Skip to main content

Impact of direct-acting antivirals on neuropsychiatric and neurocognitive dysfunction in chronic hepatitis C patients

Abstract

Background

Hepatitis C virus (HCV) infection is associated with psychiatric and cognitive dysfunctions. We aimed to investigate depression, anxiety, and cognitive function of chronic hepatitis C (CHC) patients before and after treatment with direct-acting antivirals (DAAs). Forty CHC patients (20 non-cirrhotic and 20 cirrhotic) who had undergone DAA treatment in our outpatient clinic and ten controls. We administered the Hospital Anxiety and Depression questionnaires to measure the anxiety and depression symptoms and the Cognitive Abilities Screening Instruments (CASI) to measure the cognitive function at the beginning and 3 months after the end of the treatment.

Results

Sustained virological response (SVR) was achieved in all patients. Post-treatment anxiety and depression scores showed a significant improvement than pre-treatment ones in CHC patients. Regarding CASI, before and after the treatment, a statistical significance was found in short-term memory (P = 0.001), concentration (P = 0.033), abstract thinking and judgment (P = 0.024), total (P = 0.001) in non-cirrhotic, Also, an improvement was seen in long-term memory (P = 0.015), short-term memory (P < 0.001), concentration (P = 0.024) and total (P = 0.01) in cirrhotic. However, these changes were still impaired in post-treated cirrhotic compared to controls.

Conclusions

CHC patients' anxiety, depression, and cognitive function partially improved after DAA therapy. Besides, improving the status of CHC, reversibility of cognitive dysfunction in non-cirrhotic patients may indicate the importance of treatment in early stages of liver disease.

Background

Hepatitis C virus (HCV) infection is a leading cause of chronic liver disease globally, with cirrhosis and hepatocellular carcinoma (HCC) being the most common complications [1]. Neuropsychiatry manifestations of extrahepatic symptoms of HCV infection are common such as cognitive dysfunctions, sleep disorders, depression, anxiety, and anger/hostility neurological and mental problems [2,3,4]. Compared to the general population, patients with HCV infection had a greater frequency of psychiatric problems, including substance abuse (36%), and mood disorders (28%) [5].

There are many ways that HCV infection itself can cause psychiatric symptoms. These include inflammatory routes, direct brain neurotoxicity, alterations in metabolic and neurotransmitter pathways, and immune-mediated responses [6, 7].

Also, regarding the mechanism that underline neurocognitive changes in CHC patients, a review article suggested impairments in emotional learning, memory and how this ability, together with deficient inhibitory control, are core factors in different psychopathologies. As, these impairments caused dysfunctional behaviours, such as deficit in action control and motor inhibition, that are associated with psychopathological and psychiatric conditions, which are characterized by severe impulsivity problems that can determine significant impairment or distress (due to poor regulation and capacity of control, which can be intensified in the presence of emotional stimuli) and result in neurocognitive change [8]. Other suggested mechanism suggested altered emotion perception due to brain-damaged mainly amygdala and superior temporal sulcus dysfunctions [9]. Also, several studies have in fact provided evidence on the implication of frontal lobe circuitry in altered fear extinction features in mood disorders, defining how abnormalities in the ventromedial region of prefrontal cortex (vmPFC), whose smaller volume and altered activity patterns have been observed in these patients, affect memory and emotional learning capacity [10, 11]. Finally, previous review found the tryptophan–kynurenine metabolic pathway plays the most essential role in tryptophan metabolism, producing various endogenous bioactive molecules. The activation of the metabolic pathway is linked to the pathogenesis of a wide range of diseases [12,13,14,15,16,17].

Previously used interferon (INF)-based treatments had limited efficacy and high toxicity, as well as a high rate of psychiatric manifestations [18,19,20,21] and cognitive abnormalities [22]. Interferon-free therapeutic regimens with direct-acting antivirals (DAAs) have transformed HCV treatment by raising the likelihood of cure; referred to as sustained virological response (SVR) and lowering the time of treatment significantly [23, 24]. There are no flu-like symptoms, depression, or suicidal thoughts associated with DAAs because they are not inflammatory cytokines like IFN and ribavirin (RBV). DAAs have been found to have a milder side effect profile than earlier HCV medications [25] and improved patient-reported outcomes (PROs) [26, 27].

To our knowledge, a few studies have been conducted to examine the effects of DAA medication on individuals with chronic hepatitis C (CHC) and mental problems [28]. The majority of the data came from patient-reported outcomes, which were mostly gathered during observational trials on treatment tolerance and adherence [29,30,31].

Despite effectiveness studies with DAAs that include patients with psychiatric manifestations [31, 32], few researches have particularly addressed psychiatric symptoms in DAA treatment [33, 34].

With repeated observations at baseline and after 12 weeks post-treatment, this study aims to assess and compare the impact of DAAs on cognitive functions and psychiatric disorders in CHC patients. This is the only study that we are aware of that compares the psychological side effects and cognitive function of DAA treatment in CHC patients, non-cirrhotic and cirrhotic.

Methods

This prospective study comprised 40 treatments naive CHC patients from May 2020 to April 2021. They were admitted to the Tropical Medicine and Gastroenterology Department at Assiut University's Al-Rajhi Liver Hospital in Assiut, Egypt. Patients were assigned to a 3-month regimen of sofosbuvir/daclatasvir for HCV treatment with or without ribavirin. They were separated into non-cirrhotic (n = 20) and cirrhotic (n = 20). The control people (n = 10) were chosen at random from the outpatient clinic and relatives who had been admitted to our hospital. They were found to have no symptoms or indicators of chronic liver disease, no psychological issues, negative hepatitis serological markers (HBs Ag, Anti-HCV), and normal abdominal ultrasonography. They were eligible patients were diagnosed with CHC infection based on persistent or intermittent elevations in serum transaminase levels for more than 6 months with the presence of anti-HCV antibodies and serum HCV RNA [35]. Participants were classified as non-cirrhotic or cirrhotic based on the severity of hepatic fibrosis as determined by clinical findings, fibrosis-4 (FIB-4) score, and imaging including transient elastography (TE, FibroScan, Echosens, Paris, France) [36, 37]. Cirrhosis was determined by FIB-4 score > 3.25 for advanced fibrosis/cirrhosis and TE with a liver stiffness ≥ 12.5 kPa [38].

Child–Pugh classification [39] was used to determine the severity of liver cirrhosis, and those in Classes B and C were excluded from this study. We also excluded patients with non-hepatitis C-related liver diseases, co-infection with human immunodeficiency virus (HIV) or Hepatitis B (HBV), a history of treatment with pegylated interferon or DAAs, hepatocellular carcinoma, pregnancy, comorbid condition, such as diabetes mellitus, hypertension, chronic renal disease, overt hepatic encephalopathy, and other psychiatric or neurological disorders, as well as patients who had undergone liver transplantation.

For 12 weeks, non-cirrhotic group were given DAA therapy in the form of a once-daily oral dose of sofosbuvir (400 mg) and daclatasvir (60 mg) in combination with ribavirin (1000–1200 mg) according to body weight (for cirrhotic group only) based on the protocol provided by the National Committee for Control of Viral Hepatitis in Egypt (National Community College Hispanic Council’s (NCCHC)guidelines for the management of adult patients with HCV infection) available According to the EASL Recommendations for Treatment of Hepatitis C 2016, the combination of sofosbuvir and daclatasvir is still an appropriate option [40,41,42]. All subjects were followed up 3 months after finishing DAA therapy to assess treatment response and re-evaluate their neuropsychological tests. Non-detectable HCV-RNA in serum at 12 weeks post-treatment (SVR12) was characterized as a sustained virological response (SVR) [43].

The sample size was calculated using G power software version 3.1.3 [44], using ANOVA test for comparing difference in mean of depression score between three independent mean, effect size 0.5(assumed effect size). Difference of between 3 groups under the study), alpha error prob 0.05, power (1-beta error prob) 0.80. The required sample size was total 42 patients and it was raised to 50 patients in each group.

Before beginning DAA therapy, all participants underwent a thorough medical and psychiatric assessment, which included sex, age, residence, marital status, educational level, employment, history of psychiatric illness, medical or neurological disease, and drug history.

Laboratory investigations included complete blood count, liver function tests, HCV antibodies using an Ortho HCV Version 3.0 ELISA (Ortho Diagnostics Systems, Raritan, NJ, USA), and quantitative HCV-RNA using an Artus HCV-RG-RT-PCR kit (Qiagen, Hamburg, Germany) according to the manufacturer's instructions.

All individuals were subjected to Socio-economic Status Scale and neuropsychological tests, including Hamilton Depression Rating Scale, Hamilton Anxiety Rating Scale, and Cognitive Abilities Screening Instruments (CASI).

Social-economic Status Scale [45]: It was composed of three scores: occupation, education, and social class. The last score comprised three factors: income, crowding index, and sanitation score. The total socio-economic score was 23, with scores of 19 + indicating a high socio-economic standard, 15–19 indicating a middling socio-economic standard, and less than 15 indicating a poor socio-economic standard.

Hamilton Depression Rating Scale (HDS) [46]: It was a clinical rating scale used to determine the severity of depression. The overall score was calculated by adding the scores for each item (0–4) (symptom was absent, mild, moderate, or severe). The range of scores was 0 to 54.

Hamilton Anxiety Rating Scale (HAS) [47]: It was made up of 14 items used to assess and measure the degree of anxiety. Seven items dealt with psychic anxiety, while the remaining seven dealt with somatic anxiety. Each item was assigned to a collection of symptoms categorized by nature and rated on a 5-point scale ranging from 0 (not present) to 4 (very severe), with higher scores indicating more severe anxiety. Anxiety levels varied from 0 to 56 on a scale of 0 to 56.

Cognitive Abilities Screening Instruments (CASI) [48]: It had 25 test items. They tested orientation, old and recent memory, language skills, list-making ability, abstract thinking, and judgment. The CASI total score was calculated by adding the results from each of the nine domains. The total score ranged from 0 to 100. A greater overall cognitive function was indicated by a higher score.

Statistical analysis

All statistical analyses were performed using the Statistical Package for the Social Sciences; version 26 (IBM Corp., Armonk, NY, USA). To describe descriptive data, mean ± standard deviation (SD) or frequencies and percentages were employed. The Chi-square test (χ2) or Fisher’s exact probability test was used to compare categorical variables. The Student’s t-test and ANOVA were used to analyse quantitative data. P-values less than 0.05 were used to evaluate statistical significance.

Results

Forty patients with newly diagnosed chronic hepatitis C (20 non-cirrhotic and 20 cirrhotic) were included. Their mean age was 43.5 ± 10.6 years and male sex was predominant (57.5%). The majority were rural (67.5%), unemployed (65%) and illiterate (55%). In addition, ten healthy controls (five males and 5 females with mean age of 42.9 ± 9.9) were enrolled in the study. Further sociodemographic characteristics of the studied population and their subgroups are summarized in Table 1, where no significant differences regarding age, gender, residence, education, occupation, and socio-economic level were found between different groups.

Table 1 The sociodemographic characteristics among studied groups

Those CHC patients received DAA therapy (sofosbuvir and daclatasvir ± ribavirin for 12 weeks), and they were followed up 3 months after the end of therapy, where all of the patients achieved SVR (responders).

Comparison between CHC-non-cirrhotic patients and controls regarding neuropsychological and neurocognitive changes are summarized in Table 2. Before starting the DAA therapy (pre-treatment), both hepatic patients and controls had no symptoms of anxiety or depression as measured by HAS and HDS, however, the mean values of these scores were significantly higher in patients than controls (P < 0.001 for both). On the other hand, the mean values of total CASI (P = 0.012) and its components including short-term memory (P = 0.001), and abstract thinking and judgment (P = 0.002) were significantly lower in those patients than controls.

Up to 50% of HCV patients without additional comorbidities had cognitive impairment, and the degree of this impairment was associated with the degree of fibrosis [49]. Chronic HCV infection is typically accompanied with deficits in concentration, attention, working memory speed, and other higher executive functions. Direct neurotoxicity and inflammation [50, 51] as well as hepatic encephalopathy (HE) in cirrhotic patients have been theorized to contribute to cognitive impairment [49, 51]. Following DAAs, on comparison between pre- and post-treatment neuropsychological and neurocognitive changes in non-cirrhotic patients (Table 2), we found that significantly decreasing HAS and HDS scores in post-treated patients (P < 0.001 for both, Table 2) (P = 0.031). Moreover, we found significantly raised values of total CASI score (P = 0.001) and its components including short-term memory (P = 0.001), concentration/mental manipulation (P = 0.033) and abstract thinking and judgment (P = 0.024) in non-cirrhotic patients after receiving DAA therapy (Table 3). Furthermore, these post-treatment increasing values in CASI score and its component were compatible to that of controls where, no statistically significant differences between controls and non-cirrhotic group following DAAs as shown in Table 2.

Table 2 Distribution of depression, anxiety, and cognitive function among non-cirrhotic and control groups
Table 3 Distribution of depression, anxiety, and cognitive function among cirrhotic and control groups

Before starting the DAA therapy (pre-treatment), despite the mean HAS and HDS values within normal ranges, they were significantly higher in patients than controls (P < 0.001 for both). In addition, the mean values of total CASI (P = 0.012) and its components including long-term memory (P = 0.001), short-term memory (P < 0.001), attention (P = 0.015), concentration (P = 0.012), orientation (P = 0.007), language (P = 0.003), and abstract thinking and judgment (P < 0.001) were significantly lower in cirrhotic patients than controls.

Following DAAs and SVR achievement, HDS and HAS scores had significantly decreased values in post-treated cirrhotic patients versus pre-treated ones (P < 0.001 for both, Table 3) but these changes were still impaired in post-treated patients than controls (P < 0.001 for HDS and P = 0.034 for HAS, Table 3). Furthermore, we found significantly elevated values of total CASI score (P = 0.01) and its components including long-term memory (P = 0.015) short-term memory (P < 0.001), and concentration (P = 0.024), in cirrhotic patients after receiving DAA therapy (Table 3). However, these post-treatment changes in cirrhotic group were still impaired comparing to controls where, post-treated patients had significantly lower values of CASI (P = 0.02), long-term memory (P = 0.004), short-term memory (P = 0.007), orientation (P = 0.006), language (P = 0.002) visual construction/drawing (P = 0.05), list-generation fluency (P = 0.05) and abstract thinking and judgment (P = 0.001) than controls as shown in Table 3.

Discussion

This study aimed to assess and compare the impact of DAAs on cognitive functions and psychiatric disorders in CHC patients. We had two observations at baseline and after 12 weeks post-treatment. Before therapy, cirrhotic patients had higher depression levels than non-cirrhotic patients, but these scores were significantly lower following treatment in both groups. In contrast, anxiety levels were nearly identical before therapy and dropped in both groups after treatment (Figs. 1, 2, 3).

Fig. 1
figure 1

Total CASI score among the studied groups

Fig. 2
figure 2

Total HAS score among the studied groups

Fig. 3
figure 3

Total HADI score among the studied groups

In both cirrhotic and non-cirrhotic groups, anxiety and depression scores reduced markedly after treatment. Anxiety appears to be more sensitive to treatment response in the HCV group. The considerable treatment success with DAAs may be linked to decreased anticipatory anxiety [52, 53].

Treatment with DAAs may permanently eliminate the HCV etiological component [53]. According to prior research, DAAs for example SOF/DCV, SOF/LD, SOF/VEL/RBV, SOF/DCV/RBV or SOF/LDVOBV/PTV/r + DSV regimens are well-tolerated and has high adherence with a low burden of psychiatric side effects [54, 55].

Depression is widespread in CHC patients treated with IFN [21, 22, 56], and antidepressant medicines are known to relieve depressive symptoms in such circumstances [53]. DAA treatment is superior to IFN treatment in terms of avoiding depression [56]. We employed DAA instead of IFN for our patients in this study, removing a well-known risk factor for depression. Regardless of interferon treatment, depression has been identified as a risk factor for HCV patients [53]. The extrahepatic symptoms of HCV in the central nervous system were linked to this connection [57, 58].

In HCV-infected patients, psychiatric problems and emotional distress are widespread. The elimination of HCV may reduce the number of people who require psychiatric care. As a result of treatment with DAA, patients' depression and anxiety levels decreased, and the number of patients requiring psychiatric treatment decreased [53, 59].

As a result, the reduction in depression could be attributed to the absence of HCV in this trial.

Non-cirrhotic patients had significantly lower CASI score and subscales before therapy compared to controls. These scores improved after DAAs and achievement of SVR with significant differences in the mean concentration subscale scores including short-term memory, concentration, abstract thinking and total CASI. On the other hand, cirrhotic patients had lower CASI score and subscales before and after therapy compared to controls. Short-term, long-term memories, concentration, and total CASI all improved after DAA therapy. However, these post-treatment changes in cirrhotic were still impaired comparing to controls including total CASI, long-term, short-term memories, orientation, language, visual construction/drawing, list-generation fluency and abstract thinking and judgment than controls. No statistically significant differences in CASI score and its subscale values between non-cirrhotic group and controls following DAAs indicating that treatment of early stages of CHC patients; non-cirrhotic, is important to regain the normal cognitive function. Our results are compatible with previous studies that documented the efficacy of DAAs in improving cognitive outcomes irrespective severity of liver disease [55, 59,60,61].

Previous studies have found that chronic HCV infection and non-cirrhotic and cirrhotic liver disease influence brain functions as attention, concentration, and information processing speed even before therapy begins [7, 61,62,63]. The findings of these studies were unclear as to whether HCV has a direct influence on brain function or if the impairment is mediated by chronic liver illness [22].

The ability to concentrate and the speed of memory processes were significantly impaired in patients with chronic HCV infection compared to healthy controls, according to previously published results suggesting that HCV-associated neurocognitive decline may be reversible after viral clearance [64]. Healthy controls and previous HCV patients who had eradicated the virus, on the other hand, showed no difference in neurocognitive ability [65].

Similar to other studies suggest that the improvement in several measures of cognitive function after HCV treatment is due to the influence of HCV on cognitive performance rather than psychopathology [22, 61, 66].

In addition, previous researches reported that treatment with sofosbuvir drug and simeprevir or daclatasvir drug improved cognitive performance, especially in relation to memory [55, 60].

In this study, we observed cognitive dysfunction such as impaired concentration in CHC patients. It is worth noting that at the end of the treatment term, all of the patients in this study were PCR negative, indicating an excellent serological viral response. These findings support virological hypotheses for HCV-related cognitive impairment, which suggest that HCV disease processes disrupt brain functions and impair cognition [52].

Several investigations have revealed evidence of HCV replication in the brain, which supports this notion [64, 67].

Furthermore, HCV infection has been linked to a reduction in the generation of brain-derived neurotrophic factor [68, 69]. This factor is hypothesized to be involved in the pathophysiology of cognitive and mood disorders because it plays a critical role in regeneration and neurogenesis [70]. Treating HCV with direct-acting antivirals (DAAs) could improve comorbid psychiatric disorders. However, it would be beneficial to examine the implementation of new therapeutic techniques to treat comorbid psychiatric disorders, such as non-invasive brain stimulation, that operate to ameliorate the symptoms of mental and neurological disorders [71,72,73,74].

Limitations and future directions

One of the limitations of this study was the small size of the study population. The follow-up duration was short; longer follow-up durations may provide better knowledge about psychiatric disorders. Prospective cohort studies are needed to evaluate the pretreatment and post-treatment neurocognitive changes that may occur among CHC patients to understand the impact of morphometric and functional cerebral changes in those patients before and after SVR.

In conclusion, CHC patients' anxiety, depression, and cognitive function partially improved after DAA therapy. Besides improving the status of CHC, reversibility of cognitive dysfunction in non-cirrhotic patients may indicate the importance of treatment in early stages of liver disease.

Availability of data and materials

All data generated or analysed during this study are available from corresponded on request.

Abbreviations

DAAs:

Direct-acting antivirals

CASI:

The Cognitive Abilities Screening Instruments

CHC:

Chronic hepatitis C

HCV:

Hepatitis C virus

INF:

Interferon

SVR:

Sustained virological response

NCCHC:

National Community College Hispanic Council’s

HIV:

Human immunodeficiency virus

HBV:

Hepatitis B virus

HDS:

Hamilton Depression Rating Scale

HAS:

Hamilton Anxiety Rating Scale

References

  1. Lavanchy D. Evolving epidemiology of hepatitis C virus. Clin Microbiol Infect. 2011;17(2):107–15.

    Article  CAS  PubMed  Google Scholar 

  2. Adinolfi LE, Nevola R, Lus G, Restivo L, Guerrera B, Romano C, et al. Chronic hepatitis C virus infection and neurological and psychiatric disorders: an overview. World J Gastroenterol. 2015;21(8):2269–80.

    Article  PubMed  PubMed Central  Google Scholar 

  3. Forton DM, Allsop JM, Main J, Foster GR, Thomas HC, Taylor-Robinson SD. Evidence for a cerebral effect of the hepatitis C virus. The Lancet. 2001;358(9275):38–9.

    Article  CAS  Google Scholar 

  4. Mazzaro C, Quartuccio L, Adinolfi LE, Roccatello D, Pozzato G, Nevola R, et al. A review on extrahepatic manifestations of chronic hepatitis C virus infection and the impact of direct-acting antiviral therapy. Viruses. 2021;13:11.

    Article  Google Scholar 

  5. Carithers RL Jr, Sugano D, Bayliss M. Health assessment for chronic HCV infection: results of quality of life. Dig Dis Sci. 1996;41(12 Suppl):75s–80s.

    Article  PubMed  Google Scholar 

  6. Tully DC, Hjerrild S, Leutscher PD, Renvillard SG, Ogilvie CB, Bean DJ, et al. Deep sequencing of hepatitis C virus reveals genetic compartmentalization in cerebrospinal fluid from cognitively impaired patients. Liver Int. 2016;36(10):1418–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Forton DM, Allsop JM, Main J, Foster GR, Thomas HC, Taylor-Robinson SD. Evidence for a cerebral effect of the hepatitis C virus. Lancet. 2001;358(9275):38–9.

    Article  CAS  PubMed  Google Scholar 

  8. Battaglia S, Serio G, Scarpazza C, D’Ausilio A, Borgomaneri S. Frozen in (e)motion: How reactive motor inhibition is influenced by the emotional content of stimuli in healthy and psychiatric populations. Behav Res Ther. 2021;146: 103963.

    Article  PubMed  Google Scholar 

  9. Battaglia S, Fabius JH, Moravkova K, Fracasso A, Borgomaneri S. The neurobiological correlates of gaze perception in healthy individuals and neurologic patients. Biomedicines. 2022;10(3):627.

    Article  PubMed  PubMed Central  Google Scholar 

  10. Battaglia S, Harrison BJ, Fullana MA. Does the human ventromedial prefrontal cortex support fear learning, fear extinction or both? A commentary on subregional contributions. Mol Psychiatry. 2022;27(2):784–6.

    Article  PubMed  Google Scholar 

  11. Battaglia S, Thayer JF. Functional interplay between central and autonomic nervous systems in human fear conditioning. Trends Neurosci. 2022;45(7):504–6.

    Article  CAS  PubMed  Google Scholar 

  12. Tanaka M, Vécsei L. Monitoring the kynurenine system: Concentrations, ratios or what else? Adv Clin Exp Med. 2021;30(8):775–8.

    Article  PubMed  Google Scholar 

  13. Avan R, Sahebnasagh A, Hashemi J, Monajati M, Faramarzi F, Henney NC, et al. Update on statin treatment in patients with neuropsychiatric disorders. Life. 2021;11(12):1365.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Komatsu H, Watanabe E, Fukuchi M. Psychiatric neural networks and precision therapeutics by machine learning. Biomedicines. 2021;9(4):403.

    Article  PubMed  PubMed Central  Google Scholar 

  15. Spekker E, Tanaka M, Szabó Á, Vécsei L. Neurogenic inflammation: the participant in migraine and recent advancements in translational research. Biomedicines. 2022;10(1):76.

    Article  CAS  Google Scholar 

  16. Tanaka M, Vécsei L. Editorial of Special Issue “Crosstalk between Depression, Anxiety, and Dementia: Comorbidity in Behavioral Neurology and Neuropsychiatry”. Biomedicines. 2021;9(5):517.

  17. Tanaka M, Tóth F, Polyák H, Szabó Á, Mándi Y, Vécsei L. Immune influencers in action: metabolites and enzymes of the Tryptophan-Kynurenine Metabolic Pathway. Biomedicines. 2021;9(7):734.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Udina M, Castellví P, Moreno-España J, Navinés R, Valdés M, Forns X, et al. Interferon-induced depression in chronic hepatitis C: a systematic review and meta-analysis. J Clin Psychiatry. 2012;73(8):1128–38.

    Article  CAS  PubMed  Google Scholar 

  19. Evon DM, Esserman DE, Howell MA, Ruffin RA. Pegylated interferon pharmacokinetics and self-reported depressive symptoms during antiviral treatment for chronic hepatitis C. Pharmacopsychiatry. 2014;47(6):195–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. AASLD/DSA HCV guidance panel. Hepatitis C guidance: AASLD-IDSA recommendations for testing, managing, and treating adults infected with hepatitis C virus. Hepatology. 2015;62(3):932–54.

    Article  Google Scholar 

  21. Sahar M, Hassany AMD, Hossam K, Gellan KA, Haidi KAR, Ehab FM. Prevalence and Risk Factors of Depression and Anxiety in Hepatitis C Patients Receiving Pegylated Interferon Alpha (IFN-a). Int J Curr Microbiol Appl Sci. 2017;6(10):484–93.

    Article  Google Scholar 

  22. Hassaan SH, Darwish AM, Khalifa H, Ramadan HKA, Hassany SM, Ahmed GK, et al. Assessment of cognitive functions and psychiatric symptoms in hepatitis C patients receiving pegylated interferon alpha and ribavirin: A prospective cohort study. Int J Psychiatry Med. 2019;54(6):424–40.

    Article  PubMed  Google Scholar 

  23. Adinolfi LE, Petta S, Fracanzani AL, Coppola C, Narciso V, Nevola R, et al. Impact of hepatitis C virus clearance by direct-acting antiviral treatment on the incidence of major cardiovascular events: a prospective multicentre study. Atherosclerosis. 2020;296:40–7.

    Article  CAS  PubMed  Google Scholar 

  24. Shahid I, Ibrahim MM. All oral interferon-free direct-acting antivirals as combination therapies to cure hepatitis C. Curr Mol Med. 2018;18(7):409–35.

    Article  CAS  PubMed  Google Scholar 

  25. Sulkowski MS, Vargas HE, Di Bisceglie AM, Kuo A, Reddy KR, Lim JK, et al. Effectiveness of Simeprevir Plus Sofosbuvir, With or Without Ribavirin, in Real-World Patients With HCV Genotype 1 Infection. Gastroenterology. 2016;150(2):419–29.

    Article  PubMed  Google Scholar 

  26. Younossi ZM, Stepanova M, Marcellin P, Afdhal N, Kowdley KV, Zeuzem S, et al. Treatment with ledipasvir and sofosbuvir improves patient-reported outcomes: Results from the ION-1, -2, and -3 clinical trials. Hepatology. 2015;61(6):1798–808.

    Article  CAS  PubMed  Google Scholar 

  27. Younossi ZM, Stepanova M, Jacobson IM, Asselah T, Gane EJ, Lawitz E, et al. Sofosbuvir and velpatasvir with or without voxilaprevir in direct-acting antiviral-naïve chronic hepatitis C: patient-reported outcomes from POLARIS 2 and 3. Aliment Pharmacol Ther. 2018;47(2):259–67.

    Article  CAS  PubMed  Google Scholar 

  28. Gragnani L, Lorini S, Martini L, Stasi C, Visentini M, Petraccia L, et al. Rapid improvement of psychiatric stigmata after IFN-free treatment in HCV patients with and without cryoglobulinemic vasculitis. Clin Rheumatol. 2022;41(1):147–57.

    Article  PubMed  Google Scholar 

  29. Marcellin P, Chousterman M, Fontanges T, Ouzan D, Rotily M, Varastet M, et al. Adherence to treatment and quality of life during hepatitis C therapy: a prospective, real-life, observational study. Liver Int. 2011;31(4):516–24.

    Article  CAS  PubMed  Google Scholar 

  30. Rowan PJ, Bhulani N. Psychosocial assessment and monitoring in the new era of non-interferon-alpha hepatitis C virus treatments. World J Hepatol. 2015;7(19):2209.

    Article  PubMed  PubMed Central  Google Scholar 

  31. Ho SB, Monto A, Peyton A, Kaplan DE, Byrne S, Moon S, et al. Efficacy of sofosbuvir plus ribavirin in veterans with hepatitis C virus genotype 2 infection, compensated cirrhosis, and multiple comorbidities. Clin Gastroenterol Hepatol. 2017;15(2):282–8.

    Article  CAS  PubMed  Google Scholar 

  32. Ioannou GN, Beste LA, Chang MF, Green PK, Lowy E, Tsui JI, et al. Effectiveness of sofosbuvir, ledipasvir/sofosbuvir, or paritaprevir/ritonavir/ombitasvir and dasabuvir regimens for treatment of patients with hepatitis C in the veterans affairs National Health Care System. Gastroenterology. 2016;151(3):457-71.e5.

    Article  CAS  PubMed  Google Scholar 

  33. Rowan PJ. What psychiatric screening and monitoring might be needed with the new generation of hepatitis C treatments? World J Virol. 2015;4(1):13–6.

    Article  PubMed  PubMed Central  Google Scholar 

  34. Sockalingam S, Sheehan K, Feld JJ, Shah H. Psychiatric care during hepatitis C treatment: the changing role of psychiatrists in the era of direct-acting antivirals. Am J Psychiatry. 2015;172(6):512–6.

    Article  PubMed  Google Scholar 

  35. Anwar W, Sarwar M, Hussain AB, Tariq WU, Saif M. Significance of occult HBV infection in patients with chronic hepatitis C. J Coll Physicians Surg Pak. 2006;16(3):192–5.

    PubMed  Google Scholar 

  36. Schuppan D, Afdhal NH. Liver cirrhosis. Lancet. 2008;371(9615):838–51.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Sandrin L, Fourquet B, Hasquenoph JM, Yon S, Fournier C, Mal F, et al. Transient elastography: a new noninvasive method for assessment of hepatic fibrosis. Ultrasound Med Biol. 2003;29(12):1705–13.

    Article  PubMed  Google Scholar 

  38. Foster GR, Afdhal N, Roberts SK, Bräu N, Gane EJ, Pianko S, et al. Sofosbuvir and Velpatasvir for HCV Genotype 2 and 3 Infection. N Engl J Med. 2015;373(27):2608–17.

    Article  CAS  PubMed  Google Scholar 

  39. Pugh RN, Murray-Lyon IM, Dawson JL, Pietroni MC, Williams R. Transection of the oesophagus for bleeding oesophageal varices. Br J Surg. 1973;60(8):646–9.

    Article  CAS  PubMed  Google Scholar 

  40. Ahmed OA, Safwat E, Khalifa MO, Elshafie AI, Fouad MHA, Salama MM, et al. Sofosbuvir Plus Daclatasvir in treatment of chronic hepatitis C Genotype 4 infection in a cohort of Egyptian patients: an experiment the size of Egyptian village. Int J Hepatol. 2018;2018:9616234.

    Article  PubMed  PubMed Central  Google Scholar 

  41. Poordad F, Schiff ER, Vierling JM, Landis C, Fontana RJ, Yang R, et al. Daclatasvir with sofosbuvir and ribavirin for hepatitis C virus infection with advanced cirrhosis or post-liver transplantation recurrence. Hepatology. 2016;63(5):1493–505.

    Article  CAS  PubMed  Google Scholar 

  42. EASL eAftSotL. EASL recommendations on treatment of Hepatitis C 2016. J Hepatol. 2017;66(1):153–94.

  43. Yoshida EM, Sulkowski MS, Gane EJ, Herring RW Jr, Ratziu V, Ding X, et al. Concordance of sustained virological response 4, 12, and 24 weeks post-treatment with sofosbuvir-containing regimens for hepatitis C virus. Hepatology. 2015;61(1):41–5.

    Article  CAS  PubMed  Google Scholar 

  44. Faul F, Erdfelder E, Lang AG, Buchner A. G*Power 3: a flexible statistical power analysis program for the social, behavioral, and biomedical sciences. Behav Res Methods. 2007;39(2):175–91.

    Article  PubMed  Google Scholar 

  45. Fahmy S, El-Sherbini AF. Determining simple parameters for social classifications for health research. Bull High Inst Public Health. 1983;13:95–108.

    Google Scholar 

  46. Hamilton M. Development of a rating scale for primary depressive illness. Br J Soc Clin Psychol. 1967;6(4):278–96.

    Article  CAS  PubMed  Google Scholar 

  47. Hamilton M. The assessment of anxiety states by rating. Br J Med Psychol. 1959;32:55.

    Article  Google Scholar 

  48. Evans DA, Beckett LA, Albert MS, Hebert LE, Scherr PA, Funkenstein HH, et al. Level of education and change in cognitive function in a community population of older persons. Ann Epidemiol. 1993;3(1):71–7.

    Article  PubMed  Google Scholar 

  49. Hilsabeck RC, Perry W, Hassanein TI. Neuropsychological impairment in patients with chronic hepatitis C. Hepatology. 2002;35(2):440–6.

    Article  PubMed  Google Scholar 

  50. Weissenborn K, Ennen JC, Schomerus H, Rückert N, Hecker H. Neuropsychological characterization of hepatic encephalopathy. J Hepatol. 2001;34(5):768–73.

    Article  CAS  PubMed  Google Scholar 

  51. Hilsabeck RC, Hassanein TI, Carlson MD, Ziegler EA, Perry W. Cognitive functioning and psychiatric symptomatology in patients with chronic hepatitis C. J Int Neuropsychol Soc. 2003;9(6):847–54.

    Article  PubMed  Google Scholar 

  52. Danilescu CM, Sandulescu DL, Pirlog MC, Streba CT, Rogoveanu I. Depressive and anxious symptoms in hepatitis c virus infected patients receiving DAA-Based Therapy. Diagnostics (Basel). 2021;11:12.

    Google Scholar 

  53. Kesen O, Kani HT, Yanartaş Ö, Aykut UE, Gök B, Gündüz F, et al. Evaluation of depression, anxiety and quality of life in hepatitis C patients who treated with direct acting antiviral agents. Turk J Gastroenterol. 2019;30(9):801–6.

    Article  PubMed  PubMed Central  Google Scholar 

  54. Ferenci P, Bernstein D, Lalezari J, Cohen D, Luo Y, Cooper C, et al. ABT-450/rombitasvir and dasabuvir with or without ribavirin for HCV. N Engl J Med. 2014;370(21):1983–92.

    Article  PubMed  Google Scholar 

  55. Kaur H, Dhiman RK, Kulkarni AV, Premkumar M, Singh V, Duseja AK, et al. Improvement of chronic HCV infection-related depression, anxiety, and neurocognitive performance in persons achieving SVR-12: A real-world cohort study. J Viral Hepat. 2022;29(5):395–406.

    Article  CAS  PubMed  Google Scholar 

  56. Dieperink E, Ho SB, Thuras P, Willenbring ML. A prospective study of neuropsychiatric symptoms associated with interferon-alpha-2b and ribavirin therapy for patients with chronic hepatitis C. Psychosomatics. 2003;44(2):104–12.

    Article  CAS  PubMed  Google Scholar 

  57. Carta MG, Hardoy MC, Garofalo A, Pisano E, Nonnoi V, Intilla G, et al. Association of chronic hepatitis C with major depressive disorders: irrespective of interferon-alpha therapy. Clin Pract Epidemiol Ment Health. 2007;3:22.

    Article  PubMed  PubMed Central  Google Scholar 

  58. Carta MG, Angst J, Moro MF, Mura G, Hardoy MC, Balestrieri C, et al. Association of chronic hepatitis C with recurrent brief depression. J Affect Disord. 2012;141(2–3):361–6.

    Article  PubMed  Google Scholar 

  59. Nardelli S, Riggio O, Rosati D, Gioia S, Farcomeni A, Ridola L. Hepatitis C virus eradication with directly acting antivirals improves health-related quality of life and psychological symptoms. World J Gastroenterol. 2019;25(48):6928–38.

    Article  PubMed  PubMed Central  Google Scholar 

  60. Gascon MRP, Benute GRG, Macedo EC, CapitÃo CG, Vidal JE, Smid J, et al. Cognitive assessment in patients with Hepatitis C submitted to treatment with Sofosbuvir and Simeprevir or Daclatasvir. Arq Neuropsiquiatr. 2020;78(6):342–8.

    Article  PubMed  Google Scholar 

  61. Kleefeld F, Heller S, Ingiliz P, Jessen H, Petersen A, Kopp U, et al. Interferon-free therapy in hepatitis C virus (HCV) monoinfected and HCV/HIV coinfected patients: effect on cognitive function, fatigue, and mental health. J Neurovirol. 2018;24(5):557–69.

    Article  CAS  PubMed  Google Scholar 

  62. Forton DM, Allsop JM, Cox IJ, Hamilton G, Wesnes K, Thomas HC, et al. A review of cognitive impairment and cerebral metabolite abnormalities in patients with hepatitis C infection. AIDS. 2005;19(Suppl 3):S53-63.

    Article  PubMed  Google Scholar 

  63. Weissenborn K, Tryc AB, Heeren M, Worthmann H, Pflugrad H, Berding G, et al. Hepatitis C virus infection and the brain. Metab Brain Dis. 2009;24(1):197–210.

    Article  PubMed  Google Scholar 

  64. Forton DM, Karayiannis P, Mahmud N, Taylor-Robinson SD, Thomas HC. Identification of unique hepatitis C virus quasispecies in the central nervous system and comparative analysis of internal translational efficiency of brain, liver, and serum variants. J Virol. 2004;78(10):5170–83.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Forton DM, Thomas HC, Murphy CA, Allsop JM, Foster GR, Main J, et al. Hepatitis C and cognitive impairment in a cohort of patients with mild liver disease. Hepatology. 2002;35(2):433–9.

    Article  PubMed  Google Scholar 

  66. Thein HH, Maruff P, Krahn MD, Kaldor JM, Koorey DJ, Brew BJ, et al. Improved cognitive function as a consequence of hepatitis C virus treatment. HIV Med. 2007;8(8):520–8.

    Article  CAS  PubMed  Google Scholar 

  67. Laskus T, Radkowski M, Bednarska A, Wilkinson J, Adair D, Nowicki M, et al. Detection and analysis of hepatitis C virus sequences in cerebrospinal fluid. J Virol. 2002;76(19):10064–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Modabbernia A, Ashrafi M, Keyvani H, Taslimi S, Poorkaveh A, Merat S, et al. Brain-derived neurotrophic factor predicts physical health in untreated patients with hepatitis C. Biol Psychiatry. 2011;70(5):e31–2.

    Article  CAS  PubMed  Google Scholar 

  69. Chen R, Liang FX, Moriya J, Yamakawa J, Sumino H, Kanda T, et al. Chronic fatigue syndrome and the central nervous system. J Int Med Res. 2008;36(5):867–74.

    Article  CAS  PubMed  Google Scholar 

  70. Hashimoto K, Shimizu E, Iyo M. Critical role of brain-derived neurotrophic factor in mood disorders. Brain Res Brain Res Rev. 2004;45(2):104–14.

    Article  CAS  PubMed  Google Scholar 

  71. Borgomaneri S, Battaglia S, Sciamanna G, Tortora F, Laricchiuta D. Memories are not written in stone: Re-writing fear memories by means of non-invasive brain stimulation and optogenetic manipulations. Neurosci Biobehav Rev. 2021;127:334–52.

    Article  PubMed  Google Scholar 

  72. Borgomaneri S, Battaglia S, Avenanti A, Pellegrino G. Don’t Hurt Me No More: State-dependent Transcranial Magnetic Stimulation for the treatment of specific phobia. J Affect Disord. 2021;286:78–9.

    Article  PubMed  Google Scholar 

  73. Tortella G, Selingardi PM, Moreno ML, Veronezi BP, Brunoni AR. Does non-invasive brain stimulation improve cognition in major depressive disorder? A systematic review. CNS Neurol Disord Drug Targets. 2014;13(10):1759–69.

    Article  PubMed  Google Scholar 

  74. Mutz J, Edgcumbe DR, Brunoni AR, Fu CHY. Efficacy and acceptability of non-invasive brain stimulation for the treatment of adult unipolar and bipolar depression: A systematic review and meta-analysis of randomised sham-controlled trials. Neurosci Biobehav Rev. 2018;92:291–303.

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

None.

Funding

This work was supported by a Grant from the Grant Office, Faculty of Medicine, Assiut University, Egypt (Grant No. 2016-10/30-016).

Author information

Authors and Affiliations

Authors

Contributions

GS: recruited participants, analysis, and interpreted data, and were the contributors in writing the manuscript. Gk: wrote the manuscript and analysis of data. EH, AS and SZ: recruited participants, helped in data entry, analyse, and generate result sheets and revised data interpretation and manuscript. NM and KE recruited participants, analysis, and interpreted data, and were the contributors in writing the manuscript. KO: recruited patients and made transcranial vascular assessment. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Ghaydaa A. Shehata.

Ethics declarations

Ethical approval and consent to participate

After a detailed explanation of the study's goal, methods, potential dangers, and side effects, each participant gave written informed consent to be included in the study. The Institutional Review Board of Assiut University approved the study protocol on date of October 2014 with authorization number 17200637.

Consent for publication

Not applicable.

Competing interests

The authors declare no conflicts of interests.

Additional information

Publisher's Note

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

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shehata, G.A., Ahmed, G.K., Hassan, E.A. et al. Impact of direct-acting antivirals on neuropsychiatric and neurocognitive dysfunction in chronic hepatitis C patients. Egypt J Neurol Psychiatry Neurosurg 58, 143 (2022). https://doi.org/10.1186/s41983-022-00568-5

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s41983-022-00568-5

Keywords