Abstract
Alcohol and consequent liver disease are a major burden, which has worsened during the COVID-19 pandemic. There are several facets to the pathophysiology and clinical consequences of alcohol-use disorder (AUD) and progression to alcohol-associated liver disease (ALD), which needs a concerted effort by clinicians, translational and basic science investigators. Several recent advances from bedside to bench and bench to bedside have been made in ALD. We focus this review on a case-based approach that gives a human context to these important questions across the spectrum of ALD.
INTRODUCTION:
Alcohol and consequent liver disease is increasing as a burden for patients, family members and health systems worldwide. With the COVID-19 pandemic, emerging evidence indicates an increase in alcohol consumption and incidence of alcohol-use disorder (AUD), as well as a predictive long-term impact on the incidence of alcohol-associated liver disease (ALD)1. The current world situation thus requires clinicians, translational and basic science investigators to urgently focus on this major disease process. ALD results in a wide range of presentations from pre-cirrhotic disease all the way through alcohol-associated hepatitis (AH). There have been several rapid advances in bench to bedside and from bedside to bench in ALD that focus on the patho-physiology, integration of clinical and mental health care, and liver transplantation. This review is focused on a case-based approach across the spectrum of ALD to provide a human context to these important questions.
CASE 1:
A 46-year-old man is referred to the liver clinic because of high ALT (78 IU). He is obese (BMI 35 kg/m2) with recently diagnosed hyperlipidemia. On questioning, he drinks 2–3 drinks of vodka per night and several more over the weekend. The patient’s AUDIT-10 score today is 14. He does not report any psychosocial consequences of his alcohol use, but his wife has often pointed out his drinking habits to him. His father is estranged but according to his mother had issues with alcohol. He is a Veteran of the Gulf war and was previously diagnosed with PTSD. However, he is not under therapy for this or any other mental health condition currently. A transient elastogram shows F2 fibrosis, which is also confirmed by MR elastography. MRI also shows a steatotic liver.
Can we predict if and when he will develop cirrhosis if these drinking patterns continue?
What is the impact of concomitant obesity and hyperlipidemia on risk of cirrhosis development?
How does the family history and mental health history contribute towards this risk?
What are newer biomarkers and therapies that can be used to modulate this risk?
Do we need a multi-disciplinary approach?
ALD progresses through mild steatosis, inflammation, and fibrosis to cirrhosis (ALC) and an increased risk for hepatocellular carcinoma (HCC). Heavy drinkers can develop alcohol-associated hepatitis (AH) at any stage in the progression of chronic disease. While most heavy drinkers will develop steatosis, progression to later stages of disease occurs only in a subset of heavy drinkers and likely involve interactions with both genetic and environmental factors that either offer protection for disease progression or increased risk. Both sex and genetic ancestry contribute to the risk of AUD and ALD. For example, while men may typically consume more alcohol than women, there is a growing incidence of AUD in women with greater susceptibility to the pathophysiological effects of alcohol2. Similarly, risk for AUD varies with genetic ancestry, with Native Americans at higher risk and Asians with reduced risk, but these risks may be modified by cultural influences3. Genetic ancestry/ethnicity is also a major factor in the onset and severity of ALD with Hispanic patients presenting at earlier ages than White/Caucasian patients, while African-American patients presented with less severe AH4. Development of strong protocols for risk prediction for the progression of liver disease in heavy drinkers is an important unmet clinical and public health need5
Genetic risk for progression of ALD
In addition to the role of sex and race in progression of ALD, family history/genetics can also contribute to risk for heavy drinking, as well as obesity and liver injury, three conditions present in the Veteran in this case study. Many GWAS and whole exome sequencing studies have identified multiple genetic polymorphisms, including PNPLA3, TM6SF2, MBOAT7, GCKR, HSD17B13, that contribute to NAFL/NASH6. However, large population based studies identifying genetic risk for ALD have been more limited7. The PNPLA3 polymorphism is also associated with increased risk of ALC and conveys a higher risk for mortality in patients with severe AH enrolled in the STOPAH trial8. A recent GWAS study with 1128 patients with ALC with Caucasian ancestry identified PNPLA3 and HSD17B13 as risk factors for ALC; conditional analysis, considering the PNPLA3 and HSD17B13 alleles, identified variants in Fas-associated factor family member 2 (FAF2) as protective9. These associations were validated using GWAS data from the UK Biobank9. Importantly, these data were used to develop a genetic risk score for ALC based on a combination of 3 single nucleotide polymorphisms (SNPs) (PNPLA3:rs738409, SUGP1-TM6SF2:rs10401969, HSD17B13:rs6834314) and diabetes status9.
Most of the genetic risk factors for ALD identified to date modify lipid metabolism and lipid droplet formation, consistent with abnormal lipid homeostasis in the development of steatosis. However, how these polymorphisms contribute to later stages of disease progression is not well understood. It is likely that additional genetic variants will target processes that contribute to later stage disease, including inflammation, fibrosis, and HCC. For example, a recent case control GWAS study identified polymorphisms in Wnt family members as a susceptibility locus for alcohol-related HCC10. Interestingly, these data identify germ-line polymorphisms in the Wnt-β-catenin pathway in alcohol-associated HCC10, while other studies also implicate somatic modifications in the Wnt-β-catenin family in HCC development11, 12. Genetic variance in Wnt-β-catenin pathway is consistent with aberrant regulation of cell proliferation and development of HCC10.
Heavy drinking and AUD are also moderately heritable disorders13, 14. Meta-analyses of multiple GWAS studies of AUD found that the most consistently identified SNPs associated with AUD were in genes related to alcohol metabolism15. Interestingly, recent GWAS analysis making use of the Million Veterans Program finds genetic risk for heavy drinking is not sufficient for the development of AUD14. Relatively few studies have compared the genetic risk for AUD to that for ALD16. Future analysis of genetic influences on heavy drinking and liver disease are needed to help identify specific targets for therapeutic intervention, as well as to identify patients at risk.
Environmental factors impacting progression of ALD
Obesity and risk for progression of ALD
Epidemiological data suggests that obesity and metabolic syndrome exacerbate progression of ALD17–19. This is likely related to common pathogenic mechanisms between NAFL/NASH, including increased oxidative stress, aberrant regulation of lipid metabolism and disruption of autophagy and regulation of cell death pathways19. Further, while it is well recognized that obesity and metabolic syndrome are associated with a dysfunctional and inflamed adipose tissue, more recent work finds that alcohol consumption also profoundly disrupts the function of adipose tissue, contributing to the pathogenesis of ALD20, 21. Important for treatment of heavy drinkers, cessation of drinking rapidly normalizes adipose tissue function22. Similarly, weight loss in obese patients reduced adipose inflammation23.
Microbial dysbiosis and impaired barrier function of the gut is another common feature in ALD and other forms of fatty liver. Even a single heavy bout of alcohol consumption impairs the barrier function of the intestine, leading to an increase in circulating concentrations of endotoxin/LPS24. Circulating concentrations of LPS, as well as other microbial products, are increased in patients with ALD21 and non-alcohol associated fatty liver/steatohepatitis (NAFL/NASH)25 and dysbiosis is common in both diseases25.
Importantly, as in our Case#1, it is increasingly common for patients to present with both obesity and heavy drinking18, 19, 26, likely related to the increased prevalence of obesity in the general population. The patient in our case study is typical of veterans, as more than 7 in 10 veterans who receive VA care are obese or overweight, putting them at high risk for the development of complications of metabolic syndrome, including type 2 diabetes and liver disease27. Common genetic polymorphisms associated with increased risk for NAFL/NASH and ALD also indicate common pathophysiological mechanisms18, 19. Since obesity is a risk factor for the development of diabetes, the modifying role of diabetes in genetic risk for ALC9 also points to common pathogenesis and the importance for clinicians to treat both obesity and AUD.
Is there a distinction between the pathogenesis of lean ALD vs obese ALD that could be leveraged into development of biomarkers and therapeutics? Little data is available on this important question. At this time, pre-clinical studies are in progress which may eventually impact clinical practice for the diagnosis and treatment of patients with ALD with concomitant obesity. One recent study found that the chemokine CXCL11 could distinguish patients with AH who were lean compared to obese28. Interestingly, biomarkers related to pathways of cell death can distinguish ALD from NAFL/NASH29, 30. Hepatocellular death occurs via multiple mechanisms, including apoptosis, necrosis, necroptosis and pyroptosis31. Apoptosis and necroptosis can be distinguished by the accumulation of cytokeratin-18 cleavage product M30 in the circulation. M30 is a caspase-dependent cleavage product of M65, the full-length cytokeratin 1830, 32. Interestingly, cytokeratin-18 cleavage products can distinguish patients with NASH from AH33 and the concentration of full-length cytokeratin-18 (M65), indicative of necrotic/necroptotic cell death, increases with severity of AH33, 34. Recent data also suggests that circulating concentrations of RIP3, a mediator of necroptosis, can also efficiently distinguish patients with mild AH compared to NASH29. These recent studies indicate that markers of specific cell death pathways, as well as specific chemokines, may be developed into biomarkers to distinguish ALD from NASH and also provide prognostic information in cases of mild liver disease34.
Nutrition
Undernutrition and malnutrition are commonly associated with ALD, due at least in part to the consumption of alcohol, which provides calories, but little nutritional value35, 36. Both micro and macronutrient status is compromised in ALD, likely contributing to poor clinical outcomes. Low nutritional intake is associated with increased mortality in patients with severe AH37. While nutritional support can improve outcomes in severe AH35, a recent clinical trial testing the impact of enteral nutrition in patients with AH reported that it is difficult to provide adequate nutrition to this population37. However, it is possible that outcomes for patients with more moderate ALD, particularly when associated with obesity, as in our Case#1, would be improved by routine nutritional assessment and involvement of a dietician35, 36.
PTSD:
A major factor in AUD development are concomitant mental health disorders. This Veteran suffers from PTSD, which could play a key role in AUD and ALD38. Co-existent mental health disorders such as depression, anxiety and PTSD have the potential to encourage AUD and worsen the progression towards cirrhosis and alcohol-associated hepatitis. The bidirectional transition between PTSD and AUD is sex-specific with greater vulnerability in women compared to men39. The risk of AUD is higher overall in patients with PTSD, especially within the Veterans system38. However, even in situations unrelated to deployment or war, there is a robust association with PTSD and AUD40. Cognitive testing in Veterans with cirrhosis with and without PTSD showed worsened impulse control, which was also subsequently associated with differences in gut microbial structure and function41, 42. A recent systematic review concluded that PTSD substantially increases the cue reactivity for alcohol craving43. Therefore, undiagnosed mental health disorders, especially PTSD, such as in the case of our patient, should be sought out and treated in conjunction with AUD to reduce craving and potentially improve outcomes. It is encouraging that studies have shown that continued AUD did not reduce attendance or treatment for PTSD and vice-versa44.
Treatment for heavy drinking and alcohol use disorder
AUD is diagnosed based on Diagnostic and Statistical Manual of Mental Disorders, 5th edition (DSM-5) criteria. AUDIT scoring makes use of 10 questions that assess alcohol intake, likelihood for dependence and experience of alcohol related harm; scores range from 0 (abstinence), 1–7 (low consumption and risk); 8–14 (potential harmful drinking) and >15 (likely alcohol dependence, moderate to severe AUD). Since AUDIT-10 assessment is time consuming and requires a highly trained interviewer; use of the condensed AUDIT-C has proven useful in Hepatology clinics45. However, a full AUDIT screen is necessary to complete a diagnosis of AUD. Cessation of alcohol consumption improves symptoms and mortality from ALD; however, our understanding of best practices for treatment of AUD in ALD patients is limited46, 47. Further, initiation of treatment, while available, has been limited, particularly for Veterans, perhaps due to a difficulty in the ability of hepatologists/gastroenterologists to initiate treatment48, 49. On subsequent study, there were specific patient and provider factors that underlie these AUD therapy offers and acceptance50. Recent approaches to treatment have made use of multi-disciplinary clinics including hepatologists, psychologists and social workers have shown promising preliminary results in reducing hospitalizations of ALD patients45, 46, 51, 52. These strategies are often difficult to apply in practice because of perceived knowledge gaps on behalf of hepatology and GI practitioners, especially when there is limited access to addiction specialists. These barriers can be overcome by the encouraging trend towards educating hepatology and GI staff about the pros and cons of specific medications for AUD harm reduction, relapse prevention and craving reduction to provide direct prescription and supervision of these therapies in liver clinics53, 54. In addition, it is important to rally resources related to addiction therapy in the community. It is important to recognize the treatment strategies for AUD may of necessity vary, depending on the stage of disease (see below for Cases#2 and #3). For example, pharmacotherapeutic protocols for AUD need to account for changes in hepatic drug metabolism, renal function, and interactions with immunosuppressive drugs45, 47, 52. A recent meta-analysis concluded that integrated therapy including cognitive behavioral therapy with medical care was most effective at reducing alcohol relapse in patients with ALD51.
CASE 2:
A 57-year-old woman is seen in the hepatology clinic for routine follow-up of her decompensated cirrhosis. The patient had an episode of confusion two years ago diagnosed as hepatic encephalopathy and also developed ascites. These are now controlled on lactulose and diuretics, but she occasionally has episodes of “slowing down” according to her husband. Her cirrhosis was presumed to be due to hepatitis C and alcohol. HCV was cured four years ago. The patient was a heavy alcohol user in the past with several DUIs and missed work but over the last 5 years had cut down significantly. Currently she drinks 5–8 beers over the weekend, which was corroborated by the husband who accompanied her.
In clinic today, she appears slow but is oriented to time, place, and person. She is tremulous and feels like she has trouble remembering things. Her MELD score is 18.
Questions:
What is the natural history of disease progression in those who continue to drink despite HCV eradication?
What AUD-cessation therapies can be administered in this patient group?
How do we differentiate between HE and alcohol-associated brain syndromes in this patient?
Should she be placed on the transplant waiting list?
What are the consequences of declining to place this patient on the transplant waiting list?
HCV and alcohol interaction:
HCV and alcohol are synergistic in progression to cirrhosis and further alcohol misuse worsens the progression55. During the era of interferon-based therapies, where HCV eradication was relatively difficult, there was still evidence that continued alcohol misuse despite HCV eradication could worsen cirrhosis progression56, 57. In this current day and age with DAA therapy, there is ample evidence that HCV cure can reduce progression to decompensation and reduce the development of HCC. However, after HCV cure, continued insults to the liver i.e., alcohol misuse and obesity or weight gain will enhance the progression to complications (BAVENO VII consensus in press). Therefore, it is important to counsel patients to continue to work on all etiologies of cirrhosis and for clinicians to be vigilant regarding both alcohol misuse and weight gain in patients in whom HCV has been eradicated. This is relevant even in decompensated patients such as in case 2, where there is good quality evidence that alcohol abstinence can reduce the progression to HCC and further decompensation58. Therefore, due to continued alcohol misuse, this patient remains at high-risk for HCC and should be candidate for AUD pharmacotherapy and/or behavioral therapy.
AUD Cessation therapies in decompensated cirrhosis:
In decompensated cirrhosis without AH, there is a relatively high urgency to reduce alcohol consumption, prevent relapse, and ensure that disease does not progress59, 60. AUD therapy becomes challenging in patients with decompensated cirrhosis due to hepatic and renal handling of drugs such as disulfiram and naltrexone60. However, others such as baclofen, acamprosate and topiramate can be potentially used under close multi-disciplinary supervision45, 47, 52. The evidence basis for some medications such as baclofen and gabapentin are evolving and further data are needed61. For this patient, it is also important to determine the underlying psychiatric or psychological reasons for continuing to drink despite decompensated cirrhosis62. Those conditions should also be treated simultaneously. In addition to pharmacotherapy, behavioral approaches are needed to integrate the patient’s motivations, insight into the disease process, and to assess adherence, which would be important for subsequent transplant assessment. An intriguing recent trial showed that fecal microbiota transplantation (FMT) was associated with reduction in craving and alcohol consumption in cirrhosis63.
The situation in this person is further complicated due to HE, which often reduces insight, attention and short-term memory64–66. This requires greater collaboration between hepatologists, addiction medicine specialists, and behavioral therapists to ensure understanding of the processes. As with any therapy, the insight as well as willingness to undergo therapy for AUD on the part of the patient is critical for success and adherence.
Differentiation between brain complications associated with alcohol use and cirrhosis:
The patient in question has several competing or concomitant reasons for brain dysfunction. Prior HCV, current and prior alcohol misuse, nutritional abnormalities, and HE can all contribute to her current altered mental status and prior “slowness”67. It is important to differentiate between HE and other complications since they respond differently to alcohol cessation and transplantation68 (Figure 1). The approach towards actively confused patients versus those who are outpatients with slowness differs. The following flowcharts show the approaches towards these patients (Figures 2 and 3).
A global view of several influences on brain function is critical early during the inpatient period given the major risk of withdrawal and related consequences of delirium tremens69, 70. In this patient, there is tremulousness and difficulty remembering things, but she remains oriented to time, place, and person. This is likely going to develop into withdrawal and therefore a CIWA protocol should be followed71. Given the overall “slowness” there could also be an overlay of HE in this person, which is why the evolution of brain changes over the next few hours is important. Intoxication, withdrawal, and HE have distinct symptoms; HE rarely has symptoms of excitatory motor activity, hallucinations, or confabulation which are typical in alcohol withdrawal64. In the outpatient setting, specialized neuropsychological referral may be needed. In general, covert or minimal HE does not affect long-term memory and impairment of that cognitive domain is likely related to alcohol-associated dementia65, 67. As always, caregivers and substance abuse treatment specialists are important pieces of this management plan. Specialized nutritional evaluation is also necessary to recognize and treat micronutrient deficiencies72.
Liver transplantation considerations:
There was intense debate regarding the 6-month abstinence rule for patients being considered for liver transplant, which has now been replaced by a growing consensus that a one-size fits all approach fixed-time approach is not optimal for good outcomes post-transplant45, 73. Relapse after LT has major consequences on the graft and patient health, which includes rejection and death73. These are related to sustained heavy drinking rather than occasional slips and the thinking has evolved beyond absolute abstinence towards reduction of sustained drinking74, 75. Classification of post-LT relapse as heavy or not reduces the stigma when this is encountered and could encourage patients to communicate truthfully without judgement. In a recent meta-analysis of patients with all forms of AUD in the setting of chronic liver disease, psychiatric co-morbidities, pre-transplantation abstinence <6 months, single status and smoking predicted relapse and heavy relapse76.
Therefore, we need to develop an individualized multi-disciplinary plan. For this patient, we may have time to set up a substance abuse program with monitoring of alcohol use extensively using serum biomarkers. It is also likely that the MELD would decrease with abstinence and often patients with relatively lower MELD score recover enough to be de-listed77. However, her MELD is relatively high so listing with encouraging abstinence may be an important option78. If she had additional AH, we would need a more rapid combination of behavioral and medical therapies to approach her alcohol cessation while ensuring her HE and ascites are managed appropriately. This is important because the consequences of not listing patients with AH despite abstinence are dire and can be predicted79, 80. This is further explored in case 3. The caregiver and support system needs to be present and engaged for success in pre- and post-LT settings.
CASE 3:
A 32-year-old woman was brought in by her mother to this hospital after being found collapsed in her house. The patient had been laid off during the pandemic and had missed several AA meetings and remote meetings with her alcohol counselor. According to the mother, the patient had been increasingly despondent over the last few weeks, and she suspects that “she had gone back to drinking”. Apart from the mother, the patient was in contact with her sister during the pandemic.
In the hospital, she was confused with respect to time and place but not person. She is jaundiced and has asterixis and ascites with pedal edema. These were not present during her last primary MD visit. There is also suspicion of aspiration pneumonia on her chest X ray for which she is started on antibiotics. Her MELD score is 31 and her Maddrey’s Discriminant Function is 36. On reassessment at day 4 after antibiotics, fluids and other supportive therapy, the patient is now completely alert and oriented. She is still jaundiced with MELD of 29 and Maddrey’s Discriminant Function of 32. Steroids are now started.
Questions:
What prognostic models will help define the outcome of patients with AH?
What is the role of microbial modification in the prognostication and therapy of patients with severe and moderate stages of AH?
Is this person a transplant candidate?
What are the predictors of relapse to problem drinking in this patient either before or after liver transplant?
Prognosis in AH
Traditional static scores that assess severity and prognosis for patients in AH include Maddrey’s Discriminant Function, MELD, ABIC (Age, serum Bilirubin, INR, and serum Creatinine) and GAHS (Glasgow Alcoholic Hepatitis Score). These scores are routinely used to determine if patients should be treated or not with prednisolone, the only approved treatment for AH5, 81. The more dynamic Lille score for predicting outcomes to steroid treatment in patients with AH is also widely used5, 81. Over time, changes in serum bilirubin trajectory in hospitalized patients and those placed on prednisolone over time (days 1 through 4) could determine the prognosis for the patient in Case #382. Additional prognostic indicators of specific complications contributing to mortality in AH, including bacterial or fungal infections, acute kidney injury or systemic infections, are less robust. Recent collaborative studies in many countries combining clinical trials with translational studies have begun to improve our understanding of potential biomarkers that will provide better prognostic indicators. However, very little data is available to differentiate prognosis between men and women, as well as different age groups.
Risk of bacterial and fungal infection
Patients with severe AH often develop severe infections that negatively impact short-term prognosis. Prednisolone is the first-line therapy for AH; however, the effectiveness of prednisolone in suppressing inflammatory responses in the liver is also associated with impaired resistance to microbial infections, thus negatively impacting patient survival. A recent meta-analysis, including 11 studies with more than 2000 patients with severe AH, concluded that while prednisolone improved outcomes at 28 days with a hazard ratio of 0.64; 6-month mortality was not improved83. Data from the double-blind placebo controlled STOPAH trial found that patients treated with prednisolone had almost twice the incidence of infection after treatment with prednisolone84. Fungal infections may also be increased in patient treated with prednisolone. In a meta-analysis of over 1000 patients, there was a strong association between steroid treatment and fungal infections impacting short term prognosis85. A recent multicenter cohort study of patients with AH used a combination of MELD score, ascites, WBC count, and use of corticosteroids to develop a predictive model for identifying patients at high risk of infection86.
Inflammatory signal and cell death markers as prognostic indicators
Given the critical role of the innate immune system, cell death and impaired hepatocyte regeneration in the development of ALD, recent studies have focused on identifying prognostic biomarkers within these pathways for morbidity and mortality in AH. For example, higher circulating concentrations of several markers of macrophage activation and immune danger signals were strong predictors of infection, organ failures and 90-day mortality in patients with severe AH87. Complement, another important arm of the innate immune response, is also activated in patients with AH and circulating concentrations of complement factor I and soluble C5b9 predicted mortality in AH patients as well as MELD score88. The M65 cleavage fragments of cytokeratin-18, associated with non-apoptotic hepatocyte death33, 89 and RIP3, a member of the necroptotic cell death pathway29, are both strong predictors of mortality in AH. miRNAs, both circulating or as cargo within extracellular vesicles, are another promising source for prognostic biomarkers, particularly related to regulation of hepatocyte regeneration90, 91. Other cargo in extracellular vesicles, including sphingolipids, are also potential prognostic indicators in AH92. Although these biomarkers related to inflammation and cell death are promising prognostic tools, further validation is required before they become part of clinical practice. An overview of the biomarkers and unmet needs for AH is shown in Table 1.
Table 1:
Improved approaches to decrease alcohol intake |
---|
|
Development of precision medicine biomarkers for risk of poor outcomes |
Innate immune related
|
Therapeutic avenues for treatment of AH |
Innate immune modulation
|
Modification of the microbiome
There is a growing appreciation for the contributions of intestinal microbiota in progression of ALD; recent studies have begun to interrogate intestinal microbiota as a potential therapeutic target in ALD. Numerous studies have now provided descriptive information as to the impact of alcohol use and ALD on the gut microbiome25. Excessive alcohol use, often coupled with poor nutrition or obesity, leads to intestinal dysbiosis. Dysbiosis is accompanied by additional changes in the barrier function of the intestinal epithelium, mucosal immunity and generation of microbial products into the circulation. Multiple approaches have been taken to normalize the intestinal microbiome, including use of antibiotics, probiotics, synbiotics, and fecal microbiota transfer. As we gain a better understanding of specific impact of alcohol on the microbiome, investigators are increasingly moving from untargeted approaches, e.g., anti-biotics and pro-biotics, to more targeted approaches for treatment.
Early studies used untargeted approaches to modify the gut microbiome. Based on studies in animal models, clinical trials were designed to evaluate the role of antibiotics in ALD and other chronic liver diseases25. While many of the trials showed short term improvement in some clinical parameters, changes in liver-related complications were not consistent25. Similarly, studies utilizing probiotics often show reduction in circulating cytokines and microbial metabolites. As probiotics are known to be well tolerated in patients with liver disease, more recent studies have focused on use of probiotics25. As with antibiotic studies, the clinical trials tend to involve only short-term treatment, with small sample size and only limited measures of efficacy for improving liver injury. As an example of the few randomized controlled clinical trials conducted in this area, Han and colleagues reported that short-term probiotic treatment reduced TNF-a and LPS in the circulation, as well as E. coli colony forming units, but did not improve transaminases93. The value of this study for treatment of patients with AH, as with many studies aimed at modifying the microbiome, may be limited as enrollees in the study had modest degrees of liver injury and transaminases are of limited values in diagnosing ALD. Other generalized approaches to improving the host microbial communities include the use of prebiotics, pro-biotics and synbiotics (combining pre and pro biotics)94. Multiple, relatively small studies have shown some promise, but multiple challenges have precluded the use of these compounds in clinical practice95
Current clinical trials are designed to restore specific changes in the microbiome observed in patients with ALD. For example, Lactobacillus species are typically decreased in ALD25, 95. Clinical trials to replace Lactobacillus metabolites, rather than the bacteria themselves are underway. One current clinical trial is testing efficacy and safety of the supernatant of Lactobacillus rhamnosus GG in patients with moderate AH (MELD score <21), with primary endpoint as change in MELD score after 30 days (NCT01922895).
FMT is also being investigated as a means to restore healthy gut microbiota in this setting in addition to the trial mentioned above in stable patients with cirrhosis63. Several small open label studies have shown promising results in patients with severe AH with evidence for sustained coexistence of donor and recipient species at 6–12 months after FMT95, suggesting that FMT may have a lasting impact on the recipient’s gut microbiota network. However, additional randomized clinical trials are required to confirm that FMT sustainably modifies the recipient microbiota and improves ALD without complications, even in patients with severe AH95. Studies in murine models of ALD also point to the promise of therapeutics that specifically target specific microbial species known to contribute to liver injury. For example, Schnabl’s group reported that bacteriophage targeting of cytolysin-secreting Enterococcus faecalis (E. faecalis) reduced harmful concentrations of cytolysin in the liver and reduced ethanol-induced liver96. Small molecule inhibitors of bacterial enzymes are also under investigation. For example, Brown and colleagues recently reported that small molecule inhibition of the gut microbial transformation of choline into trimethylamine (TMA), a microbial metabolite elevated in the circulation of patients with AH, prevented ethanol-induced liver injury in a murine model of ALD [Brown, et al in press eLife 2022]. These targeted precision approaches to modifying the gut microbiome hold promise but need to be further explored in clinical trials.
Transplant suitability and risk factors for relapse in patients with AH:
Practice patterns regarding transplantation in AH patients have dramatically shifted over the last decade47, 97. There is an increase in patients with AH being evaluated, listed, and undergoing transplant and a refinement of static and dynamic scoring systems98. The rate of this has increased over the COVID-19 pandemic99. Selected patients with AH do well from a survival standpoint post-LT but there are differences in the criteria for listing and selection for these patients across sites100 . A consensus conference determined that a multi-disciplinary approach focused on patients with their first episode of AH, who are not responders to traditional medical therapies without other substance abuse and good social support could be considered73. A detailed analysis by the transplant team, the addiction specialists, social workers is needed before careful transplant committee selection. There is also a recognition that with successful abstinence, it is likely that patients may not ultimately require a LT.
Specific factors that predict return to problem drinking rather than occasional slips need to be considered in consultation with addiction specialists73, 99. In the setting of AH, the SALT score (Sustained alcohol use post-Liver transplant) <5 had a negative predictive value for sustained alcohol use post-LT of 95%. The SALT score consists of >10 drinks per day at initial encounter (+4), multiple prior rehabilitation attempts (+4), prior alcohol-associated legal issues (+2) and prior illicit substance use (+1)101.
These include younger age at drinking, heavy alcohol use, poor insight into ALD, as well as other untreated mental health problems or substance abuse. In addition, a poor social support, and a long track record of non-adherences with visits and therapies are likely to indicate a high risk of alcohol relapse after liver transplantation99.
In the case of this patient, although she is young and has heavy alcohol use history, her engagement with AUD specialists, social support, and first decompensating event are favorable indicators of abstinence. There needs to be continued assessment as an inpatient of her medical status, i.e., improvement with steroids and correction of infection over time, as well as her ability to meaningfully engage with addiction specialists. Inpatient linkage with AUD specialists and their assessment of the patient’s insight, ability to follow through and support from family members will remain critical to evaluate her ultimate eligibility in case she does not respond to steroids over the next week.
FUTURE DIRECTIONS:
As illustrated by our case studies, treatment of ALD is complex and dependent on disease stage and co-morbidities. Studies utilizing ALD animal models continue to dissect out the complex interactions between alcohol consumption, diet, and the microbiome. Clinical and genetic studies at various stages of ALD are beginning to identify potential genetic markers that may either protect or contribute to disease progression. Improved phenotypic identification of disease stage, particularly for AH102, and standardization of clinical trial design103 will likely lead to successful clinical trials identifying effective ALD therapeutics. Importantly, advances in biomarker identification related to inflammation, cell death and microbial dysbiosis hold promise for personalized medicine approaches. Finally, recent advances in integrating multi-disciplinary clinics are critical for improved care for patients with ALD.
Support:
This work was supported in part by NIH grants; P50AA024333, U01 AA026938, U01 AA026264 (LEN); R21TR003095 (JSB) and VA Merit Review 2I0CX001076 (JSB)
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
Conflict of interest: None for any author
References:
- 1.Julien J, Ayer T, Bethea ED, et al. Projected prevalence and mortality associated with alcohol-related liver disease in the USA, 2019–40: a modelling study. Lancet Public Health 2020;5:e316–e323. [DOI] [PubMed] [Google Scholar]
- 2.Agabio R, Pisanu C, Gessa GL, et al. Sex Differences in Alcohol Use Disorder. Curr Med Chem 2017;24:2661–2670. [DOI] [PubMed] [Google Scholar]
- 3.Vaeth PA, Wang-Schweig M, Caetano R. Drinking, Alcohol Use Disorder, and Treatment Access and Utilization Among U.S. Racial/Ethnic Groups. Alcohol Clin Exp Res 2017;41:6–19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Levy R, Catana AM, Durbin-Johnson B, et al. Ethnic differences in presentation and severity of alcoholic liver disease. Alcohol Clin Exp Res 2015;39:566–574. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Delacôte C, Bauvin P, Louvet A, et al. A Model to Identify Heavy Drinkers at High Risk for Liver Disease Progression. Clin Gastroenterol Hepatol 2020;18:2315–2323.e6. [DOI] [PubMed] [Google Scholar]
- 6.Trépo E, Valenti L. Update on NAFLD genetics: From new variants to the clinic. J Hepatol 2020;72:1196–1209. [DOI] [PubMed] [Google Scholar]
- 7.Choudhary NS, Duseja A. Genetic and epigenetic disease modifiers: non-alcoholic fatty liver disease (NAFLD) and alcoholic liver disease (ALD). Transl Gastroenterol Hepatol 2021;6:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Atkinson SR, Way MJ, McQuillin A, et al. Homozygosity for rs738409:G in PNPLA3 is associated with increased mortality following an episode of severe alcoholic hepatitis. J Hepatol 2017;67:120–127. [DOI] [PubMed] [Google Scholar]
- 9.Schwantes-An TH, Darlay R, Mathurin P, et al. Genome-wide Association Study and Meta-analysis on Alcohol-Associated Liver Cirrhosis Identifies Genetic Risk Factors. Hepatology 2021;73:1920–1931. [DOI] [PubMed] [Google Scholar]
- 10.Trépo E, Caruso S, Yang J, et al. Common genetic variation in alcohol-related hepatocellular carcinoma: a case-control genome-wide association study. Lancet Oncol 2021. [DOI] [PubMed] [Google Scholar]
- 11.Sharma A, Mir R, Galande S. Epigenetic Regulation of the Wnt/β-Catenin Signaling Pathway in Cancer.Front Genet 2021;12:681053. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Miao CG, Yang YY, He X, et al. Wnt signaling in liver fibrosis: progress, challenges and potential directions. Biochimie 2013;95:2326–35. [DOI] [PubMed] [Google Scholar]
- 13.Tawa EA, Hall SD, Lohoff FW. Overview of the Genetics of Alcohol Use Disorder. Alcohol Alcohol 2016;51:507–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Kranzler HR, Zhou H, Kember RL, et al. Genome-wide association study of alcohol consumption and use disorder in 274,424 individuals from multiple populations. Nat Commun 2019;10:1499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Hart AB, Kranzler HR. Alcohol Dependence Genetics: Lessons Learned From Genome-Wide Association Studies (GWAS) and Post-GWAS Analyses. Alcohol Clin Exp Res 2015;39:1312–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Edwards AC, Sundquist K, Sundquist J, et al. Genetic and environmental influences on the progression from alcohol use disorder to alcohol-related medical conditions. Alcohol Clin Exp Res 2021;45:2528–2535. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Chiang DJ, McCullough AJ. The impact of obesity and metabolic syndrome on alcoholic liver disease. Clin Liver Dis 2014;18:157–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Ikejima K, Kon K, Yamashina S. Nonalcoholic fatty liver disease and alcohol-related liver disease: From clinical aspects to pathophysiological insights. Clin Mol Hepatol 2020;26:728–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Ntandja Wandji LC, Gnemmi V, Mathurin P, et al. Combined alcoholic and non-alcoholic steatohepatitis. JHEP Rep 2020;2:100101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Parker R, Kim SJ, Gao B. Alcohol, adipose tissue and liver disease: mechanistic links and clinical considerations. Nat Rev Gastroenterol Hepatol 2018;15:50–59. [DOI] [PubMed] [Google Scholar]
- 21.Wang H, Mehal W, Nagy LE, et al. Immunological mechanisms and therapeutic targets of fatty liver diseases. Cell Mol Immunol 2021;18:73–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Voican CS, Njiké-Nakseu M, Boujedidi H, et al. Alcohol withdrawal alleviates adipose tissue inflammation in patients with alcoholic liver disease. Liver Int 2015;35:967–78. [DOI] [PubMed] [Google Scholar]
- 23.Lasselin J, Magne E, Beau C, et al. Adipose inflammation in obesity: relationship with circulating levels of inflammatory markers and association with surgery-induced weight loss. J Clin Endocrinol Metab 2014;99:E53–61. [DOI] [PubMed] [Google Scholar]
- 24.Monnig MA, Lamb PS, Parra JM, et al. Immune Response to an Acute Moderate Dose of Alcohol in Healthy Young Adults. Alcohol Alcohol 2020;55:616–623. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Bluemel S, Williams B, Knight R, et al. Precision medicine in alcoholic and nonalcoholic fatty liver disease via modulating the gut microbiota. Am J Physiol Gastrointest Liver Physiol 2016;311:G1018–g1036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Parker R, Kim SJ, Im GY, et al. Obesity in acute alcoholic hepatitis increases morbidity and mortality. EBioMedicine 2019;45:511–518. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. https://www.research.va.gov/topics/obesity.cfm.
- 28.Hwang S, Ren T, Gao B. Obesity and binge alcohol intake are deadly combination to induce steatohepatitis: A model of high-fat diet and binge ethanol intake. Clin Mol Hepatol 2020;26:586–594. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Miyata T, Wu X, Fan X, et al. Differential role of MLKL in alcohol-associated and non-alcoholassociated fatty liver diseases in mice and humans. JCI Insight 2021;6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Feldstein AE, Alkhouri N, De Vito R, et al. Serum cytokeratin-18 fragment levels are useful biomarkers for nonalcoholic steatohepatitis in children. Am J Gastroenterol 2013;108:1526–31. [DOI] [PubMed] [Google Scholar]
- 31.Miyata T, Nagy LE. Programmed cell death in alcohol-associated liver disease. Clin Mol Hepatol 2020;26:618–625. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Ku NO, Strnad P, Bantel H, et al. Keratins: Biomarkers and modulators of apoptotic and necrotic cell death in the liver. Hepatology 2016;64:966–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Vatsalya V, Cave MC, Kong M, et al. Keratin 18 Is a Diagnostic and Prognostic Factor for Acute Alcoholic Hepatitis. Clin Gastroenterol Hepatol 2020;18:2046–2054. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Atkinson SR, Grove JI, Liebig S, et al. In Severe Alcoholic Hepatitis, Serum Keratin-18 Fragments Are Diagnostic, Prognostic, and Theragnostic Biomarkers. Am J Gastroenterol 2020;115:1857–1868. [DOI] [PubMed] [Google Scholar]
- 35.McClain CJ, Rios CD, Condon S, et al. Malnutrition and Alcohol-Associated Hepatitis. Clin Liver Dis 2021;25:557–570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Bhavsar-Burke I, Jansson-Knodell CL, Gilmore AC, et al. Review article: the role of nutrition in alcohol-associated liver disease. Aliment Pharmacol Ther 2021;53:1268–1276. [DOI] [PubMed] [Google Scholar]
- 37.Moreno C, Deltenre P, Senterre C, et al. Intensive Enteral Nutrition Is Ineffective for Patients With Severe Alcoholic Hepatitis Treated With Corticosteroids. Gastroenterology 2016;150:903–10 e8. [DOI] [PubMed] [Google Scholar]
- 38.Panza KE, Kline AC, Na PJ, et al. Epidemiology of DSM-5 alcohol use disorder in U.S. military veterans: Results from the National Health and Resilience in Veterans Study. Drug Alcohol Depend 2021;231:109240. [DOI] [PubMed] [Google Scholar]
- 39.Peltier MR, Roberts W, Verplaetse TL, et al. Sex Differences across Retrospective Transitions in Posttraumatic Stress and Substance Use Disorders. J Dual Diagn 2021:1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Pietrzak RH, Goldstein RB, Southwick SM, et al. Prevalence and Axis I comorbidity of full and partial posttraumatic stress disorder in the United States: results from Wave 2 of the National Epidemiologic Survey on Alcohol and Related Conditions. J Anxiety Disord 2011;25:456–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Burroughs TK, Wade JB, Ellwood MS, et al. Effect of Post-Traumatic Stress Disorder on Cognitive Function and Covert Hepatic Encephalopathy Diagnosis in Cirrhotic Veterans. Dig Dis Sci 2018;63:481–485. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Bajaj JS, Sikaroodi M, Fagan A, et al. Posttraumatic stress disorder is associated with altered gut microbiota that modulates cognitive performance in veterans with cirrhosis. Am J Physiol Gastrointest Liver Physiol 2019;317:G661–G669. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Renaud F, Jakubiec L, Swendsen J, et al. The Impact of Co-occurring Post-traumatic Stress Disorder and Substance Use Disorders on Craving: A Systematic Review of the Literature. Front Psychiatry 2021;12:786664. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kline AC, Straus E, Lyons RC, et al. Substance use predictors of attendance among veterans in integrated PTSD and alcohol use disorder treatment. J Subst Abuse Treat 2021;124:108278. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Crabb DW, Im GY, Szabo G, et al. Diagnosis and Treatment of Alcohol-Associated Liver Diseases: 2019 Practice Guidance From the American Association for the Study of Liver Diseases. Hepatology 2020;71:306–333. [DOI] [PubMed] [Google Scholar]
- 46.Mellinger JL, Winder GS, Fernandez AC, et al. Feasibility and early experience of a novel multidisciplinary alcohol-associated liver disease clinic. J Subst Abuse Treat 2021;130:108396. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Arab JP, Izzy M, Leggio L, et al. Management of alcohol use disorder in patients with cirrhosis in the setting of liver transplantation. Nat Rev Gastroenterol Hepatol 2022;19:45–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Mellinger JL, Scott Winder G, DeJonckheere M, et al. Misconceptions, preferences and barriers to alcohol use disorder treatment in alcohol-related cirrhosis. J Subst Abuse Treat 2018;91:20–27. [DOI] [PubMed] [Google Scholar]
- 49.Rogal S, Youk A, Zhang H, et al. Impact of Alcohol Use Disorder Treatment on Clinical Outcomes Among Patients With Cirrhosis. Hepatology 2020;71:2080–2092. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Chaudhari RB, Duong N, Shrestha S, et al. Patient and provider-level factors that underlie alcohol use disorder treatment offer and acceptance in veterans with cirrhosis. Alcohol Clin Exp Res 2022. [DOI] [PubMed] [Google Scholar]
- 51.Khan A, Tansel A, White DL, et al. Efficacy of Psychosocial Interventions in Inducing and Maintaining Alcohol Abstinence in Patients With Chronic Liver Disease: A Systematic Review. Clin Gastroenterol Hepatol 2016;14:191–202.e1–4; quiz e20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lucey MR, Singal AK. Integrated Treatment of Alcohol Use Disorder in Patients With Alcohol-Associated Liver Disease: An Evolving Story. Hepatology 2020;71:1891–1893. [DOI] [PubMed] [Google Scholar]
- 53.Winters AC, Aby ES, Fix OK, et al. Joining the Fight: Enhancing Alcohol Treatment Education in Hepatology. Clin Liver Dis (Hoboken) 2021;18:225–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Im GY, Mellinger JL, Winters A, et al. Provider Attitudes and Practices for Alcohol Screening, Treatment, and Education in Patients With Liver Disease: A Survey From the American Association for the Study of Liver Diseases Alcohol-Associated Liver Disease Special Interest Group. Clin Gastroenterol Hepatol 2021;19:2407–2416 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Wieland A, Everson GT. Co-existing Hepatitis C and Alcoholic Liver Disease: A Diminishing Indication for Liver Transplantation? Alcohol Alcohol 2018;53:187–192. [DOI] [PubMed] [Google Scholar]
- 56.Vandenbulcke H, Moreno C, Colle I, et al. Alcohol intake increases the risk of HCC in hepatitis C virus-related compensated cirrhosis: A prospective study. J Hepatol 2016;65:543–51. [DOI] [PubMed] [Google Scholar]
- 57.van der Meer AJ, Feld JJ, Hofer H, et al. Risk of cirrhosis-related complications in patients with advanced fibrosis following hepatitis C virus eradication. J Hepatol 2017;66:485–493. [DOI] [PubMed] [Google Scholar]
- 58.Rodriguez M, Gonzalez-Dieguez ML, Varela M, et al. Impact of Alcohol Abstinence on the Risk of Hepatocellular Carcinoma in Patients With Alcohol-Related Liver Cirrhosis. Am J Gastroenterol 2021. [DOI] [PubMed] [Google Scholar]
- 59.Pearson MM, Kim NJ, Berry K, et al. Associations Between Alcohol Use and Liver-Related Outcomes in a Large National Cohort of Patients With Cirrhosis. Hepatol Commun 2021;5:2080–2095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Caputo F, Domenicali M, Bernardi M. Diagnosis and Treatment of Alcohol Use Disorder in Patients With End-Stage Alcoholic Liver Disease. Hepatology 2019;70:410–417. [DOI] [PubMed] [Google Scholar]
- 61.Anton RF, Latham P, Voronin K, et al. Efficacy of Gabapentin for the Treatment of Alcohol Use Disorder in Patients With Alcohol Withdrawal Symptoms: A Randomized Clinical Trial. JAMA Intern Med 2020;180:728–736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.DiMartini AF, Leggio L, Singal AK. Barriers to the management of alcohol use disorder and alcohol-associated liver disease: strategies to implement integrated care models. Lancet Gastroenterol Hepatol 2022;7:186–195. [DOI] [PubMed] [Google Scholar]
- 63.Bajaj JS, Gavis EA, Fagan A, et al. A Randomized Clinical Trial of Fecal Microbiota Transplant for Alcohol Use Disorder. Hepatology 2021;73:1688–1700. [DOI] [PubMed] [Google Scholar]
- 64.Vilstrup H, Amodio P, Bajaj J, et al. Hepatic encephalopathy in chronic liver disease: 2014 Practice Guideline by the American Association for the Study of Liver Diseases and the European Association for the Study of the Liver. Hepatology 2014;60:715–35. [DOI] [PubMed] [Google Scholar]
- 65.Weissenborn K, Ennen JC, Schomerus H, et al. Neuropsychological characterization of hepatic encephalopathy. J Hepatol 2001;34:768–73. [DOI] [PubMed] [Google Scholar]
- 66.Bajaj JS, Wade JB, Sanyal AJ. Spectrum of neurocognitive impairment in cirrhosis: Implications for the assessment of hepatic encephalopathy. Hepatology 2009;50:2014–21. [DOI] [PubMed] [Google Scholar]
- 67.Bajaj JS, Duarte-Rojo A, Xie JJ, et al. Minimal Hepatic Encephalopathy and Mild Cognitive Impairment Worsen Quality of Life in Elderly Patients With Cirrhosis. Clin Gastroenterol Hepatol 2020;18:3008–3016 e2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Bajaj JS. Alcohol, liver disease and the gut microbiota. Nat Rev Gastroenterol Hepatol 2019;16:235–246. [DOI] [PubMed] [Google Scholar]
- 69.Davis BC, Bajaj JS. Effects of Alcohol on the Brain in Cirrhosis: Beyond Hepatic Encephalopathy. Alcohol Clin Exp Res 2018;42:660–667. [DOI] [PubMed] [Google Scholar]
- 70.Singal AK, Bataller R, Ahn J, et al. ACG Clinical Guideline: Alcoholic Liver Disease. Am J Gastroenterol 2018;113:175–194. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Sullivan JT, Sykora K, Schneiderman J, et al. Assessment of alcohol withdrawal: the revised clinical institute withdrawal assessment for alcohol scale (CIWA-Ar). Br J Addict 1989;84:1353–7. [DOI] [PubMed] [Google Scholar]
- 72.Rossi RE, Conte D, Massironi S. Diagnosis and treatment of nutritional deficiencies in alcoholic liver disease: Overview of available evidence and open issues. Dig Liver Dis 2015;47:819–25. [DOI] [PubMed] [Google Scholar]
- 73.Asrani SK, Trotter J, Lake J, et al. Meeting Report: The Dallas Consensus Conference on Liver Transplantation for Alcohol Associated Hepatitis. Liver Transpl 2020;26:127–140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Pfitzmann R, Schwenzer J, Rayes N, et al. Long-term survival and predictors of relapse after orthotopic liver transplantation for alcoholic liver disease. Liver Transpl 2007;13:197–205. [DOI] [PubMed] [Google Scholar]
- 75.DiMartini A, Day N, Dew MA, et al. Alcohol consumption patterns and predictors of use following liver transplantation for alcoholic liver disease. Liver Transpl 2006;12:813–20. [DOI] [PubMed] [Google Scholar]
- 76.Chuncharunee L, Yamashiki N, Thakkinstian A, et al. Alcohol relapse and its predictors after liver transplantation for alcoholic liver disease: a systematic review and meta-analysis. BMC Gastroenterol 2019;19:150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Pose E, Torrents A, Reverter E, et al. A notable proportion of liver transplant candidates with alcohol-related cirrhosis can be delisted because of clinical improvement. J Hepatol 2021;75:275–283. [DOI] [PubMed] [Google Scholar]
- 78.Louvet A, Labreuche J, Artru F, et al. Main drivers of outcome differ between short term and long term in severe alcoholic hepatitis: A prospective study. Hepatology 2017;66:1464–1473. [DOI] [PubMed] [Google Scholar]
- 79.Musto J, Stanfield D, Ley D, et al. Recovery and outcomes of patients denied early liver transplantation for severe alcohol-associated hepatitis. Hepatology 2022;75:104–114. [DOI] [PubMed] [Google Scholar]
- 80.Lee BP, Samur S, Dalgic OO, et al. Model to Calculate Harms and Benefits of Early vs Delayed Liver Transplantation for Patients With Alcohol-Associated Hepatitis. Gastroenterology 2019;157:472–480 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Singal AK, Mathurin P. Diagnosis and Treatment of Alcohol-Associated Liver Disease: A Review. Jama 2021;326:165–176. [DOI] [PubMed] [Google Scholar]
- 82.Parker R, Cabezas J, Altamirano J, et al. Trajectory of Serum Bilirubin Predicts Spontaneous Recovery in a Real-World Cohort of Patients With Alcoholic Hepatitis. Clin Gastroenterol Hepatol 2022;20:e289–e297. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Louvet A, Thursz MR, Kim DJ, et al. Corticosteroids Reduce Risk of Death Within 28 Days for Patients With Severe Alcoholic Hepatitis, Compared With Pentoxifylline or Placebo-a Meta-analysis of Individual Data From Controlled Trials. Gastroenterology 2018;155:458–468 e8. [DOI] [PubMed] [Google Scholar]
- 84.Vergis N, Atkinson SR, Knapp S, et al. In Patients With Severe Alcoholic Hepatitis, Prednisolone Increases Susceptibility to Infection and Infection-Related Mortality, and Is Associated With High Circulating Levels of Bacterial DNA. Gastroenterology 2017;152:1068–1077 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Singal AK, Kodali S, Vucovich LA, et al. Diagnosis and Treatment of Alcoholic Hepatitis: A Systematic Review. Alcohol Clin Exp Res 2016;40:1390–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Penrice DD, Shah S, Kezer CA, et al. Risk Prediction of Nosocomial and Posthospital Discharge Infections in Alcohol-Associated Hepatitis. Hepatol Commun 2021;5:2096–2103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Saha B, Tornai D, Kodys K, et al. Biomarkers of Macrophage Activation and Immune Danger Signals Predict Clinical Outcomes in Alcoholic Hepatitis. Hepatology 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Fan X, McCullough RL, Huang E, et al. Diagnostic and Prognostic Significance of Complement in Patients With Alcohol-Associated Hepatitis. Hepatology 2021;73:983–997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Bissonnette J, Altamirano J, Devue C, et al. A prospective study of the utility of plasma biomarkers to diagnose alcoholic hepatitis. Hepatology 2017;66:555–563. [DOI] [PubMed] [Google Scholar]
- 90.Yang Z, Zhang T, Kusumanchi P, et al. Transcriptomic Analysis Reveals the MicroRNAs Responsible for Liver Regeneration Associated With Mortality in Alcohol-Associated Hepatitis. Hepatology 2021;74:2436–2451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Momen-Heravi F, Saha B, Kodys K, et al. Increased number of circulating exosomes and their microRNA cargos are potential novel biomarkers in alcoholic hepatitis. J Transl Med 2015;13:261. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Sehrawat TS, Arab JP, Liu M, et al. Circulating Extracellular Vesicles Carrying Sphingolipid Cargo for the Diagnosis and Dynamic Risk Profiling of Alcoholic Hepatitis. Hepatology 2021;73:571–585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Han SH, Suk KT, Kim DJ, et al. Effects of probiotics (cultured Lactobacillus subtilis/Streptococcus faecium) in the treatment of alcoholic hepatitis: randomized-controlled multicenter study. Eur J Gastroenterol Hepatol 2015;27:1300–6. [DOI] [PubMed] [Google Scholar]
- 94.Han Y, Glueck B, Shapiro D, et al. Dietary Synbiotic Supplementation Protects Barrier Integrity of Hepatocytes and Liver Sinusoidal Endothelium in a Mouse Model of Chronic-Binge Ethanol Exposure. Nutrients 2020;12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Siddiqui MT, Cresci GAM. Microbiota reprogramming for treatment of alcohol-related liver disease. Transl Res 2020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Duan Y, Llorente C, Lang S, et al. Bacteriophage targeting of gut bacterium attenuates alcoholic liver disease. Nature 2019;575:505–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Mathurin P, Moreno C, Samuel D, et al. Early liver transplantation for severe alcoholic hepatitis. N Engl J Med 2011;365:1790–800. [DOI] [PubMed] [Google Scholar]
- 98.Louvet A, Labreuche J, Artru F, et al. Combining Data From Liver Disease Scoring Systems Better Predicts Outcomes of Patients With Alcoholic Hepatitis. Gastroenterology 2015;149:398–406 e8; quiz e16–7. [DOI] [PubMed] [Google Scholar]
- 99.Bittermann T, Mahmud N, Abt P. Trends in Liver Transplantation for Acute Alcohol-Associated Hepatitis During the COVID-19 Pandemic in the US. JAMA Netw Open 2021;4:e2118713. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Lee BP, Mehta N, Platt L, et al. Outcomes of Early Liver Transplantation for Patients With Severe Alcoholic Hepatitis. Gastroenterology 2018;155:422–430 e1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Lee BP, Vittinghoff E, Hsu C, et al. Predicting Low Risk for Sustained Alcohol Use After Early Liver Transplant for Acute Alcoholic Hepatitis: The Sustained Alcohol Use Post-Liver Transplant Score. Hepatology 2019;69:1477–1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Crabb DW, Bataller R, Chalasani NP, et al. Standard Definitions and Common Data Elements for Clinical Trials in Patients With Alcoholic Hepatitis: Recommendation From the NIAAA Alcoholic Hepatitis Consortia. Gastroenterology 2016;150:785–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Dasarathy S, Mitchell MC, Barton B, et al. Design and rationale of a multicenter defeat alcoholic steatohepatitis trial: (DASH) randomized clinical trial to treat alcohol-associated hepatitis. Contemp Clin Trials 2020;96:106094. [DOI] [PMC free article] [PubMed] [Google Scholar]