Pharmacometabolomics Identifies Candidate Predictor Metabolites of an L-carnitine Treatment Mortality Benefit in Septic Shock ============================================================================================================================= * Michael A. Puskarich * Theodore S. Jennaro * Christopher E. Gillies * Charles R. Evans * Alla Karnovsky * Cora E. McHugh * Thomas L. Flott * Alan E. Jones * Kathleen A. Stringer * On behalf of the RACE Trial Investigators ## Abstract **Background** Sepsis-induced metabolic dysfunction contributes to organ failure and death. L-carnitine has shown promise for septic shock, but a recent study demonstrated a non-significant reduction in mortality. **Methods** A pharmacometabolomics study of patients (n=250) in a Phase II trial of L-carnitine to identify metabolic profiles predictive of a 90-day mortality benefit from L-carnitine. The independent predictive value of each pre-treatment metabolite concentration, adjusted for L-carnitine dose, on 90-day mortality was determined by logistic regression. A grid-search analysis maximizing the Z-statistic from a binomial proportion test identified specific metabolite threshold levels that discriminated L-carnitine responsive patients. Threshold concentrations were further assessed by hazard-ratio and Kaplan-Meier estimate. **Findings** Accounting for L-carnitine treatment and dose, 11 1H-NMR metabolites and 12 acylcarnitines were independent predictors of 90-day mortality. Based on the grid-search analysis numerous acylcarnitines and valine were identified as candidate metabolites of drug response. Acetylcarnitine emerged as highly viable for the prediction of an L-carnitine mortality benefit due to its abundance and biological relevance. Using its most statistically significant threshold concentration, patients with acetylcarnitine ≥35µM were less likely to die at 90 days if treated with L-carnitine (18 g) versus placebo (p=0.01 by log rank test). **Interpretation** Metabolomics identified independent predictors of 90-day sepsis mortality. Our proof-of-concept approach shows how pharmacometabolomics may be useful for tackling the heterogeneity of sepsis and informing clinical trial design. Also, metabolomics can help understand mechanisms of sepsis heterogeneity and variable drug response, since sepsis induces alterations in numerous metabolite concentrations. Key words * sepsis * metabolomics * acylcarnitines * branched chain amino acids * nuclear magnetic resonance * liquid chromatography-mass spectroscopy ## Introduction Sepsis represents the leading cause of death in the intensive care unit and the single most expensive inpatient diagnosis, representing more than $17 billion in healthcare costs annually in the United States.1-3 Septic shock carries a particularly poor prognosis, with short-term mortality rates of approximately 40%. Among the many physiologic disturbances associated with sepsis is a profound shift in metabolism.4 Hyperlactatemia represents one of the hallmarks of sepsis and is now considered a criterion for the diagnosis of septic shock.5 However, hyperglycemia, lipolysis, and protein catabolism are also common and similarly associated with increased mortality.4,6 Manipulation of these processes represents an underdeveloped but promising target for novel pharmacotherapies. Despite the concerning sepsis mortality statistics and an increasingly focused research effort on the condition, clinical trials of novel sepsis pharmacotherapies have traditionally yielded disappointing results. While the causes of the failure of clinical trials to further novel treatments are multifactorial, the highly heterogeneous nature of sepsis certainly contributes to these results.7,8 This highlights the need to forge a better understanding of the heterogeneity and complexity of the clinical illness by identifying sepsis endotypes.9 In doing so, strategies for enriched patient selection could be used to improve the precision of clinical trials. Importantly, predictive and prognostic enrichment strategies for clinical trials have been advocated by many and have been issued as guidance by regulatory agencies like the U.S. Food and Drug Administration.10-12 We recently completed a phase II, Bayesian adaptive dose-finding randomized control trial comparing L-carnitine (6, 12, or 18 g) treatment to saline (placebo) for the early treatment of septic shock. None of the tested doses of L-carnitine resulted in a significant reduction in sequential organ failure assessment (SOFA) score at 48 hours, though the highest and best performing dose (18 g) demonstrated a non-significant 3% and 6% absolute mortality reduction at 28 days in the intention to treat and per protocol analyses compared to saline placebo, respectively. In parallel with the planning of the original trial, we designed an ancillary metabolomics study, the L-Carnitine Pharmacometabolomics in Sepsis (CaPS) study, to identify candidate metabolites of drug response that could serve to endotype a heterogeneous septic shock cohort and direct the design of a clinical enrichment strategy for a phase III trial. A number of studies have demonstrated the importance of energy-related metabolites for the differentiation of sepsis survivors and the identification of sepsis endotypes,4,6,13-16 most of which are readily detected by nuclear magnetic resonance (NMR) spectroscopy6,14,15 and targeted liquid chromatography – mass spectroscopy (LC-MS) assays.16 Furthermore, we have previously demonstrated the utility of metabolomics in predicting drug response (pharmacometabolomics) in sepsis15 employing relatively quantified NMR metabolites and acylcarnitines generated by an LC-MS assay. With this background in mind, we hypothesized that serum concentrations of acylcarnitines and/or other metabolites could differentiate patients that disproportionately benefit from L-carnitine treatment as measured by mortality. ## Methods ### Study design This study utilized pre-treatment serum samples collected from 236 of the 250 patients enrolled in the Rapid Administration of Carnitine (RACE) in Sepsis clinical trial.17 The parent trial was approved by each site’s institutional review board, all patients or their surrogate gave written informed consent, and it was registered at [clinicaltrials.gov](http://clinicaltrials.gov) prior to initiation (NCT 01665092). Details of the blood samples included in the study are provided in the supplementary material, Figure 1. Serum samples were assayed for acylcarnitines by LC-MS16 and by quantitative proton (1H) nuclear magnetic resonance (NMR) as previously described.18,19 More details about the methods for these measurements can be found in the supplementary material. ![Figure 1:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/31/2021.01.28.21250687/F1.medium.gif) [Figure 1:](http://medrxiv.org/content/early/2021/01/31/2021.01.28.21250687/F1) Figure 1: Pre-treatment acetylcarnitine (C2) concentration as a predictive clinical trial enrichment strategy. We present four scenarios to illustrate how different threshold concentrations of acetylcarnitine (C2), a high abundant acylcarnitine, would have impacted the outcome of the Rapid Administration of Carnitine (RACE) in Sepsis clinical trial in patients treated with either L-carnitine (18 g) or placebo. In scenario 1, no threshold concentration is used so the entire RACE cohort (n=236) is eligible. The sample size of 170 patients represents those that received either L-carnitine (18 g; n=100) or placebo (n=70). The hazard ratio is not significant, and consistent with the parent trial, the Kaplan-Meier curve shows no mortality benefit of L-carnitine (p=0·57). In scenario 2, an acetylcarnitine (C2) threshold concentration of ≥21µM is used. Forty-four percent (n=104) of the RACE cohort met this criterion and of these, 68 patients received either L-carnitine (18 g) or placebo. The hazard ratio is not improved, and the Kaplan-Meier curve shows no mortality benefit of L-carnitine (p=0·59). In scenario 3, an acetylcarnitine (C2) threshold concentration of ≥30µM is used. Twenty-seven percent (n=64) of the RACE cohort met this criterion and of these, 42 patients received either L-carnitine (18 g) or placebo. The hazard ratio is significant and favors L-carnitine (18 g); the Kaplan-Meier curve shows a mortality benefit of L-carnitine (p=0·04). Finally, scenario 4 uses the acetylcarnitine (C2) concentration associated with the maximum Z-statistic (see Table 4 in the supplementary material), ≥35µM. Twenty-three percent (n=54) of the RACE cohort met this criterion and of these, 37 patients received either L-carnitine (18 g) or placebo. The hazard ratio is significant, and the Kaplan-Meier curve shows a mortality benefit of L-carnitine (p=0·01). Kaplan-Meier curves were interpreted using log-rank (Mantel-Cox) tests; hazard ratios were calculated using Mantel-Haenszel. ### Outcomes We elected to use mortality as the outcome of our analysis because the primary end point of the RACE trial (reduction in SOFA score at 48 h) was not met, but the 18 g dose of L-carnitine resulted in a trend towards a reduction in mortality. Based on data suggesting a substantial continued decline in mortality among sepsis patients beyond 28 days and preliminary data from our phase I data suggesting continued benefit from L-carnitine treatment on longer term mortality rates,20 we elected to assess the cumulative distribution mortality function to find the optimal time frame for assessment of mortality (28, 90, 180, or 365 days). By 90-days, ∼90% of the deaths had occurred, (see Figure 2 in the supplementary material), based on this analysis, we chose 90-day mortality as the primary clinical outcome. View this table: [Table 1:](http://medrxiv.org/content/early/2021/01/31/2021.01.28.21250687/T1) Table 1: Demographics and clinical characteristics of the cohort, stratified by 90-day mortality View this table: [Table 2.](http://medrxiv.org/content/early/2021/01/31/2021.01.28.21250687/T2) Table 2. Logistic regression model for the prediction of 90-day mortality adjusted for treatment (L-carnitine dose or placebo) View this table: [Table 3.](http://medrxiv.org/content/early/2021/01/31/2021.01.28.21250687/T3) Table 3. Logistic regression interaction model testing the relationship between metabolite predictors and mortality by treatment (L-carnitine dose or placebo) for the prediction of 90-day mortality ranked by ascending p value up to 0·05 View this table: [Table 4.](http://medrxiv.org/content/early/2021/01/31/2021.01.28.21250687/T4) Table 4. Significant metabolites# from logistic regression interaction model ranked by descending maximum Z-statistic ![Figure 2:](http://medrxiv.org/https://www.medrxiv.org/content/medrxiv/early/2021/01/31/2021.01.28.21250687/F2.medium.gif) [Figure 2:](http://medrxiv.org/content/early/2021/01/31/2021.01.28.21250687/F2) Figure 2: A clinical trial enrichment strategy could optimize clinical trial design for heterogeneous critical illnesses like sepsis. An example of a scheme for a hypothetical phase III clinical trial of supplement L-carnitine for the treatment of septic shock that uses an *a priori* determined acetylcarnitine (C2) threshold concentration to determine whether a patient is enrolled and randomized to receive either L-carnitine (18 g) or placebo. ### Statistical analyses Descriptive data are reported as means and standard deviations, medians with interquartile ranges, or proportions as appropriate. Differences in categorical outcomes were compared using chi-square tests, while Student t-tests and Wilcoxon rank sum tests were used to compare continuous variables. The aims of our primary analyses were to: 1) determine the relationship between individual metabolites and 90-day mortality; 2) determine if the relationship between a predictive metabolite and mortality depends on treatment allocation; and 3) using metabolites most associated with mortality, determine the optimal (threshold) metabolite level that could be used to identify patients with septic shock most likely to respond favorably to L-carnitine treatment. Collectively, and similar to other secondary analyses or ancillary studies of clinical and observational trials,21-25 achievement of these goals would provide clinical proof of concept of a metabolically informed strategy to tackle the heterogeneity of sepsis that also could be used for a predictive enrichment design of a phase III study.10,11 Since metabolomics data are on different scales due to varying abundance, in preparation for statistical analyses, data were natural-log transformed and Z-score normalized to have a mean of 0 and a standard deviation of 1.26,27 We began our analysis using partial least squares-discriminant analysis (PLS-DA)28 to visualize the overall metabolic heterogeneity of the study participants and determine whether there were metabolic differences between sexes and the treatment groups. We followed PLS-DA by an assessment of the predictive value of individual metabolites on 90-day mortality. To accomplish this, we constructed a series of logistic regression models and adjusted for treatment assignment.29 We then tested if the relationship between metabolite predictors and mortality varied across treatment groups using a logistic regression interaction model. Age and SOFA score were considered as covariates in further multivariable modelling. To test the potential clinical application of our pharmacometabolomics approach, after identifying metabolites strongly related to 90-day mortality that also had a significant interaction with treatment allocation, we aimed to identify the specific concentration or levels of these candidate metabolites that could be used to predict which patients would be most likely to derive a mortality benefit from L-carnitine. To achieve this, we used a grid-search methodology to compute the Z-statistic from the binomial proportion test at every possible threshold metabolite concentration or level.30 For this example, since the 18g dose of L-carnitine was the most efficacious in the RACE trial and would be the one most likely to be tested in a phase III trial, we used the Z-statistic to quantify the standardized difference in the proportion of deaths between those patients who received L-carnitine (18 g) and those who received placebo. For this analysis, the metabolite level at each threshold was used as the criterion for inclusion into the proportion test. We then computed a two-sample (binomial) proportion test31 which compared the proportion of patients treated with L-carnitine who died by 90 days to those that were treated with placebo. This permitted the identification of metabolite levels associated with a range of Z-statistics including the maximum Z-statistic and their corresponding p values. The Z-statistic simultaneously accounts for the difference in the proportion of patients who died in the treatment versus placebo groups and the sizes of each group, thereby suggesting the most optimal metabolite threshold level. Metabolites were then ranked by descending maximum Z-statistic. Similar approaches have been used by other studies that have sought to identify the responder population in clinical and observational trials.21-25 To further illustrate the implications of the use of different metabolite concentrations as predictors of mortality, hazard ratios were calculated using the Mantel-Haenszel method and Kaplan-Meier curves were constructed (log-rank [Mantel-Cox] test). Metabolite concentration cut points were selected according to different trial scenarios and the grid-search analysis described above. All statistical tests except for hazard ratios (Mantel-Haenszel) and log-rank (Mantel-Cox) tests which were done using PRISM, were performed in R studio (R version 3.6.2 (2019-12-12) Copyright 2019 The R Foundation for Statistical Computing) and figures were constructed in R and PRISM (version 8.4.3, June 10, 2020). ## Results Of the 250 participants randomized in the parent trial, 1H-NMR metabolomics and acylcarnitine data were available from 228 and 236 patient serum samples, respectively (see Figure 1 in the supplementary material). We identified and quantified 27 serum metabolites by 1H-NMR and 24 acylcarnitines by LC-MS (see Table 1 in the supplementary material). Representative 1H-NMR and LC spectra are shown in Figures 3 and 4 in the supplementary material. All-cause 90-day mortality was 124/236 (52·5%), while 28-day and 1-year mortality were 104/236 (44·1%) and 136/236 (57·6%), respectively. Clinical and demographic variables of the cohort stratified by the primary outcome are summarized in **Table 1**. As expected, patients who died were older and had a higher SOFA score. The PLS-DA plots of the acylcarnitine data and the NMR metabolites by treatment category (supplementary Figure 5A and B) and sex (supplementary Figure 6A and B) illustrate the metabolic heterogeneity of the study cohort and do not demonstrate any significant metabolic distinction between these groups. We then conducted multivariate logistic regression using L-carnitine dose and metabolites as covariates (base model) and applied a conservative Bonferroni correction for multiple comparisons. The base model identified in 11/27 1H-NMR metabolites and 12/24 acylcarnitines that significantly discriminated 90-day mortality (**Table 2;** the complete list can be found in Table 2 of the supplementary material). We then tested whether the relationship between predictive metabolites and mortality depends on treatment allocation. This was done with the addition of an interaction term between L-carnitine dose and metabolite level (interaction model) which reduced the number of significant metabolites from 23 to 14, of which all but three metabolites were acylcarnitines (**Table 3**; a comprehensive list can be found in Table 3 in the supplementary material); these were not in range to withstand a conservative adjustment (e.g., Bonferroni) for multiple comparisons. In this analysis, a statistically significant and negative interaction term indicates that the predicted probability of 90-day mortality for a given metabolic feature is lower at higher doses of L-carnitine. To determine whether the signals found in the base and interaction models was merely due to factors associated with the risk of death, we controlled for both age32 and SOFA score33. Several acylcarnitines and choline tolerated this adjustment (see Table 4 in the supplementary material for the full list of metabolites); notably, lactate was not significant in either model (p=0·96 and p=0·22, respectively). As these findings were not evident in the parent clinical trial and they suggest that there may be a sepsis endotype that may derive a therapeutic benefit from supplement L-carnitine, we hypothesized that a pharmacometabolomics approach may aid in defining this sub-group of patients. To identify the candidate metabolites, we took a hypothesis-generating approach and considered all metabolites with significant (≤ 0·05) unadjusted p values (n=14 in the logistic regression interaction model; **Table 3**) and assessed the Z-statistic of each. Based on this analysis, the metabolites with significant (FDR-corrected p values ≤ 0·05) maximum Z-statistics included a number of acylcarnitines as well as the branched chain amino acid, valine (**Table 4**; also see supplementary Table 5). In addition to the Z-statistic values, to identify candidate metabolites, we also considered the prevalence of the acylcarnitine signal, the known potential of acetylcarnitine (C2) to predict drug responsiveness15 and its close metabolic relationship with L-carnitine. Furthermore, the maximum Z-statistic of C12 and C8:1 represented a lower percentage of the clinical cohort than either C5 or acetylcarnitine (C2). As such, we selected acetylcarnitine (C2) as the most viable metabolite candidate to demonstrate the utility of our pharmacometabolomics approach. As examples, we assessed several concentrations of both acetylcarnitine (C2) and valine, including the ones at the maximum Z-statistic, 35 µM (p=0·002; as measured by LC-MS) (**Figure 1**) and 88µM (p=0·009), respectively **(**also see Figure 7 and Table 5 in the supplementary material). These analyses illustration how a pharmacometabolomics may aid in the design of a precision trial of L-carnitine for the treatment of septic shock using the scheme as illustrated in **Figure 2**. ## Discussion Our pharmacometabolomics study, CaPS, aimed to identify pre-treatment, sepsis-induced metabolic derangements in survivors and non-survivors treated with L-carnitine. We found that there are likely metabolically distinct groups (endotypes) of patients that do proportionally better when they receive an 18 g dose of supplemental L-carnitine. These findings imply that a precision, clinical trial enrichment strategy using pharmacometabolomics could help combat the heterogeneity of sepsis and drug response, which is known to have contributed to numerous negative clinical studies.7 Here we show that a pharmacometabolomics approach identified clinically indistinguishable sepsis endotypes are more likely to derive a mortality benefit from treatment with L-carnitine (18 g), a finding not evident in the metabolically naive parent trial. To accomplish this, we used a metabolomics analysis to capture high abundant polar compounds (quantitative 1H-NMR) and acylcarnitines (LC-MS) in serum samples collected from patients enrolled in a phase II clinical trial of L-carnitine therapy.17 Using this approach, similar to our prior study16, we found a prevalent acylcarnitine signal. From this profile, we selected acetylcarnitine (C2) and valine to illustrate how different threshold concentrations could influence mortality outcome in patients randomized to either placebo of L-carnitine (18 g). Specifically, patients with higher (e.g., ≥ 30µM) acetylcarnitine (C2) levels at enrollment may be more likely to derive a treatment benefit as defined by decreased intermediate term (90-day) mortality; this benefit is maximized at acetylcarnitine (C2) concentrations ≥ 35µM. While severity of illness could contribute to this finding, clinical variables alone do not seem to account for the identification of the drug-responsive endotype since the finding is retained when accounting for factors associated with the risk of death (age and SOFA score, see supplementary Table 4). Notably, we also found that serum concentrations of the branched chain amino acid, valine, could also be used to identify a mortality benefit of L-carnitine but not to as great an extent as acetylcarnitine (C2). Collectively, these data suggest that there are patients that are in clinically occult subgroups. Should these data be validated, metabolically informed clinical trial design35 and ultimately, precision treatment strategies, could represent a new paradigm of sepsis care. These data provide the groundwork and rationale for a pharmacometabolomics directed clinical trial to test L-carnitine therapy efficacy for septic shock using a specific concentration of a key metabolite (e.g., acetylcarnitine (C2)) to guide inclusion criteria (**Figure 2**). Importantly, the current study also shows that numerous metabolites may have predictive value for sepsis mortality, even after controlling for factors associated with the risk of death (see Table 4 in the supplementary material). These data provide further evidence that sepsis induces broad metabolic disruption that is linked to patient outcomes, corroborating prior studies.36,37 Of note, numerous acylcarnitines, including unsaturated acylcarnitines, predicted mortality, suggesting significant disruption in fatty acid metabolic pathways.37 Overall, the broad range in disruption of acylcarnitines may reflect differential and variable mobilization of fatty acids,38 rather than disruption of a specific enzyme or pathway. We have previously demonstrated this in a smaller cohort of septic shock patients.16 Despite this variance, acetylcarnitine (C2) was the most robust predictor of overall sepsis mortality. This corroborates a previous study that identified acetylcarnitine (C2) as being associated with the severity of sepsis-induced organ dysfunction, inflammation and infection.36 Acetylcarnitine (C2) also happens to be one of only two compounds (with L-carnitine) detected by both the LC-MS and NMR analytical platforms; regardless of the detection method, it performed similarly in the regression models. Interestingly, acetylcarnitine (C2) outperformed the more clinically ubiquitous lactate level in predicting sepsis mortality. After correcting for age and SOFA score, lactate was not a significant independent predictor (supplementary Table 4) whereas acetylcarnitine (C2) retained its predictive value following this correction, which suggests the potential for its use as an adjunctive clinical test for risk prognosis. However, as our cohort was highly selected and involved only participants receiving vasopressors (which affect glycolysis and lactate production)39,40 who were already resuscitated, it would be inappropriate to interpret these data to imply that lactate does not serve an important role in the early identification and prognosis patients with suspected infection. In particular, serial lactate levels and its clearance rate have been used to assess the adequacy of resuscitation and lactate is included in the sepsis definition.5,41-43 Nevertheless, limitations of lactate have been recognized41 and, notably, others have demonstrated that acylcarnitines outperform lactate in predicting sepsis mortality.37 Our data suggest that acetylcarnitine (C2) may represent a superior risk stratification tool in a selected cohort of fully resuscitated patients undergoing treatment with vasopressor infusions. We also learned from the CaPS study that pre-treatment serum L-carnitine concentrations did not predict a L-carnitine treatment mortality benefit, suggesting against the hypothesis that serum L-carnitine deficiency drives the response to supplemental L-carnitine in sepsis patients. Rather, in aggregate, these data provide evidence to support the hypothesis that sepsis induces an impairment in the mobilization of acetyl groups. While there may be a number of biologically plausible hypotheses, our findings could be due to sepsis-induced increased intracellular accumulation of acetyl-CoA secondary to its decreased metabolism via the tricarboxylic acid cycle (TCA) or enhanced acetyl-CoA production via fatty acid (beta-oxidation) metabolism (Figure 9 in the supplementary material). Consequently, increases in acetyl-CoA are managed by a number of mechanisms one of which is via the mitochondrial enzyme, carnitine acetyltransferase (EC 2.3.1.7). Carnitine acetyltransferase transfers acetyl groups to carnitine, displacing the hydrogen atom in its hydroxyl group44 converting it to the membrane-permeable, acetylcarnitine (C2) (Figure 9 in the supplementary material). Acetylcarnitine (C2), the shortest of the acylcarnitines, is important because it plays a controlling role over acetyl-CoA on metabolic substrate switching and therefore, enables metabolic flexibility.44 As the need for ATP increases, acetyl-CoA is diverted to the TCA cycle. However, in sepsis, the TCA cycle may fail to metabolize these groups resulting in excess acetyl-CoA and subsequent elevation in measured serum acetylcarnitine (C2) concentrations. The elevation in acetylcarnitine (C2) may reflect the ability of L-carnitine to serve as route for the disposal of excess acetyl groups which has been demonstrated in the myocardium45 and during exercise.46 However, unlike acetylcarnitine (C2), the metabolic link between L-carnitine therapeutic response and BCAA concentrations is less clear. We and others have shown that levels of branched chain amino acids (BCAA) influence sepsis outcome15,37 and shock resolution.47 It is possible that patients with elevated BCAA blood concentrations represent those with a metabolic reserve that enables them to more efficiently utilize supplemental L-carnitine48 but in general, the mechanisms of BCAA signaling and metabolic mechanisms of action are poorly understood.49 In aggregate, our findings suggest that the magnitude of sepsis-induced disruptions in energy metabolism may be associated with a therapeutic benefit of L-carnitine. This relationship and the mechanisms that underlie it warrant further interrogation. Despite the encouraging results of our study, we acknowledge that there are several important weaknesses. We recognize that “real-time” metabolomics is not feasible in clinical practice and that routine measurement of these compounds, including acetylcarnitine (C2), for routine clinical use is not currently available. We also employed a limited, focused metabolomic approach, measuring high abundant polar compounds (1H-NMR) and acylcarnitines. We acknowledge that a broad, untargeted approach may have yielded additional compounds predictive of outcomes or treatment response. With our targeted approach, we still made multiple comparisons testing involving over 50 metabolites in this study, which opens the door to false positive findings. Our findings persisted after application of a conservative Bonferroni correction, but we acknowledge that the predictive capacity of acetylcarnitine (C2) and valine, when accounting for interactions between baseline metabolite and treatment assignment (interaction model), was not amenable to correction for multiple comparisons. As such, and given that this was an ancillary study, we acknowledge that any conclusions regarding the accurate prediction of clinical drug responsiveness are only hypothesis generating and will require rigorous prospective testing. We did, however, show how the use of a number of different acetylcarnitine (C2) and valine concentrations would influence the mortality outcome of the RACE trial (**Figure 1** and supplementary Figure 7). These were merely used as examples to illustrate the utility of a pharmacometabolomics approach and despite including almost 250 patients, we acknowledge that our results may overestimate the true effect size and will require validation in an external cohort. Nevertheless, even though these subgroups represent ≤ 50% of the total RACE trial cohort, they highlight the value of a predictive enrichment strategy that could be used to design a phase III clinical trial of L-carnitine supplementation for septic shock. Importantly, the pharmacometabolomics approach was developed concurrent with the design of the parent trial, and the conceptual model was based on and is consistent with our preliminary work in a unique, though smaller cohort,15 strengthening the validity of the findings. In summary, an ancillary pharmacometabolomics study, CaPS, of the parent clinical trial, RACE, found numerous predictors, independent of intervention, age and SOFA score, for 90-day mortality in septic shock including many acylcarnitines and other metabolites such as tyrosine, betaine, lysine and glycine. We also demonstrate the translational value of the work by showing how the application of a pharmacometabolomics-based clinical trial enrichment strategy, using pre-treatment acetylcarnitine (C2) concentrations as an example, could be used to identify the responder population, a sepsis endotype, that may derive a mortality benefit from L-carnitine supplementation. This represents a unique clinical trial enrichment strategy that could be employed to improve the efficiency of a phase III L-carnitine efficacy study in patients with septic shock9 and other emerging therapeutics in heterogeneous critical illnesses. These findings also support the notion that distinct metabolic endotypes contribute to sepsis heterogeneity. ## Supporting information supplement [[supplements/250687_file02.pdf]](pending:yes) ## Data Availability The metabolomics data sets and R code will be made publicly available at the time of publication of the peer-reviewed manuscript. [https://doi.org/10.21228/M8VX0Z](https://doi.org/10.21228/M8VX0Z) ## Data sharing The metabolomics data sets and R code will be made publicly available at the time of publication of the peer-reviewed manuscript. ## Contributors Authors’ contributions to the study: Study concept and design: MAP, AEJ, KAS; oversight of metabolomics data generation: KAS; LC-MS acylcarnitines data generation: CRE; generation of quantitative NMR metabolomics data: CEM, TLF; analysis data and statistical analyses: MAP, CEG, TSJ, KAS; data interpretation: MAP, CEG, TSJ, AK, KAS; data verification: MAP, TSJ, AEJ, KAS; drafting of the manuscript and its revision for important intellectual content: MAP, CEG, TSJ, CRE, AK, CEM, TLF, AEJ, KAS; final approval of the version submitted for publication: MAP, CEG, TSJ, CRE, AK, CEM, TLF, AEJ, KAS ## Declaration of interests * Competing interests: The author(s) declare(s) that they have no competing interests. No authors or their institutions received any payments or services in the past 36 months from a third party that could be perceived to influence or give the appearance of potentially influencing, the submitted work. ## Funding statement * None of the authors or their institutions at any time received payment or services from a third party for any aspect of the submitted work. * This study was supported by the National Institute of General Medical Sciences (NIGMS) via R01GM103799 (AEJ), K23GM113041 (MAP) and R01GM111400 (KAS). CEG’s and TSJ’s contributions were supported, in part, by the Michigan Institute for Data Science “Propelling Original Data Science” grant from the University of Michigan; TSJ also received support from the American Foundation of Pharmaceutical Education. The content is solely the responsibility of the authors and does not necessarily represent the official views of NIGMS or the NIH. ## Acknowledgements We acknowledge the contributions of the RACE Trial Investigators group; Nathan I. Shapiro, MD, MPH (Department of Emergency Medicine, Beth Israel Deaconess Medical Center, Boston, MA); Faheem W. Guirgis, MD (Department of Emergency Medicine, University of Florida College of Medicine–Jacksonville, FL); Michael Runyon, MD, MPH (Department of Emergency Medicine, Carolinas Medical Center, Charlotte, NC); Jason Y. Adams, MD (Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, University of California, Davis, CA); Robert Sherwin, MD (Department of Emergency Medicine, Wayne State University, Detroit, MI); Ryan Arnold, MD (Department of Emergency Medicine, Christiana Care Health System, Wilmington, DE); Brian W. Roberts, MD, MSc (Department of Emergency Medicine, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, NJ); Michael C. Kurz, MD, MS (Department of Emergency Medicine, The University of Alabama School of Medicine at Birmingham, Birmingham, AL); Henry E. Wang, MD, MS (Department of Emergency Medicine, The University of Texas Health Science Center at Houston, Houston, TX); Jeffrey A. Kline, MD (Department of Emergency Medicine, Indiana University School of Medicine, Indianapolis, IN); D. Mark Courtney, MD (Department of Emergency Medicine, Northwestern University, Chicago, IL); Stephen Trzeciak, MD, MPH (Department of Medicine, Cooper University Hospital, Cooper Medical School of Rowan University, Camden, NJ); Sarah A. Sterling, MD (Department of Emergency Medicine, The University of Mississippi Medical Center, Jackson, MS); Utsav Nandi, MD (Department of Emergency Medicine, The University of Mississippi Medical Center, Jackson, MS); Deepti Patki, MS (Department of Emergency Medicine, The University of Mississippi Medical Center, Jackson, MS); Kert Viele, PhD (Berry Consultants, Austin, TX). * Received January 28, 2021. * Revision received January 28, 2021. * Accepted January 31, 2021. * © 2021, Posted by Cold Spring Harbor Laboratory This pre-print is available under a Creative Commons License (Attribution-NonCommercial-NoDerivs 4.0 International), CC BY-NC-ND 4.0, as described at [http://creativecommons.org/licenses/by-nc-nd/4.0/](http://creativecommons.org/licenses/by-nc-nd/4.0/) ## References 1. 1.Fleischmann C, Scherag A, Adhikari NKJ, et al. Assessment of Global Incidence and Mortality of Hospital-treated Sepsis. Current Estimates and Limitations. Am J Respir Crit Care Med 2016; 193(3): 259–72. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1164/rccm.201504-0781OC&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26414292&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 2. 2.Gaieski DF, Edwards JM, Kallan MJ, Carr BG. Benchmarking the incidence and mortality of severe sepsis in the United States. Crit Care Med 2013; 41(5): 1167–74. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/CCM.0b013e31827c09f8&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23442987&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000317661000019&link_type=ISI) 3. 3.Rudd KE, Johnson SC, Agesa KM, et al. Global, regional, and national sepsis incidence and mortality, 1990-2017: analysis for the Global Burden of Disease Study. Lancet 2020; 395(10219): 200–11. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(19)32989-7&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 4. 4.Langley RJ, Tipper JL, Bruse S, et al. Integrative “omic” analysis of experimental bacteremia identifies a metabolic signature that distinguishes human sepsis from systemic inflammatory response syndromes. Am J Respir Crit Care Med 2014; 190(4): 445–55. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1164/rccm.201404-0624OC&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=25054455&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 5. 5.Singer M, Deutschman CS, Seymour CW, et al. The Third International Consensus Definitions for Sepsis and Septic Shock (Sepsis-3). JAMA 2016; 315(8): 801–10. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2016.0287&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26903338&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 6. 6.Liu Z, Triba MN, Amathieu R, et al. Nuclear magnetic resonance-based serum metabolomic analysis reveals different disease evolution profiles between septic shock survivors and non-survivors. Crit Care 2019; 23(1): 169. 7. 7.Seymour CW, Kennedy JN, Wang S, et al. Derivation, Validation, and Potential Treatment Implications of Novel Clinical Phenotypes for Sepsis. JAMA 2019; 321(20): 2003–17. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1001/jama.2019.5791&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=http://www.n&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 8. 8.Leligdowicz A, Matthay MA. Heterogeneity in sepsis: new biological evidence with clinical applications. Critical Care 2019; 23(1): 80. 9. 9.NAGMSC Working Group on Sepsis: Final Report. In: National Institute of General Medical Sciences NIH, editor. [https://www.nigms.nih.gov/News/reports/Documents/nagmsc-working-group-on-sepsis-final-report.pdf](https://www.nigms.nih.gov/News/reports/Documents/nagmsc-working-group-on-sepsis-final-report.pdf): NIH; 2019. 10. 10.United States Food and Drug Administration. Enrichment Strategies for Clinical Trials to Support Determination of Effectiveness of Human Drugs and Biological Products: Guidance for Industry. 2019. [https://www.fda.gov/regulatory-information/search-fda-guidance-documents/enrichment-strategies-clinical-trials-support-approval-human-drugs-and-biological-products](https://www.fda.gov/regulatory-information/search-fda-guidance-documents/enrichment-strategies-clinical-trials-support-approval-human-drugs-and-biological-products) (accessed August 26 2020). 11. 11.Prescott HC, Calfee CS, Thompson BT, Angus DC, Liu VX. Toward Smarter Lumping and Smarter Splitting: Rethinking Strategies for Sepsis and Acute Respiratory Distress Syndrome Clinical Trial Design. Am J Respir Crit Care Med 2016; 194(2): 147–55. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1164/rccm.201512-2544CP&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=27244481&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 12. 12.Vincent JL. The coming era of precision medicine for intensive care. Crit Care 2017; 21(Suppl 3): 314. 13. 13.Johansson PI, Nakahira K, Rogers AJ, et al. Plasma mitochondrial DNA and metabolomic alterations in severe critical illness. Crit Care 2018; 22(1): 360. 14. 14.Mickiewicz B, Duggan GE, Winston BW, Doig C, Kubes P, Vogel HJ. Metabolic profiling of serum samples by 1H nuclear magnetic resonance spectroscopy as a potential diagnostic approach for septic shock. Crit Care Med 2014; 42(5): 1140–9. 15. 15.Puskarich MA, Finkel MA, Karnovsky A, et al. Pharmacometabolomics of l-carnitine treatment response phenotypes in patients with septic shock. Ann Am Thorac Soc 2015; 12(1): 46–56. 16. 16.Puskarich MA, Evans CR, Karnovsky A, Das AK, Jones AE, Stringer KA. Septic Shock Nonsurvivors Have Persistently Elevated Acylcarnitines Following Carnitine Supplementation. Shock 2018; 49(4): 412–9. 17. 17.Jones AE, Puskarich MA, Shapiro NI, et al. Effect of Levocarnitine vs Placebo as an Adjunctive Treatment for Septic Shock: The Rapid Administration of Carnitine in Sepsis (RACE) Randomized Clinical Trial. JAMA Netw Open 2018; 1(8): e186076. 18. 18.Labaki WW, Gu T, Murray S, et al. Serum amino acid concentrations and clinical outcomes in smokers: SPIROMICS metabolomics study. Sci Rep 2019; 9(1): 11367. 19. 19.McHugh CE, Flott TL, Schooff CR, et al. Rapid, Reproducible, Quantifiable NMR Metabolomics: Methanol and Methanol: Chloroform Precipitation for Removal of Macromolecules in Serum and Whole Blood. Metabolites 2018; 8(4). 20. 20.Puskarich MA, Kline JA, Krabill V, Claremont H, Jones AE. Preliminary safety and efficacy of L-carnitine infusion for the treatment of vasopressor-dependent septic shock: a randomized control trial. JPEN J Parenter Enteral Nutr 2014; 38(6): 736–43. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1177/0148607113495414&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=23851424&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 21. 21.Anderson BJ, Calfee CS, Liu KD, et al. Plasma sTNFR1 and IL8 for prognostic enrichment in sepsis trials: a prospective cohort study. Crit Care 2019; 23(1): 400. 22. 22.Man M, Close SL, Shaw AD, et al. Beyond single-marker analyses: mining whole genome scans for insights into treatment responses in severe sepsis. Pharmacogenomics J 2013; 13(3): 218–26. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1038/tpj.2012.1&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22310353&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 23. 23.Martucci G, McNally D, Parekh D, et al. Trying to identify who may benefit most from future vitamin D intervention trials: a post hoc analysis from the VITDAL-ICU study excluding the early deaths. Crit Care 2019; 23(1): 200. 24. 24.Stanski NL, Stenson EK, Cvijanovich NZ, et al. PERSEVERE Biomarkers Predict Severe Acute Kidney Injury and Renal Recovery in Pediatric Septic Shock. Am J Respir Crit Care Med 2020; 201(7): 848–55. 25. 25.Li Q, Parikh H, Butterworth MD, et al. Longitudinal Metabolome-Wide Signals Prior to the Appearance of a First Islet Autoantibody in Children Participating in the TEDDY Study. Diabetes 2020; 69(3): 465–76. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiZGlhYmV0ZXMiO3M6NToicmVzaWQiO3M6ODoiNjkvMy80NjUiO3M6NDoiYXRvbSI7czo1MDoiL21lZHJ4aXYvZWFybHkvMjAyMS8wMS8zMS8yMDIxLjAxLjI4LjIxMjUwNjg3LmF0b20iO31zOjg6ImZyYWdtZW50IjtzOjA6IiI7fQ==) 26. 26.Everitt B, Hothorn T, SpringerLink (Online service). An introduction to applied multivariate analysis with R. New York: Springer; 2011. 27. 27.van den Berg RA, Hoefsloot HC, Westerhuis JA, Smilde AK, van der Werf MJ. Centering, scaling, and transformations: improving the biological information content of metabolomics data. BMC Genomics 2006; 7: 142. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1186/1471-2164-7-142&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16762068&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 28. 28.Worley B, Powers R. Multivariate Analysis in Metabolomics. Curr Metabolomics 2013; 1(1): 92–107. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.2174/2213235X11301010092&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=26078916&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 29. 29.Zhang Z, Seibold H, Vettore MV, Song WJ, Francois V. Subgroup identification in clinical trials: an overview of available methods and their implementations with R. Ann Transl Med 2018; 6(7): 122. 30. 30.Lipkovich I, Dmitrienko A, Denne J, Enas G. Subgroup identification based on differential effect search—A recursive partitioning method for establishing response to treatment in patient subpopulations. Statistics in Medicine 2011; 30(21): 2601–21. [PubMed](http://medrxiv.org/lookup/external-ref?access_num=21786278&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 31. 31.Simon R. Clinical trials for predictive medicine. Stat Med 2012; 31(25): 3031–40. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1002/sim.5401&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22714719&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 32. 32.Martin GS, Mannino DM, Moss M. The effect of age on the development and outcome of adult sepsis. Crit Care Med 2006; 34(1): 15–21. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/01.CCM.0000194535.82812.BA&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16374151&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000234381900003&link_type=ISI) 33. 33.Vincent JL, de Mendonca A, Cantraine F, et al. Use of the SOFA score to assess the incidence of organ dysfunction/failure in intensive care units: results of a multicenter, prospective study. Working group on “sepsis-related problems” of the European Society of Intensive Care Medicine. Crit Care Med 1998; 26(11): 1793–800. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/00003246-199811000-00016&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=9824069&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000076999100016&link_type=ISI) 34. 34.Storey JD. A direct approach to false discovery rates. J Roy Stat Soc B 2002; 64: 479– 98. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1111/1467-9868.00346&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=WOS:00017742&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) 35. 35.Vincent J-L, Sakr Y. Clinical trial design for unmet clinical needs: a spotlight on sepsis. Expert Rev Clin Pharmacol 2019; 12(9): 893–900. 36. 36.Chung KP, Chen GY, Chuang TY, et al. Increased Plasma Acetylcarnitine in Sepsis Is Associated With Multiple Organ Dysfunction and Mortality: A Multicenter Cohort Study. Crit Care Med 2019; 47(2): 210–8. 37. 37.Langley RJ, Tsalik EL, van Velkinburgh JC, et al. An integrated clinico-metabolomic model improves prediction of death in sepsis. Sci Transl Med 2013; 5(195): 195ra95. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6MTE6InNjaXRyYW5zbWVkIjtzOjU6InJlc2lkIjtzOjEzOiI1LzE5NS8xOTVyYTk1IjtzOjQ6ImF0b20iO3M6NTA6Ii9tZWRyeGl2L2Vhcmx5LzIwMjEvMDEvMzEvMjAyMS4wMS4yOC4yMTI1MDY4Ny5hdG9tIjt9czo4OiJmcmFnbWVudCI7czowOiIiO30=) 38. 38.Costa CC, de Almeida IT, Jakobs C, Poll-The BT, Duran M. Dynamic changes of plasma acylcarnitine levels induced by fasting and sunflower oil challenge test in children. Pediatr Res 1999; 46(4): 440–4. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1203/00006450-199910000-00013&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=10509365&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000082752600013&link_type=ISI) 39. 39.Annane D, Vignon P, Renault A, et al. Norepinephrine plus dobutamine versus epinephrine alone for management of septic shock: a randomised trial. Lancet 2007; 370(9588): 676–84. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(07)61344-0&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=17720019&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000249131400032&link_type=ISI) 40. 40.Levy B, Gibot S, Franck P, Cravoisy A, Bollaert PE. Relation between muscle Na+K+ ATPase activity and raised lactate concentrations in septic shock: a prospective study. Lancet 2005; 365(9462): 871–5. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/S0140-6736(05)71045-X&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=15752531&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000227444600029&link_type=ISI) 41. 41.Puskarich MA, Shapiro NI, Massey MJ, Kline JA, Jones AE. Lactate Clearance in Septic Shock Is Not a Surrogate for Improved Microcirculatory Flow. Acad Emerg Med 2016; 23(6): 690–3. 42. 42.Arnold RC, Shapiro NI, Jones AE, et al. Multicenter study of early lactate clearance as a determinant of survival in patients with presumed sepsis. Shock 2009; 32(1): 35–9. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1097/SHK.0b013e3181971d47&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=19533847&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000267081400006&link_type=ISI) 43. 43.Jansen TC, van Bommel J, Schoonderbeek FJ, et al. Early lactate-guided therapy in intensive care unit patients: a multicenter, open-label, randomized controlled trial. Am J Respir Crit Care Med 2010; 182(6): 752–61. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1164/rccm.200912-1918OC&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=20463176&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000282162100008&link_type=ISI) 44. 44.Muoio DM, Noland RC, Kovalik JP, et al. Muscle-specific deletion of carnitine acetyltransferase compromises glucose tolerance and metabolic flexibility. Cell Metab 2012; 15(5): 764–77. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1016/j.cmet.2012.04.005&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=22560225&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000303694100023&link_type=ISI) 45. 45.Schroeder MA, Atherton HJ, Dodd MS, et al. The cycling of acetyl-coenzyme A through acetylcarnitine buffers cardiac substrate supply: a hyperpolarized 13C magnetic resonance study. Circ Cardiovasc Imaging 2012; 5(2): 201–9. [Abstract/FREE Full Text](http://medrxiv.org/lookup/ijlink/YTozOntzOjQ6InBhdGgiO3M6MTQ6Ii9sb29rdXAvaWpsaW5rIjtzOjU6InF1ZXJ5IjthOjQ6e3M6ODoibGlua1R5cGUiO3M6NDoiQUJTVCI7czoxMToiam91cm5hbENvZGUiO3M6ODoiY2lyY2N2aW0iO3M6NToicmVzaWQiO3M6NzoiNS8yLzIwMSI7czo0OiJhdG9tIjtzOjUwOiIvbWVkcnhpdi9lYXJseS8yMDIxLzAxLzMxLzIwMjEuMDEuMjguMjEyNTA2ODcuYXRvbSI7fXM6ODoiZnJhZ21lbnQiO3M6MDoiIjt9) 46. 46.Kiens B. Skeletal muscle lipid metabolism in exercise and insulin resistance. Physiol Rev 2006; 86(1): 205–43. [CrossRef](http://medrxiv.org/lookup/external-ref?access_num=10.1152/physrev.00023.2004&link_type=DOI) [PubMed](http://medrxiv.org/lookup/external-ref?access_num=16371598&link_type=MED&atom=%2Fmedrxiv%2Fearly%2F2021%2F01%2F31%2F2021.01.28.21250687.atom) [Web of Science](http://medrxiv.org/lookup/external-ref?access_num=000234758500005&link_type=ISI) 47. 47.Puskarich MA, McHugh C, Flott TL, et al. Serum Levels of Branched Chain Amino Acids Predict Duration of Cardiovascular Organ Failure in Septic Shock. Shock 2020. 48. 48.Hiraoka A, Kiguchi D, Ninomiya T, et al. Can L-carnitine supplementation and exercise improve muscle complications in patients with liver cirrhosis who receive branched-chain amino acid supplementation? Eur J Gastroenterol Hepatol 2019; 31(7): 878–84. 49. 49.Bonvini A, Coqueiro AY, Tirapegui J, Calder PC, Rogero MM. Immunomodulatory role of branched-chain amino acids. Nutr Rev 2018; 76(11): 840–56.