Sie sind auf Seite 1von 9

VIRAL IMMUNOLOGY

Volume 30, Number 9, 2017 Review


ª Mary Ann Liebert, Inc.
Pp. 1–9
DOI: 10.1089/vim.2017.0009

Hepatitis C Virus-Associated Extrahepatic Manifestations


in Lung and Heart and Antiviral Therapy-Related
Cardiopulmonary Toxicity
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

Syeda Zainab Ilyas,1,* Rabia Tabassum,1,* Haroon Hamed,2 Shafiq Ur Rehman,1 and Ishtiaq Qadri2

Abstract

Besides liver cirrhosis and hepatocellular carcinoma, chronic hepatitis C virus (HCV) infection is associated
with many extrahepatic manifestations (EHMs). HCV exhibits lymphotropism that is responsible for various
EHM. An important characteristic of HCV is escape from the immune system, which enables it to produce
chronic infections and autoimmune disorders along with accumulation of circulating immune complexes. These
EHMs have large spectrum, because they affect many organs such as heart, lungs, kidney, brain, thyroid, and skin.
HCV-related cardiac and pulmonary manifestations include myocarditis, cardiomyopathies, cardiovascular dis-
eases (i.e., Stroke, ischemic heart disease), chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis,
asthma, and interstitial lung diseases. This review discusses etiology and pathogenesis of HCV-associated cardiac
and pulmonary manifestations and how different genes, immune system, indirectly linked factors (mixed cryo-
globulinemia), liver cirrhosis, and antiviral treatment are involved in HCV-related heart and lung diseases,
however, their exact mechanism is not clear.

Keywords: chronic hepatitis C virus, extrahepatic manifestations, cardiac and pulmonary manifestations,
pathogenesis, mixed cryoglobulinemia, antiviral treatment

Introduction The HCV infects about 3% of world population, while 17–


37% HCV-infected people also suffer with HCV-related heart

H epatitis C virus (HCV) infection is a chronic blood-


borne disease that mainly affects the liver, but due to
lymphotropic nature of this virus, it also leads to many ex-
diseases (51). The most common cardiac manifestations are
cardiomyopathies, myocarditis, atherosclerosis, cardiovascu-
lar disorders (stroke and ischemic heart disease), coronary
trahepatic manifestations (EHMs) (7). Several studies found artery disease (CAD), and peripheral artery disease (PAD)
EHMs of the disease in 70–74% of HCV patients (24). One (28). Along with heart diseases, HCV-associated pulmonary
of the major attributes of HCV is its ability to escape from complications are also reported. HCV-related lung patholo-
activity of host immune system leading to chronic infec- gies could be the primary outcome of the infection, however,
tion. During chronic infections, accumulation of circulat- there are certain other pathologies such as pulmonary vas-
ing immune complexes (CIC) and activation of autoimmune culature and damage of pulmonary parenchyma attributed to
responses lead to extra hepatic manifestations (26). In addi- mixed cryoglobulinemia (MC), interferon therapy liver cir-
tion to this, certain EHMs are also associated with advanced rhosis (60), hepatopulmonary syndrome, and portopulmonary
liver disease. HCV being both hepatotropic and lymphotropic hypertension (3). The primary or direct outcomes of the HCV
virus, acts as a stimulator of the immune system (23). infection on lung pathologies include the chronic obstructive
Therefore, beside the ordinary cardiovascular risk factors pulmonary disease (COPD), idiopathic pulmonary fibrosis
(hypertension, obesity, diabetes, physical inactivity, smok- (IPF), asthma, and interstitial lung diseases (60).
ing, and high cholesterol), it acts as an indirect risk factor for In this study, we will discuss the etiology and pathogenesis
various cardiopulmonary diseases (10). Although the cases of HCV-associated cardiac and pulmonary manifestations
of HCV-related cardiac diseases are present all around the and how different genes, immune system, indirectly linked
globe, they are highly prevalent in Asia, Africa, and under- factors (diabetes and cryoglobulinemia), and antiviral treat-
developed countries (41). ment contribute to the HCV-related cardiopulmonary diseases.

1
Department of Microbiology and Molecular Genetics, University of the Punjab, Lahore, Pakistan.
2
Department of Biological Sciences, King Abdul Aziz University, Jeddah, Kingdom of Saudi Arabia.
*These two authors contributed equally to this work.
2 ILYAS ET AL.

HCV-Related Cardiomyopathies and Myocarditis (TNF-a) are the two major cytokines produced by macro-
phages, responsible for recruiting inflammatory cells and
Cardiomyopathies can be defined as a group of hetero- factors toward the myocardium (71). This mechanism
geneous diseases of myocardium. It includes idiopathic di- suggests that chronic HCV can act as a risk factor for the
lated, hypertrophic or restrictive disease cardiomyopathies, development of myocarditis and subsequently to cardio-
arrhythmogenic right ventricular cardiomyopathy, and in- myopathies (Fig. 1).
flammation of the myocardium or myocarditis (66). One Many studies have shown a stronger association between
of the major reasons of myocarditis is cardiotropic viruses increased expressions of TNF-a and depressed myocardial
(66) such as enteroviruses, coxsackievirus B, adenovirus, function with its increasing concentrations in plasma and
and parvovirus (52). Recently, a high prevalence of HCV myocardium of patients. The TNF-a binds to two different
infection has been noted in patients with hypertrophic car- receptors (TNF-a receptor 1 [TNFRI] and TNF-a receptor 2
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

diomyopathy, dilated cardiomyopathy (DCM), and myo- [TNFRII]) present on the surface of cardiomyocytes, and
carditis (49). Among all of the HCV-associated cardiac cause negative inotropic effects. In cardiac myocytes, con-
disorders, DCM is a relatively common disorder, which may traction and relaxation depend on cyclic release and re-
lead to severe heart failure. Besides this, HCV infection causes moval of calcium ions from the sarcoplasmic reticulum (84).
induction of proliferative stimuli that lead to the formation of Extracellular calcium ions enter the cell via L-type Ca+2
myocytes responsible for cardiomyopathy and hypertrophic channels during membrane depolarization and help in the
cardiomyopathy (HCM). HCM refers to a heart defect char- release of calcium ions from the sarcoplasmic reticulum.
acterized by increased thickness of the wall of the myocardium, The SR Ca+2/Mg+2-ATPase (SERCA) present in the sarco-
mostly left ventricle, causing functional impairment of the heart. plasmic reticulum of myocytes is involved in the calcium
It is possible that both direct viral cytotoxic and indirect uptake during muscular movement and higher concentra-
immune-mediated mechanisms of myocardial tissue damage tions of TNF-a restrict L-type Ca+2 channels and affect the
are crucial in the pathogenesis of viral-induced cardiac systolic phase by inhibiting high concentration of calcium
disease (40). Previously, immunological mechanisms were ions (87). It also affects the relaxation phase by inhibiting
described for chronic liver disease-mediated myocarditis SERCA (16).
and cardiomyopathies. Patients having chronic HCV- Another mechanism by which TNF-a affects the myo-
mediated myocarditis had normal serum levels of hepatic cardium is by stimulating the production of nitric oxide
transaminases, until the final stages of heart failure (51). In (NO), which acts as an endogenous inhibitor and has a
case of congestive hepatopathy, the main driver of this negative inotropic effect on the myocardium. In the pres-
condition is activation of cytotoxic T cell response in liver ence of proinflammatory cytokine (TNF-a), the concentra-
tissue. When protein products of HCV stimulate the toll-like tion of inducible nitric oxide synthase (iNOS), (calcium
receptors, CD4+ cell-mediated T helper-1 (Th1) response is independent, a nonexpressive isoform of NO synthase) is
induced, which results in release of interleukin-12 (IL-12), increased. Expression of iNOS in the myocardium causes
interferon (IFN)-a and -b from monocytes, macrophages, the decrease in the contractile function of the heart and
and neutrophils. Highly activated CD4+ cells produce IFN-c, isolated cardiomyocytes (71).
which stimulates the macrophages to produce proin- Autoimmunity is another important mechanism of HCV-
flammatory cytokines. IL-1 and tumor necrosis factor alpha linked myocarditis and DCM. The presence of some human

FIG. 1. Postulated behavior of the HCV-mediated immune activation, resulting development of myocarditis, which
ultimately leads to cardiomyopathies. DCM, dilated cardiomyopathy; HCM, hypertrophic cardiomyopathy; HCV, hepatitis
C virus; IFN-a, interferon alpha; IL-12, interleukin-12; Th1, T helper-1; TNF-a, tumor necrosis factor alpha.
HCV-ASSOCIATED EXTRAHEPATIC MANIFESTATIONS 3

leukocyte antigen (HLA) and non-HLA haplotypes ge- HCV infection increases the risk of developing PAD.
netically predispose some HCV patients toward HCV- PAD responsible for the development of calcium and fat
associated cardiomyopathies (71). All human HLA class II plaques in the arteries, which upon increase in size lead to
loci (HLA-DP, HLA DQ. HLA-DR) show polymorphism, atherosclerosis (32). Some data from a Taiwanese study
which affects the epitope’s affinity to bind with different indicated that HCV-infected patients are at higher risk of
peptides involved in the pathogenesis of some autoimmune developing PAD compared with noninfected patients. The
diseases (11). In HCV-related cardiomyopathies, certain incidence of PAD development in HCV-infected patients
genes have been found to play their roles. The gene for occurs in the first and third years of disease (12). HCV
human major histocompatibility complex (MHC) is located chronic infection and related inflammation may be associ-
on the short arm of chromosome 6 and encodes for several ated with several unfavorable pathologic conditions, in-
protein products involved in immune function, including cluding atherosclerosis (6). Chronic HCV infection causes
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

complement, TNF-a, and HLA complex. Any polymor- liver fibrosis, which induces metabolic changes that are in-
phism in this gene is proposed as determinant for the sus- volved in the production of atherosclerosis (39). In HCV-
ceptibility to various diseases. infected patients, atherosclerosis is the main cause of strokes
MHC class II genes may play their roles, in conferring and heart attacks. It has been reported in different studies
susceptibility to HCV infection, and may influence the de- that HCV infection is an independent risk factor for strokes,
velopment of different phenotypes of cardiomyopathy (62). cerebrovascular diseases, and ischemic heart diseases. HCV-
Recently, several studies have reported that dilated and hy- infected patients showed a marked association between in-
pertrophic cardiomyopathies are associated with certain al- flammatory markers and atherosclerosis (12).
leles of the HLA system. For example, the HLA-DPB1*0901 The liver is a major inflammatory regulator; it regulates both
and HLA-DRB1*1201 have been reported to be more preva- local and systemic inflammation by the production of mole-
lent in chronic HCV-infected patients who develop DCM, and cules having inflammatory-inducing properties. When HCV
HLA DRB1*0901 and DQB1*0303 haplotypes are more infects liver, inflammatory cascade begins. Proinflammatory
prevalent in chronic HCV-infected patients with hypertrophic and inflammatory cytokines, including IL 1-b, TNF-a, and -b,
cardiomyopathy (50). Another study described that HCV- are produced, which cause local inflammation and fibrosis
linked DCM was due to alleles of non-HLA genes rather than (90). Steatosis is another factor that has contributed in the
with those of the HLA genes. Specifically, a group of in- mechanism of EHM of HCV via the production of inflam-
vestigators mapped the gene factor, responsible for HCV- matory markers such as IL-6 and TNF-a. These molecules play
related DCM at non-HLA gene locus spanning from the roles in the inhibition of insulin signaling pathway, and adi-
NF-kappa-B inhibitor-like protein 1 (NFKBIL1) to the MHC ponectin levels subsequently results in the progression of
class I chain-related protein A (MICA) gene loci within the steatosis and IR (43). In some HCV patients, a high level of
MHC class III-class I boundary region (78). inflammatory markers due to steatosis is a major cause of CAD
(9). The other mechanism that causes inflammation due to
HCV is oxidative stress. Imbalance between antioxidants and
Cardiovascular Diseases
reactive oxygen species is linked with local and systemic in-
Chronic hepatitis C (CHC) is a major independent risk flammation. Other factors that are involved in oxidative stress
factor for the development of cardiovascular diseases damage include necrosis, tissue regeneration, apoptosis, is-
(CVDs), including atherosclerosis, cardiovascular disorders chemia, and fibrinogenesis (89) (Fig. 2).
(stroke and ischemic heart disease), CAD, and PAD (15).
According to the World Health Organization, stroke and
Idiopathic Pulmonary Fibrosis
ischemic heart disease are the two major causes of death
worldwide. People suffering from chronic HCV have in- IPF is a chronic disease of the lungs that includes low-
creased risk of CAD, PAD, and cerebrovascular disease grade inflammation of alveoli, leading to the derangement of
(28). CHC also develops the major risk factors for other lung interstitium. IPF usually develops in cases of chronic
metabolic diseases such as insulin resistance (IR), diabetes HCV infection (19). It is characterized by the accumulation
mellitus (DM), atherosclerosis, and hypertension (39). of extracellular matrix, impaired gas exchange, and worsen-
It has been assumed that HCV promotes atherogenesis ing dyspnea (58,88). Various pathologic features of IPF in-
and causes CVDs by stimulating different biological clude epithelial cell injury, diffuse alveolar damage, variable
mechanisms. These mechanisms involve liver steatosis, fi- inflammation, lung damage and fibrosis, and temporal and
brosis, diabetes, IR, HCV colonization and proliferation spatial heterogeneity in lung areas (27,58). The precise cause
within arterial walls, excessive and imbalance production of for the IPF is unknown (67), however, HCV is said to be a
cytokines, oxidative stress, endotoxemia, and hyperhomo- triggering agent for inflammation in the lung interstitium that
cysteinemia (89). Among the various CVDs, HCV mainly leads to lung fibrosis (23,64). The relationship between HCV
promotes the atherosclerosis. Recent studies indicated that infection and IPF was confirmed by a retrospective study,
HCV is associated with carotid atherosclerosis and carotid which reports 11% of patients with HCV-induced cirrhosis,
intima-media thickness independent of other risk factors of who also showed obstructive airway disease (4).
CVDs (35). In a cross-sectional study, association between There are many factors that contribute to the chronic lung
HCV and carotid artery plaques was indicated by the pres- diseases such as persistent antigens, viral infection, lung
ence of anti-HCV antibodies and HCV core protein in the injury, and immune cell activation (47). HCV-related IPF is
serum of HCV-affected patients (36). HCV infection is as- said to be an outcome of occult infection, in which virus (an
sociated with coronary artery plaques and high levels of inflammatory agent) disrupts the normal healing process of
HCV-mediated plaque calcification. the lung, resulting in lung fibrosis (70). In case of IPF, both
4 ILYAS ET AL.
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

FIG. 2. Factors related to HCV that causes chronic inflammation, which plays a role in hepatic and systemic manifes-
tations. HCC, hepatocellular carcinoma.

lytic and latent viruses have found to play their roles in IPF+HCV patients were improved when treated with anti-
causing lung fibrosis. In case of IPF, HCV RNA is also virals, valacyclovir and ganciclovir (58,88). In addition to
detectable in bronchoalveolar lavage (BAL) fluid, and in this, IPF+HCV patients have higher mean age suggestive of
lung biopsies of patients, indicating the direct involvement the immunosenescence, particularly in case of viral infec-
of the virus in IPF (8), however, the presence of HCV-RNA tions (25,59), along with different spatial or temporal mor-
at lower quantity is indicative of virus airway transmission; phology of the lung indicative of reminiscent infection
as no difference was found between the T cell and B cell (27,58). Besides this, a large population of activated lym-
populations among the cases and controls (34). Certain other phocytes is observed in the BAL fluids of IPF patients with
immune cells, such as neutrophil, eosinophil, macrophages, enhanced expression of the host defense genes (58,59,86).
mast cells, and fibroblast cause some nonspecific inflam-
matory reactions (47), ultimately resulting in lung fibrosis.
Chronic Obstructive Pulmonary Disease
In HCV patients, immune complexes found to have no role
in causing the IPF, because fluorescence microscopy of the COPD is characterized by a chronic inflammation of the
lung biopsies has not revealed any immune complex. lung parenchyma tissue with varying degree of airway ob-
There was a high prevalence of anti-HCV antibodies struction (60). COPD can be a result of spontaneous pul-
against the C100-3 antigen (a polypeptide containing 363 monary reactions, or sometimes it develops in response to
amino acids) of HCV in the sera of IPF patients (82). In HCV- certain medical treatment. In 1990, COPD was considered
related IPF, virus-induced immune-mediated fibroblast acti- as the sixth most common cause of death that is estimated to
vation is thought to have a key role in pathogenesis (17), as be the third most common cause of death in 2020 (79).
these fibroblast cells are activated after the viral infection. COPD is characterized by the fixed airflow restriction and
With these activated fibroblasts, epithelial cells, mast cells, forced expiratory volume (FEV) as these factors can be used
monocytes, and macrophages have found to release higher as prognostic markers for the disease (79). Various viral
levels of IL-10, IL-13, and IL-4 (47). IL-4 and IL-13 act as infections aggravate the previously caused COPD and
proinflammatory mediators and either directly or indirectly asthma (58). HCV can either initiate or increase the severity
affect the fibroblast activation (85). In this type 2 cytokine of the disease, as HCV+COPD have reduced values for
environment, an increase in the levels of IL-4 and IL-13 results pulmonary function tests, including FEV1 and carbon
in the production of increased extracellular matrix proteins and monoxide diffusion capacity of the lungs (DLCO) values,
fibroproliferation. Excessive deposition of the extracellular compared with the HCV-COPD patients (79).
matrix proteins in the lungs causes pulmonary fibrosis (85). HCV exacerbates the COPD directly by causing chronic
IPF can also be an outcome of autoantibodies produced in inflammation due to latent viral infection and indirectly by
response to the HCV infection as there is a relationship triggering the HCV-specific T-Lymphocytic response. Be-
between the Jo-1 autoantibody and IPF (83). sides this, HCV infection reduces the lung functions in pa-
From the published data, various evidences have tients suffering from COPD. In patients with COPD, the
been provided for viral etiology of IPF. The conditions of disease conditions mainly worsened due to the chronic HCV
HCV-ASSOCIATED EXTRAHEPATIC MANIFESTATIONS 5

conditions. In a study, various anthropological factors such RNA (70) in patients of HCV+MC further indicates the
as mean age, sex, exposure to environmental pollutants, risk involvement of HCV in causing MC.
factors for acquisition of HCV, and smoking habits were Cardiovascular manifestations of MC include ischemic
assessed to find their associations with HCV. A significant heart disease with coronary vasculitis, valvular heart disease
difference has been found only in the mean age of the two with mitral valvular damage, pericarditis, acute myocardial
groups, as patients with HCV and COPD have higher mean infarction, congestive heart failure, and dilative and hyper-
ages compared with those with HCV and no COPD (77), trophic cardiomyopathies (5). Pulmonary alterations that are
thereby suggesting an association between chronic HCV, mostly resulted from MC are mild in nature, while some-
with longer duration to COPD. In patients with HCV and times they lead to the severe outcomes such as diffuse al-
increased mean ages, there is a long period of immune ac- veolar damage and organizing pneumonia. In HCV patients,
tivation and inflammation due to chronic viral infection that various cytokines, inflammatory cells, and immune com-
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

aggravates the COPD. Because of long-term viral replica- plexes act as major contributors of heart diseases, however,
tion in the body, the expression of CD8+ T lymphocytes is in the serum of MC+HCV patients, there is a direct in-
upregulated. This upregulation activates a cascade of in- volvement of viral antigens (bind with immunoglobulins)
flammatory pathways resulting in the release of inflamma- and various immune complexes that ultimately deposit in
tory mediators, which causes the airway obstruction in lung interstitium (23).
COPD (60). In addition, in HCV patients, increased level of In patients with HCV and MC, various B cell subsets and
IL-8 causes the inflammatory response in the body as well as autoantibodies are observed, which cause lung fibrosis. CIC
suppresses the action of IFN-c (79). comprises IgG, IgM, IgA, C3, and C4 and were detected in
HCV patients with heart diseases in different proportions. For
example, patients with valvular disease present high levels of
Mixed Cryoglobulinemia
C3, however, the levels of IgM are higher in patients with
myocarditis (14). Cytokine zTNF4 (a member of the tumor
Cryoglobulinemia is a blood disorder, characterized by necrosis factor family) was found at higher levels in patients
the presence of special blood immunoglobulins called with MC-linked HCV. CXCL10-mRNA is overexpressed in
cryoglobulins. The MC is a systemic vasculitis with various hepatocytes in HCV patients and plays a role in the patho-
clinical symptoms (23). Cryoglobulins are common in the genesis of disease through recruitment of inflammatory cells
blood of HCV patients, as MC is found to be the most (T cells) to the site of inflammation. Presence of high levels of
common and severe EHM of HCV infection (70). The as- CXCL10 in serum results in histological severity and globular
sociation of MC with HCV has been defined on the basis of inflammation. In HCV+MC patients, certain genetic muta-
its higher prevalence and pathogenesis in HCV patients (93). tions also play their roles in overexpansion of the B cell
Cryoglobulins starts to precipitate or deposit in the vessels community. Among these mutations, there includes translo-
when blood is chilled (below 37C), thus restricting the blood cation of the bcl2 gene. This mutation results in the over-
flow in vessels resulting in vasculitis (24,94). Longer duration expression of antiapoptotic bcl2 gene, resulting in increased
of HCV infection and clonal B cell expansion in hepatocytes survival of B cell populations (76,94).
is among the many factors that cause MC (68,75). Along with
many other complications of MC, pulmonary hemorrhage
and cardiovascular disorders are found to be common in HCV Cardiopulmonary Diseases Related
patients (65). Among the patients of HCV, percentage of MC- to Liver Cirrhosis
affected patients is 19–50% (26), however, its prevalence HCV is one of the major causes of liver cirrhosis in the
varies with the geographical heterogeneity. In HCV patients world. Chronic liver inflammation is responsible for liver
the organs that are mostly affected by MC include the lungs, cirrhosis that leads to severe liver diseases such as liver
joints, nerves, and kidneys (70). cancer and liver failure. Cardiac and liver diseases are
In HCV patients, MC is the result of antigen-driven mutually interrelated (63). The major cardiac manifestations
lymphoproliferative process of B cells. HCV is known to that arise due to liver cirrhosis are cirrhotic cardiomyopathy
reside on the peripheral lymphocytes circulating in the body (CCM), pericardial effusion, systolic and diastolic dys-
where it replicates and increases its number. HCV has function, and noncirrhotic high-output heart failure that
ability to bind with the CD81 ligand present on the B- occurs due to hepatic arteriovenous shunts. The exact defi-
Lymphocyte through E2 protein. E2 protein present on viral nition of CCM was reported in 2005 by experts in annual
envelope mimics the human IgS (26), thus, it activates meeting of the World Gastroenterology Organization (57).
complement cascade, resulting in the formation of immune The main clinical features of CCM are cardiac dysfunction
complexes upon its binding to CD81 (33). Furthermore, this (systolic and diastolic), hyperdynamic circulation, and ab-
binding triggers expression of a special set of genes en- normal electrophysiological findings (73).
coding the variable region (VH, VK) of immunoglobulins In patients suffering from chronic liver diseases, cardiac
and of those involved in production of IgM-RF with cross- dysfunction is the main heart problem that depends on many
reactive WA-idiotype (69,94). In type 2 MC, gene expres- factors. These factors include defects in the cardiac b-
sion results in the production of polyclonal antibodies (IgG) adrenergic receptor system, plasma membrane fluidity,
followed by production of monoclonal antibodies (IgG, abnormalities in the membrane calcium channels, and
IgM, and IgA) after the emergence of a single-type domi- pathological effects of many humoral factors such as NO,
nant B cell. While in case of type 3 MC, only polyclonal cytokines, carbon monoxide, and endocannabinoids (56).
(IgG, IgM) antibodies are produced (26). In addition, the HCV-related CCM shows three major pathophysiologic
presence of cryoglobulins mainly complexed with HCV abnormalities, which include cardiac electrophysiological
6 ILYAS ET AL.

abnormalities, structural and functional ventricular abnor- ing myofibroblast cells start to deposit in the lung tissues,
malities, and abnormal ventricular response to stress (44). thereby causing the lung tissue fibrosis (85). Pulmonary
Other cardiac manifestations related to cirrhosis are en- sarcoidosis is another IFN-a-related complication resulted
docarditis, arrhythmias, and tachycardia (45). Arrhythmias from the stimulation of Th1 response and macrophages, along
are associated with hepatic pulmonary syndrome. In liver with the induction of autoantibodies due to IFN therapy (2).
cirrhosis, different markers such as natriuretic peptides, cy- The mechanism is known to cause pulmonary sarcoidosis,
tokeratin 18, TNF, and microRNAs are produced in high granuloma formation in lung tissues, and macrophage acti-
number. These biomarkers are used to access cardiac diseases vation due to dysregulated IFN-c.
due to chronic liver diseases (61). Despite many pulmonary To date, only few cases of IFN-related sarcoidosis have
diseases that are attributable to the lymphoproliferative nature been reported in patients of myelogenous leukemia, multiple
of HCV, certain pulmonary alterations can be the direct myeloma, and HCV (1,2,54). IFN treatment is also re-
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

outcome of advanced liver diseases such as liver cirrhosis in sponsible for causing pulmonary arterial hypertension
HCV patients. Among these outcomes, there include hepa- (PAH) in HCV patients (72). Cardiovascular complica-
topulmonary syndrome and portopulmonary hypertension tions due to PEG-IFN-a include congestive heart failure,
(29). myocardial infarction, life-threatening arrhythmias, ische-
Hepatopulmonary syndrome is characterized by altering mic heart disease, CAD, pericarditis, and DCM (38). These
gas function in the lungs and shunting of blood from right to cardiac complications occur only after prolong treatment of
left side because of the intrapulmonary vascular dilatations. IFN-a therapy. Anemia is a common outcome of Peg-IFN/
Any level of liver damage, including liver cirrhosis and RBV treatment in many HCV patients, however, some pa-
portal hypertension, contributes to disease. Among the HCV tients who have CAD cannot tolerate anemia (18). Besides
patients, it is more prevalent among those who are suffering this, DCM was completely resolved when Peg IFN-a and
from the advanced liver diseases (20,29). Until now, endo- RBV therapy was stopped (92).
thelial cell injury in the lungs has been said to be a main In the United States, the triple therapy, including the
reason for the intrapulmonary vascular dilatations. In HCV RBV, PEGylated IFN, and protease inhibitors (boceprevir
patients, endothelial cell injury occurs due to increased and telaprevir), has been adopted in 2011 (80). The direct
levels of bile acid and cytokines (TNF-a). Bile acids are acting antivirals (DAAs) reduced the duration of therapy up
produced due to liver cirrhosis, while cytokines mainly re- to 24–28 weeks in *50% of patients, while responsible for
sult from HCV-induced immune activation (29,30). TNF-a the improvement in sustained virological response (SVR)
is known to perform the proinflammatory function that rate in naive and experienced patients. These benefits could
further promotes the leukocyte–endothelium interaction or be achieved only when DAAs are used in combination with
sometimes activates the arachidonic acid pathway (70). The peg-IFN/RBV (91), but the treatment is not effective in case
second outcome of advanced liver diseases is porto- of patients suffering with cirrhosis. In last 3 years, DAAs
pulmonary hypertension. It is characterized by the altered revolutionized the treatment of HCV by using multiple
pressure in the pulmonary arteries and veins. About 5% of classes of DAAs in IFN-free regimens (46).
the patients suffered from cirrhosis also develop porto- Until now a large number of antivirals have been ap-
pulmonary hypertension (29). proved by the European Medicine Agency (EMA) and the
Food and Drug Administration (FDA) starting with boce-
previr and telaprevir in 2011 for the treatment of HCV ge-
HCV Antiviral Therapy and Cardiopulmonary
notype 1, followed by the approval of four new protease/
Complications
polymerase inhibitors: sofosbuvir, simeprevir, daclatasvir,
Before 2011, the approved therapies for treating the chronic and ledipasvir, and a NS3/4A protease inhibitor asunaprevir
HCV infection include IFN, PEGylated IFN-a alone, or in in 2014 (74). A new combination of ombitasvir/paritaprevir
combination with ribavirin (RBV) (42). IFN and RBV com- was also approved as a treatment option for HCV in the year
bination is in use for treating the HCV infection, however, 2015. Recently in 2016, another combination of grazoprevir/
PEGylated IFN a-2b is approved for the treatment of chronic elbasvir has been recommended by EMA. These antivirals
HCV as a monotherapy. Antiviral-related lung pathologies are are associated with higher SVR rate (90–100%) as it has
mostly rare, but are potentially fatal with an incidence of 0.4% been proven from various pivotal studies (53). Use of
to <1% (13). The spectrum of these pathologies includes di- multiple classes of the DAAs has been proven to be effec-
verse outcomes such as interstitial pneumonitis, bronchiolitis tive in cirrhosis patients because short and tolerated regi-
obliterans organizing pneumonia (BOOP) (secondary orga- mens increased the SVR rate up to 95% (22). This treatment
nizing pneumonia), pulmonary sarcoidosis, pleural effusion, has limited EHMs, but mostly it affects the kidneys.
and exacerbation of asthma (13). Interstitial pneumonia (IP) Although the efficacy of DAAs therapy is higher, it has
and sarcoidosis are among the well-documented complica- some severe side effects such as lymphopenia, hemolytic
tions of the IFN therapy of HCV patients (3). The incidence anemia, and decomposition of cirrhosis (80). These side
rate of IP is estimated to be 0.01–0.3% (37). effects also favor the lung infections by bacterial pathogens
The exact mechanism for the toxicity of lungs by IFN-a is such as mycobacterium abscessus (80). Pneumonia is an-
not well understood, however, IFN-a causes the inhibition other outcome of cirrhotic HCV patients who were given
of suppressor T cells and enhances the expression of cyto- DAA therapy. DAAs are responsible for the cardiovascular
toxic T cells (55). In addition, it increases the release of manifestation in those HCV patients suffering with DM
fibrinogenic cytokines, thus causing the lung injury. In the (48). The most common side effects of the DAAs include
presence of higher level of type 2 cytokines (IL-13 and IL- fatigue, headache, nausea, and insomnia. Percentage of
4), the extracellular matrix proteins produced by activat- HCV patients suffering with anemia is lower when treated
HCV-ASSOCIATED EXTRAHEPATIC MANIFESTATIONS 7

with simeprevir/sofosbuvir compared with the RBV, how- 8. Brunetti G, Delmastro M, Nava S, et al. Detection of HCV-
ever, two serious complications, including renal failure and RNA in bronchoalveolar lavage from a woman with pul-
aspiration pneumonia, were considered for treatment-related monary fibrosis. Respir Med 2003;97:736–738.
patients’ death. Besides, this simeprevir and sofosbuvir were 9. Butt AA, Xiaoqiang W, Budoff M, et al. Hepatitis C virus
also associated with pulmonary manifestations, including mild infection and the risk of coronary disease. Clin Infect Dis
diffuse emphysema, PAH, and interstitial pneumonitis (31,81). 2009;49:225–232.
Among various DAAs, ombitasvir/paritaprevir/ritonavir and 10. Cacoub P, Gragnani L, Comarmond C, et al. Extrahepatic
dasabuvir were found to have serious pulmonary and cardiac manifestations of chronic hepatitis C virus infection. Dig
complications, including acute respiratory failure, sinus Liver Dis 2014;46:S165–S173.
tachycardia, ventricular extrasystoles, and acute transient 11. Charron D. HLA class II disease associations: molecular
basis. J Autoimmun 1992;5:45–53.
stroke (21).
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

12. Chen Y-J, Lin M-S, Hsu K-Y, et al. Prevalence of


Conclusion asymptomatic peripheral arterial disease and related risk
factors in younger and elderly patients in Taiwan. An-
Although HCV causes the liver cirrhosis and hepatocel- giology 2014;65:396–401.
lular carcinoma, chronic HCV infection is involved in many 13. Chopra A, Marak C, Alappan N, et al. Organizing pneu-
EHMs that affect every organ specially heart and lungs via a monia associated with pegylated interferon a and ribavirin
variety of mechanisms. The crucial cardiopulmonary dis- therapy. Case Rep Pulmonol 2015;2015:794592.
eases related to HCV include myocarditis, cardiomyopa- 14. Cristea A, Rus H, Niculescu F, et al. Characterization of
thies, CVDs, strokes, COPD, IPF, asthma, and interstitial lung circulating immune complexes in heart disease. Immunol
diseases. Mostly cardiopulmonary manifestations of HCV are Lett 1986;13:45–49.
immune-mediated and some occurred due to chronic inflam- 15. Domont F, and Cacoub P. Chronic hepatitis C virus in-
mation. Cryoglobulinemia, liver cirrhosis, and HCV antiviral fection, a new cardiovascular risk factor? Liver Int
therapy are the most significant source of HCV-related lung 2016;36:621–627.
and heart diseases in those persons suffering with advanced 16. Duncan DJ, Hopkins PM, and Harrison SM. Negative
inotropic effects of tumour necrosis factor-alpha and
liver diseases. The classic anti-HCV therapy (IFN-RBV) is also
interleukin-1beta are ameliorated by alfentanil in rat ven-
linked with many adverse effects, however, with the discovery
tricular myocytes. Br J Pharmacol 2007;150:720–726.
of new directly acting anti-HCV compounds the IFN-RBV- 17. Egan JJ, Woodcock AA, and Stewart JP. Viruses and idio-
mediated side effects will be decreased with the more efficient pathic pulmonary fibrosis. Eur Respir J 1997;10:1433–1437.
viral control. 18. El-Zayadi A-R. Hepatitis C comorbidities affecting the
course and response to therapy. World J Gastroenterol
Acknowledgment 2009;15:4993–4999.
IQ received funding from KACST large grant #162-34. 19. Erturk A, Tokgonul AN, Capan N, et al. Pulmonary alter-
ations in patients with chronic HCV infection. Dig Liver
Author Disclosure Statement Dis 2006;38:673–676.
20. Fallon MB, Krowka MJ, Brown RS, et al. Impact of he-
No competing financial interests exist. patopulmonary syndrome on quality of life and survival in
liver transplant candidates. Gastroenterology 2008;135:
References
1168–1175.
1. Abdi EA, Nguyen GK, Ludwig RN, et al. Pulmonary sar- 21. Feld JJ, Kowdley KV, Coakley E, et al. Treatment of HCV
coidosis following interferon therapy for advanced renal with ABT-450/r-ombitasvir and dasabuvir with ribavirin. N
cell carcinoma. Cancer 1987;59:896–900. Engl J Med 2014;370:1594–1603.
2. Akay BN, Ekmekci P, Sanli H, et al. Cutaneous, pulmonary 22. Feld JJ, Moreno C, Trinh R, et al. Sustained virologic re-
and hepatic sarcoidosis associated with autoimmune compli- sponse of 100% in HCV genotype 1b patients with cirrhosis
cations during interferon-alpha treatment for hepatitis C virus receiving ombitasvir/paritaprevir/r and dasabuvir for 12
infection. J Eur Acad Dermatol Venereol 2006;20:442–445. weeks. J Hepatol 2016;64:301–307.
3. Aliannejad R, and Ghanei M. Hepatitis C and pulmonary 23. Ferri C, La Civita L, Fazzi P, et al. Interstitial lung fibrosis
fibrosis: hepatitis C and pulmonary fibrosis. Hepat Mon and rheumatic disorders in patients with hepatitis C virus
2011;11:71–73. infection. Br J Rheumatol 1997;36:360–365.
4. Al-Moamary MS, Gorka T, Al-Traif IH, et al. Pulmonary 24. Franciscus A. An overview of extrahepatic manifestations
changes in liver transplant candidates with hepatitis C cir- of hepatitis C. HCSP Fact Sheet 2015;7:1–6.
rhosis. Saudi Med J 2001;22:1069–1072. 25. Fülöp T, Larbi A, and Pawelec G. Human T cell aging and
5. Antonelli A, Ferri C, Ferrari SM, et al. Endocrine mani- the impact of persistent viral infections. Front Immunol
festations of hepatitis C virus infection. Nat Clin Pract 2013;4:1–9.
Endocrinol Metab 2009;5:26–34. 26. Galossi A, Guarisco R, Bellis L, et al. Extrahepatic mani-
6. Boddi M, Abbate R, Chellini B, et al. Hepatitis C virus festations of chronic HCV infection. J Gastrointestin Liver
RNA localization in human carotid plaques. J Clin Virol Dis 2007;16:65–73.
2010;47:72–75. 27. Gilani SR, Vuga LJ, Lindell KO, et al. CD28 down-
7. Bowen DG, and Walker CM. Adaptive immune responses regulation on circulating CD4 T-cells is associated with
in acute and chronic hepatitis C virus infection. Nature poor prognoses of patients with idiopathic pulmonary fi-
2005;436:946–952. brosis. PLoS One 2010;5:e8959.
8 ILYAS ET AL.

28. Gill K, Ghazinian H, Manch R, et al. Hepatitis C virus as a 47. Lukacs NW, Hogaboam C, Chensue SW, et al. Type 1/type
systemic disease: reaching beyond the liver. Hepatol Int 2 cytokine paradigm and the progression of pulmonary fi-
2016;10:415–423. brosis. Chest 2001;120:5–8.
29. Goldberg DS, and Fallon MB. The art and science of di- 48. Maan R, and van der Meer AJ. Recent advances in man-
agnosing and treating lung and heart disease secondary to aging chronic HCV infection: focus on therapy in patients
liver disease. Clin Gastroenterol Hepatol 2015;13:2118– with severe liver disease. F1000Res 2016;5:pii: F1000.
2127. DOI: 10.12688/f1000research.7399.1.
30. Gomez FP, Martinez-Palli G, Barbera JA, et al. Gas ex- 49. Matsumori A. Hepatitis C virus and cardiomyopathy. Herz
change mechanism of orthodeoxia in hepatopulmonary 2000;25:249–254.
syndrome. Hepatology 2004;40:660–666. 50. Matsumori A. Hepatitis C virus infection and cardiomy-
31. Helmers RA, Byrne TJ, Wesselius LJ, et al. Serious pul- opathies. Circ Res 2005;96:144–147.
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

monary toxicity secondary to novel hepatitis C antiviral 51. Matsumori A. Global alert and response network for hep-
therapy in a liver transplant recipient. Mayo Clin Proc atitis C virus-derived heart diseases: a call to action. CVD
2015;90:1294–1297. Prev Control 2009;4:109–118.
32. Hsu Y-H, Muo C-H, Liu C-Y, et al. Hepatitis C virus in- 52. Matsumori A, Shimada T, Chapman NM, et al. Myocarditis
fection increases the risk of developing peripheral arterial and heart failure associated with hepatitis C virus infection.
disease: a 9-year population-based cohort study. J Hepatol J Card Fail 2006;12:293–298.
2015;62:519–525. 53. McCaughan GW. New therapies against HCV: expected
33. Hu SL, and Jaber BL. Ribavirin monotherapy for hepatitis risks and challenges associated with their use in the liver
C virus-associated membranous nephropathy. Clin Nephrol transplant setting. J Hepatol 2012;57:1361–1367.
2005;63:41–45. 54. Michavila IA, Sanchez MM, Ronchel JP, et al. Sarcoidosis
34. Idilman R, Cetinkaya H, Savas I, et al. Bronchoalveolar following combined ribavirin and interferon therapy: a case
lavage fluid analysis in individuals with chronic hepatitis C. report and review of the literature. Arch Bronconeumol
J Med Virol 2002;66:34–39. 2004;40:45–49.
35. Ishizaka N, Ishizaka Y, Takahashi E, et al. Association 55. Midturi J, Hoffman MS, Hurley D, et al. Spectrum of
between hepatitis C virus seropositivity, carotid-artery pulmonary toxicity associated with the use of interferon
plaque, and intima-media thickening. Lancet 2002;359: therapy for hepatitis C: case report and review of the lit-
133–135. erature. Clin Infect Dis 2004;39:1724–1729.
36. Ishizaka Y, Ishizaka N, Takahashi E, et al. Association 56. Moller S, and Bernardi M. Interactions of the heart and the
between hepatitis C virus core protein and carotid athero- liver. Eur Heart J 2013;34:2804–2811.
sclerosis. Circ J 2003;67:26–30. 57. Møller S, and Henriksen JH. Cardiovascular complications
37. Ji FP, Li ZX, Deng H, et al. Diagnosis and management of of cirrhosis. Gut 2008;57:268–278.
interstitial pneumonitis associated with interferon therapy 58. Moore BB, Fry C, Zhou Y, et al. Inflammatory leukocyte
for chronic hepatitis C. World J Gastroenterol 2010;16: phenotypes correlate with disease progression in idiopathic
4394–4399. pulmonary fibrosis. Front Med 2014;1:pii: 00056. DOI:
38. Karbasi-Afshar R. Treatment of hepatitis C virus infection 10.3389/fmed.2014.
and associated vascular complications: a literature review. 59. Moore BB, and Moore TA. Viruses in idiopathic pulmo-
Iran J Med Sci 2014;39:238–246. nary fibrosis. Etiology and exacerbation. Ann Am Thorac
39. Katsi V, Felekos I, Skevofilax S, et al. Cardiovascular Soc 2015;12 Suppl 2:186–192.
disease and hepatitis C virus infection: an irrelevant state- 60. Moorman JP, Fitzgerald SM, Prayther DC, et al. Induction
ment or a hot relationship? Cardiol Rev 2015;23:11–17. of p38- and gC1qR-dependent IL-8 expression in pulmo-
40. Kawai C. From myocarditis to cardiomyopathy: mecha- nary fibroblasts by soluble hepatitis C core protein. Respir
nisms of inflammation and cell deathlearning from the past Res 2005;6:105.
for the future. Circulation 1999;99:1091–1100. 61. Mozos I. Arrhythmia risk in liver cirrhosis. World J He-
41. Kawai C, and Matsumori A. Dilated cardiomyopathy up- patol 2015;7:662–672.
date: infectious-immune theory revisited. Heart Fail Rev 62. Naruse T, and Inoko H. HLA and hepatitis C virus positive
2013;18:703–714. cardiomyopathy. Jpn J Clin Med 2000;58:212–217.
42. Kumar KS, Russo MW, Borczuk AC, et al. Significant 63. Naschitz JE, Slobodin G, Lewis RJ, et al. Heart diseases
pulmonary toxicity associated with interferon and ribavirin affecting the liver and liver diseases affecting the heart. Am
therapy for hepatitis C. Am J Gastroenterol 2002;97:2432– Heart J 2000;140:111–120.
2440. 64. Rabea AEM, Zidan M, Daabis R, et al. Prevalence of
43. Leandro G, Mangia A, Hui J, et al. Relationship between chronic hepatitis C virus (HCV) infection in patients with
steatosis, inflammation, and fibrosis in chronic hepatitis C: idiopathic pulmonary fibrosis. Egypt J Chest Dis Tuberc
a meta-analysis of individual patient data. Gastroenterology 2015;64:907–913.
2006;130:1636–1642. 65. Ragab G, and Hussein MA. Vasculitic syndromes in hep-
44. Lee RF, Glenn TK, and Lee SS. Cardiac dysfunction in atitis C virus: a review. J Adv Res 2017;8:99–111.
cirrhosis. Best Pract Res Clin Gastroenterol 2007;21:125– 66. Richardson P, McKenna W, Bristow M, et al. Report of the
140. 1995 World Health organization/International Society and
45. Liu H, and Lee S. Cardiopulmonary dysfunction in cir- Federation of cardiology Task Force on the definition and
rhosis. J Gastroenterol Hepatol 1999;14:600–608. classification of cardiomyopathies. Circulation 1996;93:
46. Lok AS, Gardiner DF, Lawitz E, et al. Preliminary study of 841–842.
two antiviral agents for hepatitis C genotype 1. N Engl J 67. Ryu JH, Daniels CE, Hartman TE, et al. Diagnosis of in-
Med 2012;366:216–224. terstitial lung diseases. Mayo Clinic Proc 2007;82:976–986.
HCV-ASSOCIATED EXTRAHEPATIC MANIFESTATIONS 9

68. Saadoun D, Asselah T, Resche-Rigon M, et al. Cryoglo- 82. Ueda T, Ohta K, Suzuki N, et al. Idiopathic pulmonary
bulinemia is associated with steatosis and fibrosis in fibrosis and high prevalence of serum antibodies to hepa-
chronic hepatitis C. Hepatology 2006;43:1337–1345. titis C virus. Am Rev Respir Dis 1992;146:266–268.
69. Saadoun D, Bieche I, Maisonobe T, et al. Involvement of 83. Weidensaul D, Imam T, Holyst MM, et al. Polymyositis,
chemokines and type 1 cytokines in the pathogenesis of pulmonary fibrosis, and hepatitis C. Arthritis Rheum 1995;
hepatitis C virus–associated mixed cryoglobulinemia vas- 38:437–439.
culitis neuropathy. Arthritis Rheum 2005;52:2917–2925. 84. Wier WG, Egan TM, Lopez-Lopez JR, et al. Local control of
70. Saleh AM, Elalfy H, Arafa MM, et al. Association between excitation-contraction coupling in rat heart cells. J Physiol
HCV induced mixed cryoglobulinemia and pulmonary af- 1994;474:463–471.
fection: the role of TNF-alpha in the pathogenesis of pul- 85. Wynn TA. Integrating mechanisms of pulmonary fibrosis.
monary changes. Egypt J Chest Dis Tuberc 2014;63:259–265. J Exp Med 2011;208:1339–1350.
Downloaded by Gothenburg University Library from online.liebertpub.com at 09/28/17. For personal use only.

71. Sanchez MJ, and Bergasa NV. Hepatitis C associated car- 86. Yang IV, Luna LG, Cotter J, et al. The peripheral blood
diomyopathy: potential pathogenic mechanisms and clini- transcriptome identifies the presence and extent of disease
cal implications. Med Sci Monit 2008;14:55–63. in idiopathic pulmonary fibrosis. PLoS One 2012;7:
72. Savale L, Sattler C, Gunther S, et al. Pulmonary arterial e37708.
hypertension in patients treated with interferon. Eur Respir 87. Yokoyama T, Vaca L, Rossen RD, et al. Cellular basis for
J 2014;44:1627–1634. the negative inotropic effects of tumor necrosis factor-alpha
73. Sawant P, Vashishtha C, and Nasa M. Management of in the adult mammalian heart. J Clin Invest 1993;92:2303–
cardiopulmonary complications of cirrhosis. Int J Hepatol 2312.
2011;2011:280569. 88. Yoshinouchi T, Naniwa T, Shimizu S, et al. Expression of
74. Scavone C, Sportiello L, Rafaniello C, et al. New era in chemokine receptors CXCR3 and CCR4 in lymphocytes of
treatment options of chronic hepatitis C: focus on safety of idiopathic nonspecific interstitial pneumonia. Respir Med
new direct-acting antivirals (DAAs). Expert Opin Drug Saf 2007;101:1258–1264.
2016;15:85–100. 89. Younossi Z, Stepanova M, Nader F, et al. Associations of
75. Sene D, Ghillani-Dalbin P, Thibault V, et al. Longterm chronic hepatitis C with metabolic and cardiac outcomes.
course of mixed cryoglobulinemia in patients infected with Aliment Pharmacol Ther 2013;37:647–652.
hepatitis C virus. J Rheumatol 2004;31:2199–2206. 90. Zampino R, Marrone A, Restivo L, et al. Chronic HCV in-
76. Sene D, Limal N, and Cacoub P. Hepatitis C virus- fection and inflammation: clinical impact on hepatic and extra-
associated extrahepatic manifestations: a review. Metab hepatic manifestations. World J Hepatol 2013;5:528–540.
Brain Dis 2004;19:357–381. 91. Zeuzem S, Andreone P, Pol S, et al. Telaprevir for retreat-
77. Serpil E, Ahmet O, and Ayten K. Hepatitis C virus infec- ment of HCV infection. N Engl J Med 2011;364:2417–2428.
tion and chronic obstructive pulmonary disease. Hepat Mon 92. Zhao W, Ji F, Yu S, et al. Dilated cardiomyopathy and
2009;2009:39–44. hypothyroidism associated with pegylated interferon and ri-
78. Shichi D, Kikkawa EF, Ota M, et al. The haplotype block, bavirin treatment for chronic hepatitis C: case report and
NFKBIL1-ATP6V1G2-BAT1-MICB-MICA, within the literature review. Braz J Infect Dis 2014;18:110–113.
class III-class I boundary region of the human major his- 93. Zignego AL, and Craxi A. Extrahepatic manifestations of
tocompatibility complex may control susceptibility to hepatitis C virus infection. Clin Liver Dis 2008;12:611–636.
hepatitis C virus-associated dilated cardiomyopathy. Tissue 94. Zignego AL, Giannini C, and Gragnani L. HCV and lym-
Antigens 2005;66:200–208. phoproliferation. Clin Dev Immunol 2012;2012:980942.
79. Silva DR, Stifft J, Cheinquer H, et al. Prevalence of hep-
atitis C virus infection in patients with COPD. Epidemiol
Infect 2010;138:167–173. Address correspondence to:
80. Soza A, Labbe P, Arrese M, et al. Mycobacterium ab- Dr. Ishtiaq Qadri
scessus pulmonary infection during hepatitis C treatment Department of Biological Sciences
with telaprevir, peginterferon and ribavirin. Ann Hepatol King Abdul Aziz University
2015;14:132–136. Jeddah 110809
81. Tamaki K, and Okubo A. Simeprevir with peginterferon Kingdom of Saudi Arabia
and ribavirin induced interstitial pneumonitis: first case
report. World J Gastroenterol 2015;21:1009–1013. E-mail: ishtiaq80262@yahoo.com

Das könnte Ihnen auch gefallen