NH, Schmidt AJM, Arntz A, Winter FAM. Psychometric properties of the multidimensional pain inventory, Dutch language version (MPI-DLV). Behav Res Ther. 1999;37(2):167–182. doi:10.1016/S0005-7967(98)00137-5

    26. Kalkman CJ, Visser K, Moen J, Bonsel GJ, Grobbee DE, Moons KGM. Preoperative prediction of severe postoperative pain. Pain. 2003;105(3):415–423. doi:10.1016/S0304-3959(03)00252-5

    27. Althaus A, Hinrichs-Rocker A, Chapman R, et al. Development of a risk index for the prediction of chronic post-surgical pain. European J Pain. 2012;16(6):901–910. doi:10.1002/j.1532-2149.2011.00090.x

    28. van der Bij AK, de Weerd S, Cikot RJLM, Steegers EAP, Braspenning JCC. Validation of the Dutch short form of the state scale of the Spielberger State-trait anxiety inventory: considerations for usage in screening outcomes. Public Health Genomics. 2003;6(2):84–87. doi:10.1159/000073003

    29. Hajian-Tilaki K. Sample size estimation in diagnostic test studies of biomedical informatics. J Biomed Inform. 2014;48:193–204. doi:10.1016/j.jbi.2014.02.013

    30. Moons KGM, Royston P, Vergouwe Y, Grobbee DE, Altman DG. Prognosis and prognostic research: what, why, and how? BMJ. 2009;338(feb23 1):b375–b375. doi:10.1136/bmj.b375

    31. de Hond AAH, Steyerberg EW, van Calster B. Interpreting area under the receiver operating characteristic curve. Lancet Digit Health. 2022;4(12):e853–e855. doi:10.1016/S2589-7500(22)00188-1

    32. Fletcher D, Lavand’homme P. Towards better predictive models of chronic post-surgical pain: fitting to the dynamic nature of the pain itself. Br J Anaesth. 2022;129(3):281–284. doi:10.1016/j.bja.2022.06.010

    33. Martinez V, Lehman T, Lavand’homme P, et al. Chronic postsurgical pain A European survey. Eur J Anaesthesiol. 2024;41(5):351–362. doi:10.1097/EJA.0000000000001974

    34. Glare P, Aubrey KR, Myles PS. Transition from acute to chronic pain after surgery. Lancet. 2019;393(10180):1537–1546. doi:10.1016/S0140-6736(19)30352-6

    35. Nijs J, George SZ, Clauw DJ, et al. Central sensitisation in chronic pain conditions: latest discoveries and their potential for precision medicine. Lancet Rheumatol. 2021;3(5):e383–e392. doi:10.1016/S2665-9913(21)00032-1

    36. Feizerfan A, Sheh G. Transition from acute to chronic pain. Continuing Educ Anaesth Crit Care Pain. 2015;15(2):98–102. doi:10.1093/bjaceaccp/mku044

    37. Papadomanolakis-Pakis N, Haroutounian S, Christiansen CF, Nikolajsen L. Prediction of chronic postsurgical pain in adults: a protocol for multivariable prediction model development. BMJ Open. 2021;11(12):e053618. doi:10.1136/bmjopen-2021-053618

    38. Montes A, Roca G, Cantillo J, Sabate S. Presurgical risk model for chronic postsurgical pain based on 6 clinical predictors: a prospective external validation. Pain. 2020;161(11):2611–2618. doi:10.1097/j.pain.0000000000001945

    39. Katz J, Weinrib A, Fashler S, et al. The Toronto General Hospital Transitional Pain Service: development and implementation of a multidisciplinary program to prevent chronic postsurgical pain. J Pain Res. 2015;695. doi:10.2147/JPR.S91924

    40. Carley ME, Chaparro LE, Choinière M, et al. Pharmacotherapy for the prevention of chronic pain after surgery in adults: an updated systematic review and meta-analysis. Anesthesiology. 2021;135(2):304–325. doi:10.1097/ALN.0000000000003837

    41. Chaparro LE, Smith SA, Moore RA, Wiffen PJ, Gilron I. Pharmacotherapy for the prevention of chronic pain after surgery in adults. Cochrane Database Syst Rev. 2013;2021(6). doi:10.1002/14651858.CD008307.pub2

    42. Fitzcharles MA, Cohen SP, Clauw DJ, Littlejohn G, Usui C, Häuser W. Chronic pain 2 nociplastic pain: towards an understanding of prevalent pain conditions. Lancet. 2021;397(10289):2098–2110. doi:10.1016/S0140-6736(21)00392-5

    43. Langford DJ, Reichel JF, Zhong H, et al. Machine learning research methods to predict postoperative pain and opioid use: a narrative review. Reg Anesth Pain Med. 2025;50(2):102–109. doi:10.1136/rapm-2024-105603

    Continue Reading

  • Better Artificial Intelligence (AI) Stock: SoundHound AI vs. C3.ai

    Better Artificial Intelligence (AI) Stock: SoundHound AI vs. C3.ai

    • SoundHound AI and C3.ai are pure-play artificial intelligence (AI) software companies with massive opportunities ahead.

    • SoundHound AI stock is more richly valued than C3.ai, but may have a greater runway for growth ahead.

    • Choosing between the two stocks may ultimately boil down to the risk tolerance levels of investors.

    • 10 stocks we like better than SoundHound AI ›

    The adoption of artificial intelligence (AI) software is increasing at an incredible pace because of the productivity and efficiency gains this technology is capable of delivering, and the good part is that this niche is likely to sustain a healthy growth rate over the long run.

    According to ABI Research, the AI software market is expected to clock a compound annual growth rate (CAGR) of 25% through 2030, generating $467 billion in annual revenue at the end of the decade. That’s why it would be a good time to take a closer look at the prospects of SoundHound AI (NASDAQ: SOUN) and C3.ai (NYSE: AI) — two pure-play AI companies that could help investors capitalize on a couple of fast-growing niches within the AI software market — and check which one of them is worth buying right now.

    Image source: Getty Images.

    SoundHound AI provides a voice AI platform where its customers can create conversational AI assistants and voice-based AI agents that can be deployed for multiple uses, such as taking orders in restaurants, car infotainment systems, and customer service applications, among others.

    This particular market is growing at a nice clip, as deploying AI-powered voice solutions can help companies improve productivity and efficiency, since they will be able to automate tasks. Companies can now significantly improve their customer interaction experiences, thanks to the availability of round-the-clock multilingual AI agents and assistants.

    Not surprisingly, SoundHound AI has been witnessing a robust growth in demand for its voice AI solutions, which explains the solid revenue growth in the past year.

    SOUN Revenue (TTM) Chart

    SOUN Revenue (TTM) data by YCharts.

    But here’s what investors should look forward to: The conversational AI market could grow at an annual average rate of almost 24% through 2030, generating over $41 billion in annual revenue by the end of the decade. SoundHound AI has been growing at a much faster pace than the overall market, suggesting it is gaining a bigger share of this lucrative space.

    SoundHound’s revenue guidance of $167 million at the mid-point for 2025, is nearly double the revenue it reported last year. Importantly, its cumulative subscriptions and bookings backlog stood at a massive $1.2 billion last year. This metric is a measure of the potential revenue that the company expects to “realize over the coming several years,” suggesting it can maintain its healthy growth rates for a long time to come thanks to the AI-fueled opportunity it’s sitting on.

    C3.ai is a pure-play enterprise AI software platform provider that enables its customers to build generative AI applications and agentic AI solutions. The company claims that it provides 130 comprehensive enterprise AI applications ready for deployment across industries such as oil and gas, manufacturing, financial services, utilities, chemicals, defense, and others.

    It has been in the news of late for receiving a bigger contract worth $450 million from the U.S. Air Force for maintaining aircraft, ground assets, and weapons systems for the next four years. However, this is just one of the many contracts that the company has been landing lately.

    C3.ai’s offerings are used across diverse industries, and its customer base includes the likes of Baker Hughes, which recently expanded its partnership with the company; local and state government bodies across multiple U.S. states; and companies such as Ericsson, Bristol Myers Squibb, Chanel, and others. The company’s fast-expanding customer base and the bigger contracts that it is signing with existing customers explain why there has been an uptick in C3.ai’s growth of late.

    AI Revenue (TTM) Chart

    AI Revenue (TTM) data by YCharts.

    The company finished fiscal 2025 (which ended on April 30) with a 25% increase in its revenue to $389 million. Management expects another 20% increase in total revenue in fiscal 2025. Consensus estimates suggest that C3.ai is likely to report similar growth next year, followed by an acceleration in fiscal 2028.

    AI Revenue Estimates for Current Fiscal Year Chart

    AI Revenue Estimates for Current Fiscal Year data by YCharts.

    There’s a strong possibility, however, that C3.ai will exceed expectations and its own forecast for growth this year. That’s because C3.ai ended the previous fiscal year with 174 pilot projects, which it calls initial production deployments. The good part is that the company has been converting its pilots into contracts at a healthy rate.

    C3.ai turned 66 of its initial production deployments into long-term contracts in fiscal 2025. The company ended fiscal 2024 with 123 pilot projects, which means that it has a conversion rate of more than 50%. So the robust increase in the company’s pilot projects last year means that it could close more such initial production deployments into full agreements in the current fiscal year, going by past trends.

    So there is a strong possibility of C3.ai’s growth rate exceeding Wall Street’s expectations, which should ideally turn out to be a tailwind for its stock price in the long run.

    While it is clear both SoundHound and C3.ai are growing at a nice pace because of AI, the former’s growth rate is much higher. However, to buy SoundHound stock, investors will have to pay a handsome price-to-sales ratio of nearly 38. C3.ai, on the other hand, is trading at a much more attractive 8 times sales, which is almost in line with the U.S. technology sector’s average sales multiple.

    So, investors looking for a mix of steady growth and attractive valuation can consider buying shares of C3.ai. However, if you have a higher appetite for risk and are willing to pay for a stock with a richer valuation, then consider buying SoundHound AI, as its faster growth could help it clock more upside, though the expensive valuation also exposes it to more volatility.

    Before you buy stock in SoundHound AI, consider this:

    The Motley Fool Stock Advisor analyst team just identified what they believe are the 10 best stocks for investors to buy now… and SoundHound AI wasn’t one of them. The 10 stocks that made the cut could produce monster returns in the coming years.

    Consider when Netflix made this list on December 17, 2004… if you invested $1,000 at the time of our recommendation, you’d have $699,558!* Or when Nvidia made this list on April 15, 2005… if you invested $1,000 at the time of our recommendation, you’d have $976,677!*

    Now, it’s worth noting Stock Advisor’s total average return is 1,060% — a market-crushing outperformance compared to 180% for the S&P 500. Don’t miss out on the latest top 10 list, available when you join Stock Advisor.

    See the 10 stocks »

    *Stock Advisor returns as of June 30, 2025

    Harsh Chauhan has no position in any of the stocks mentioned. The Motley Fool has positions in and recommends Bristol Myers Squibb. The Motley Fool recommends C3.ai. The Motley Fool has a disclosure policy.

    Better Artificial Intelligence (AI) Stock: SoundHound AI vs. C3.ai was originally published by The Motley Fool

    Continue Reading