Epidemiological studies have demonstrated an increased risk of developing B-cell lymphomas in patients with chronic hepatitis C virus (HCV) infection. However, the strength of the association shows great geographic discrepancies, with higher relative risk in countries with high HCV prevalence. It remains unclear whether additional environmental and genetic factors are involved or if the international variability is simply a consequence of the variable infection prevalence. Therefore, a causal relationship remains controversial. Other confounding factors may affect the comparability of the different studies, including the method of HCV assessment, the selection of normal controls, the lymphoma classification used, and the year of publication. The most convincing proof is the observation, mainly limited to some indolent subtypes, of B-cell lymphoma regressions after HCV eradication with IFN and ribavirin. However, the molecular mechanisms of the HCV-induced lymphomagenesis are mainly hypothetical. According to the model considered to be most plausible, lymphoma growth is a consequence of the continuous antigenic stimulation of the B-cell immunologic response induced by the chronic viral infection. This review summarizes the current epidemiological and biological evidence of a role of HCV in lymphomagenesis, describing the putative mechanisms for a causative relationship. The clinical characteristics and management difficulties of the HCV-associated lymphomas are also discussed. HCV treatment with IFN cannot be given safely in concomitance with cytotoxic lymphoma treatment because of hematological and liver toxicity. However, novel and better tolerated antiviral regimens are under development and will hopefully make the treatment of both lymphoma and hepatitis easier in the future.

Learning Objectives
  • To understand the current lines of evidence linking HCV infection and lymphomagenesis

  • To review the specific problems of the clinical management of patients with chronic HCV infection

The hepatitis C virus (HCV) is a major cause of acute hepatitis and chronic liver disease. Worldwide, ∼150 million people are estimated to be chronically infected and up to 500 000 people die each year from hepatitis C-related liver diseases.1,2  There are important geographical variations in the extent of HCV chronic infection, with the highest prevalence (>10%) in Egypt, Central Africa, Mongolia, and Bolivia.3  It has been estimated that ∼3-4 million people in the United States4  and 15 million people in Europe are chronically infected2 ; Asia has the largest population of infected persons, with the highest number in China (estimated at 13 million) and India (estimated at 9.5 million).5 

HCV is not only hepatotropic, but also lymphotropic, because it equally infects both hepatocytes and lymphocytes. A large body of epidemiological, clinical, and biological data have suggested an association between HCV infection and the pathogenesis of at least a portion of B-cell non-Hodgkin lymphomas (NHLs). However, a causative relationship has not been fully confirmed and the etiopathogenetic mechanisms remain largely speculative.

The initial finding that generated a large number of epidemiological studies on the association between HCV and lymphoproliferative disorders was the extremely high prevalence of HCV infection in patients with type II mixed cryoglobulinemia (MC).6  Type II MC is a disorder characterized by circulating cryoglobulins consisting of complexes of polyclonal IgG and monoclonal IgM rheumatoid factors that become insoluble at reduced temperature. The main clinical features are palpable purpura, arthralgia, weakness, organ involvement (liver, kidney), peripheral neuropathy, and vasculitis. The histological lesions are secondary to vasculitis, which is caused by immune complexes deposited in small vessels. The antigenic component of these immune complexes has been shown to be highly enriched in HCV-RNA.6  Nearly all MC patients are HCV infected, even in the absence of chronic liver disease. Conversely, the MC prevalence in patients with HCV infection shows a great geographic heterogeneity. Low levels of circulating cryoglobulins are frequently detected in HCV-positive individuals, but only a minority has an overt cryoglobulin vasculitis.7,8  The biological basis of the relation between MC and HCV is not entirely elucidated. Analysis of the immunoglobulin heavy- and light-chain genes highlights the predisposition of HCV to select a restricted B-cell repertoire in response to chronic antigenic stimulation, favoring the preferential use of the IGHV1-69, IGKV3-A27, and IGKV3-20.9,10  Monoclonal B lymphocytes have indeed been detected in the liver, bone marrow, and peripheral blood of patients with MC8  and, in a multicenter Italian study, the risk of developing a B-cell lymphoma in HCV-infected patients with symptomatic cryoglobulinemia was 35 times higher than in the general population.11 

Numerous studies have found a high prevalence of HCV seropositivity in patients with B-cell lymphoproliferative disorders, particularly B-cell NHL, including cases in which MC was not present. The evidence for an association with T-cell lymphoma, Hodgkin lymphoma, and plasma cell disorders is less convincing.12-15  The association with B-cell NHL is particularly evident in countries with a high prevalence of HCV infection. There are important geographic variations of the prevalence of HCV-associated lymphomas, ranging from 0% to 50%. This variability may simply reflect the HCV epidemiology, but other confounding factors may affect the comparability of the different studies, including the timing and rate of HCV assessment in lymphoma patients, the method of HCV assessment, the selection of normal controls, the lymphoma classification used, and the year of publication.12-14  Two systematic reviews of >60 studies indicate that, overall, 13%–18% of B-cell lymphomas are associated with HCV infection (Table 1), but there is an important heterogeneity among studies and in study design.14,16 

Table 1.

Systematic reviews14,16  and meta-analyses12-14,16,17  addressing the risk of HCV-associated lymphomas

Systematic reviews14,16 and meta-analyses12-14,16,17 addressing the risk of HCV-associated lymphomas
Systematic reviews14,16 and meta-analyses12-14,16,17 addressing the risk of HCV-associated lymphomas

CI indicates confidence interval; and N/A, not applicable.

Table 1 also summarizes the findings of meta-analyses, which take into account the case-control studies and cohort studies published up to 2008.12-14,16,17  Globally, the relative risk of being infected is approximately 2-4 times higher among B-cell lymphoma patients than in the general population. However, because the virus is sporadic in some areas and endemic in others, a clear correlation between HCV and lymphoma has emerged mainly in populations with a high prevalence of the virus. Lower or no evident correlation has been demonstrated in low prevalence countries such as Scandinavia, the United Kingdom, and or Canada. In the United States, 2 large studies involving the National Cancer Institute–Surveillance, Epidemiology, and End Results (NCI-SEER) registry18  and the US Veterans Affairs health system19  have documented a modest, but significant, increase of the risk of B-cell lymphoma in patients with chronic HCV infection. Similarly, the large European multicenter case-control study EPILYMPH reported an estimated 2-fold lymphoma risk.20  Nevertheless, it should be noted that some studies have reported a low prevalence of lymphoma in infected people living in areas with a high prevalence of HCV (Thailand, Macedonia) and others have reported a relatively high prevalence of HCV-associated lymphomas in countries with a relatively low prevalence of the virus (Japan), indicating that additional genetic or environmental factors may be needed for the development of an overt lymphoma. In most meta-analyses, a high geographic heterogeneity is evident; nevertheless, in the countries where HCV prevalence is high, up to 10% of NHL may be attributable to the infection. Compared with the strong correlation between HCV and hepatocarcinoma, there is only a moderate risk for lymphoma development in HCV-positive patients and the presence of a causal link between HCV and lymphomagenesis remains controversial.21  However, interestingly, elimination of the virus may reduce the incidence of NHL in patients with chronic HCV infection.22 

HCV is currently classified into at least 6 major genotypes that are variably distributed in different countries (Table 2). The influence of viral genotypes on the pathogenesis and course of hepatitis is controversial, but genotype determination is important for the prediction of response to antiviral treatment. No clear association has emerged in the published literature between genotypes and the risk of lymphoma, and the genotype distribution in HCV-associated lymphoma apparently mirrors that of the countries where the epidemiologic studies have been conducted.15 

Table 2.

Principal characteristics of HCV genotypes*

Principal characteristics of HCV genotypes*
Principal characteristics of HCV genotypes*
*

HCV Genotype 7, extremely rare, is a provisional entity discovered in patients who were presumably infected in Central Africa.

†HCV is a bloodborne virus and infection is commonly due to unsafe injection practices: inadequate sterilization of medical equipment and blood transfusion. Transfusion of unscreened blood and blood products was the main route before the 1990s; today, in Western countries, HCV is rarely transmitted by transfusion due to thorough screening of the blood and effective viral inactivation procedures.

RBV indicates ribavirin; HCC, hepatocellular carcinoma; and IVDU, intravenous drug use.

It is still debated whether some specific lymphoma subtypes are more closely associated with HCV. The B-cell NHL subtypes most frequently described as being associated with HCV are marginal zone lymphomas (MZLs), in particular splenic MZLs (SMZLs), extranodal (mainly nongastric) MZL of mucosa-associated lymphoid tissue (MALT), lymphoplasmacytic lymphoma (LPL), and diffuse large B-cell lymphoma (DLBCL).10,15,23  In a meta-analysis of 15 case-control studies and 3 prospective cohorts published between 1997 and 2005, no clear difference emerged to suggest that NHL develops more frequently in specific histological subtypes.13  However, the EPILYMPH study identified DLBCL, MZL, and LPL as being the most frequently associated with HCV infection.20  Subsequently, the very large International Lymphoma Epidemiology Consortium (InterLymph) case-control study provided a subtype-specific analysis confirming that HCV infection was increasingly associated with DLBCL [odds ratio (OR) = 2.24], MZL (OR = 2.47), and LPL (OR = 2.57).17  DLBCL is also the most common subtype among HCV-negative NHL, whereas MZL and LPL are relatively uncommon. Table 3 reports the lymphoma subtype distribution of 38 HCV-associated lymphoma treated at the Oncology Institute of Southern Switzerland between 2000 and 2013, which shows a more frequent involvement of extranodal marginal zone and DLBCL. This seems mostly in keeping with the aforementioned reports; it should be noted, however, that HCV antibodies were investigated in less than half of the patients seen in the same period at our institution.

Table 3.

Frequency of anti-HCV antibodies detection in different histological subtypes in a cohort of 384 B-cell lymphoma patients with HCV serology tested at diagnosis and treated at the Oncology Institute of Southern Switzerland between 2000 and 2013

Frequency of anti-HCV antibodies detection in different histological subtypes in a cohort of 384 B-cell lymphoma patients with HCV serology tested at diagnosis and treated at the Oncology Institute of Southern Switzerland between 2000 and 2013
Frequency of anti-HCV antibodies detection in different histological subtypes in a cohort of 384 B-cell lymphoma patients with HCV serology tested at diagnosis and treated at the Oncology Institute of Southern Switzerland between 2000 and 2013

CI indicates confidence interval.

*One-sided 97.5% CI.

†This estimate of HCV prevalence among B-cell lymphoma may be biased. Indeed, ∼550 additional lymphoma patients seen in the same period were not tested for HCV at diagnosis. Most likely, patients with an expected increased risk for the infection (eg, those from endemic countries, those with a history of blood transfusions or of intravenous use of drugs, and those with elevate transaminases or a history of liver disease) were tested more frequently than the others, leading to a potential overestimation of the HCV prevalence.

In general, the broad clinicopathological heterogeneity of NHLs is maintained in the subgroup of HCV-associated lymphoma. However, at least in some studies, in addition to the predominance of diffuse large cell and marginal-zone histology, HCV-associated lymphomas appear to carry distinctive clinicopathological features that may partly depend on the presence of HCV infection. At onset, they often present splenic localization or extranodal involvement at target organs of HCV infection such as the liver and the salivary glands.24  Compared with HCV-negative patients, they also show a more frequent presence of increased transaminase levels, monoclonal gammopathies, autoimmune phenomena, rheumatoid factor, and asymptomatic cryoglobulinemia.15 

DLBCLs are the most common aggressive lymphomas arising in HCV-infected patients. A few studies25-28  have specifically described the main clinicopathological characteristics of HCV-associated DLBCL (Table 4) that very frequently present at an advanced stage mainly due to extranodal localizations, elevated lactate dehydrogenase (which may, at least in part, result from the concomitant hepatitis), and unfavorable International Prognostic Index score (Table 4). Whether these patients have a significantly worse outcome than HCV-negative lymphoma patients remains unclear. In a large survey of patients included in the Groupe d'Etude des Lymphomes de l'Adulte (GELA) trials, HCV-positive DLBCL patients had more frequent spleen involvement and were more frequently transformed from low-grade lymphoma than HCV-negative DLBCL patients, but histological transformation was not associated with a worse outcome.25  The Fondazione Italiana Linfomi (FIL, Italian Lymphoma Foundation) recently developed a specific prognostic score for this subset of patients based on a multicenter retrospective series of 535 patients with HCV-associated DLBCL treated with anthracycline-based regimens (with rituximab in 255 cases). Severe hepatotoxicity was observed in 14% of patients and it was not increased in patients receiving rituximab. The 3-year overall survival and progression-free survival rates were 71% and 55%, respectively. At multivariate analysis, Eastern Cooperative Oncology Group performance status of 2 or over, serum albumin below 3.5 g/dL, and HCV-RNA viral load over 1000 KIU/mL displayed an adverse prognostic significance. The combination of these 3 factors in a new “HCV-Prognostic Score” discriminated 3 risk groups with significantly different outcomes (low = 0; intermediate = 1; high-risk = ≥2 factors). The estimated overall survival at 3 years was 92% for the low-risk group, 77% for the intermediate-risk group, and 39% for the high-risk group. The 3-year progression-free survival rate was 81%, 61%, and 19% in the low-, intermediate- and high-risk groups, respectively. This score retained prognostic value in the subgroups of patients treated with and without rituximab and performed better than the International Prognostic Index.28 

Table 4.

Comparison of clinical features at presentation between different studies of DLBCL associated with HCV25-28 

Comparison of clinical features at presentation between different studies of DLBCL associated with HCV25-28
Comparison of clinical features at presentation between different studies of DLBCL associated with HCV25-28

NR indicates not reported; OS, overall survival; RFS, relapse-free survival; EFS, event-free survival; and PFS, progression-free survival.

*The working formulation was used to define histology by Tomita et al,27  reporting 20 large cell lymphomas and other aggressive histologies; this study included also 4 cases of T-cell origin.

Among indolent lymphomas, 2 rare, peculiar clinical presentations were recently described in HCV-infected patients: primary SMZL with MC type II and subcutaneous “lipoma-like” extranodal marginal zone B-cell lymphoma of MALT type. In the former, ∼70% of patients are women and most have symptomatic cryoglobulinemia (vasculitis, arthralgia, peripheral neuropathy) and circulating villous lymphocytes. HCV eradication with IFN and ribavirin has been reported to induce a complete lymphoma remission in most cases and should be the first choice of treatment.29  In subcutaneous “lipoma-like” extranodal marginal zone B-cell lymphoma of MALT type, the large majority of cases were described in elderly women. The disease is characteristically confined to the subcutaneous tissue; the skin and the cutaneous adnexa are not involved. Typical lesions are single or multiple soft and mobile subcutaneous nodules underlying a normal-appearing skin. The clinical course is usually indolent. Analogous to what has been described in splenic lymphomas, objective lymphoma regressions have been reported after HCV eradication.30 

HCV is an enveloped, positive-stranded RNA virus belonging to the Flaviviridae family that infects and replicates directly inside hepatocytes,31  but does not integrate into the host genome and does not contain an obvious oncogene.9  Being a positive-stranded RNA virus, HCV has to convert its positive strand into a negative intermediate RNA strand before replication. Detection of a positive HCV-RNA strand indicates the presence of the virus, but only the detection of a negative-stranded intermediate provides evidence of a true viral replication in a target cells. Viral replication has been demonstrated clearly in hepatocytes, supporting a direct role for HCV in the development of hepatocellular carcinoma.31  The HCV-RNA negative strand and the NS3 viral nonstructural protein, which is a main component of the HCV replication cycle and indicates viral mRNA translation and polyprotein production, have been detected in PBMCs.32,33  However, which is the principal HCV-target population of the immune system is still debated, as is the capacity of the virus to replicate in vivo in the infected lymphocytes.34 

Replication in the lymphocytes is not sufficient to fully demonstrate a pathogenetic role in NHL development. The strongest evidence for a causal relationship between HCV and lymphoma comes from the observation of lymphoma regression after antiviral treatment. This was first shown in 9 patients with splenic lymphoma who had a lymphoma remission after IFNα and ribavirin antiviral treatment.35  A direct antitumor effect of IFNα therapy was ruled out (in fact, 6 additional patients with splenic lymphoma, but without HCV infection, had no hematologic response to the antiviral treatment).35  These results were reproduced by other studies showing that viral eradication can be an effective treatment for HCV-associated indolent lymphomas (Table 5). The hematological response rate across different studies is ∼75%.36,37  A large Japanese study comparing 501 consecutive patients with chronic HCV who had never received IFNα therapy with 2708 consecutive HCV-infected patients who received IFNα therapy showed a significantly higher incidence of lymphoma in patients with persistent infection than in patients with sustained virologic response (SVR), demonstrating that HCV eradication protects against the development of malignant lymphoma.22 

Table 5.

Hematological responses after antiviral treatment of HCV-associated indolent NHLs (only series with >3 cases are included)35,44,45,48,55-57 

Hematological responses after antiviral treatment of HCV-associated indolent NHLs (only series with >3 cases are included)35,44,45,48,55-57
Hematological responses after antiviral treatment of HCV-associated indolent NHLs (only series with >3 cases are included)35,44,45,48,55-57

RBV indicates ribavirin; CR, complete response; ORR, overall response rate (complete + partial); SLVL, splenic marginal zone lymphoma with circulating villous lymphocytes; SMZL, splenic marginal zone lymphoma; EMZL, extranodal marginal zone lymphoma of the mucosa-associated lymphoid tissue; and NMZL, nodal marginal zone lymphoma.

*Five additional patients were treated with PegIFNα alone.

The clinical responses observed after IFNα treatment in splenic HCV-positive lymphomas are similar to those observed in gastric MALT lymphoma after antibiotic treatment38  and clearly indicate a pathogenic role for HCV and lymphomagenesis, but do not explain the mechanism(s) by which HCV may promote lymphoma growth. A causal link may be based on different theoretical mechanisms (summarized by Marcucci and Mele9 ): a viral immunosuppressive effect on the tumor cells, but a significant immunodeficiency is not usually detectable; the coinfection by another unknown oncogenic virus, but no evidence has emerged supporting this hypothesis; a direct oncogenic role of HCV; and an indirect, antigen-driven stimulation of the lymphoma growth. These different putative oncogenic mechanisms of HCV-induced lymphoma-genesis do not necessarily have to be mutually exclusive.

Apparently, the lymphoma subtypes that are most frequently HCV associated originate from germinal center or postgerminal center lymphocytes, suggesting a possible antigen-driven proliferation. Indeed, a monoclonal/oligoclonal B-cell expansion has been shown in circulating B cells, as well as in the bone marrow or intrahepatic B cells of HCV-infected patients.31 

The HCV-E2 envelope protein binds the CD81 expressed on B cells. At the same time, HCV may bind a specific BCR on the B-cell surface. It has been hypothesized that dual binding of both the CD19/CD21/CD81 complex and the BCR can result in a decreased threshold for B-cell activation and proliferation.31  This mechanism, however, has been challenged by Ng et al, who did not confirm the ability of lymphoma BCRs from patients with B-NHL and chronic HCV infection to bind HCV antigens.39  Nevertheless, chronic antigenic stimulation may play a significant role in the development of an initially polyclonal B-cell expansion and predispose to genetic aberrations.

The monoclonal rheumatoid factor-like IgM component of the type II MC and monoclonal antibodies derived from HCV-infected patients were found to have restricted combinations of the IGHV gene repertoire.9,10  HCV-NHLs have been found to express analog regions of the IgV genes and to undergo somatic hypermutation, indicating affinity maturation driven by a selective antigen pressure. Analogous to gastric MALT lymphoma associated with Helicobacter pylori chronic infection, it can be hypothesized that, in the context of a persistent antigenic stimulation, successive genetic abnormalities can progressively affect a B-cell clone among the reactive B cells of the chronic inflammatory tissue and give rise to the HCV-associated B-cell lymphoma. In this model, type II MC can be considered a premalignant condition associated with chronic antigenic stimulation of B-cell clones, which, after “a second hit” represented by additional genomic alterations, can progress to overt lymphoma. It should be noted that HCV eradication is an effective treatment for HCV-associated MC. However, cryoglobulinemia may sometimes persist after antiviral treatment as an expression of acquired independence from the antigenic stimulus.

As summarized in a recent review,21  it remains unclear which signaling pathways mediate HCV-induced lymphomagenesis. The proinflammatory IL-6 has been suggested to contribute to the development of cryoglobulinemia and B-cell NHL in HCV-infected patients via stimulation of TLR2.21  In transgenic mice, the expression of B-lymphocyte stimulator/B-activating factor (BLyS/BAFF, a TNF-family cytokine) has appeared associated with the clinical progression of HCV infection and may activate NF-κB, JNK, and ERK pathways, stimulating B-cell proliferation.21  Gene expression profiling of lymphoma B cells from HCV-transgenic mice has revealed a modified expression of various genes, suggesting that the activation of both canonical and alternative NF-κB pathways and down-regulation of miR26b may contribute to lymphomagenesis, and a reduced expression of miR-26b has been found in HCV-positive SMZL patients.21 

Several studies have addressed the role of chromosomal aberration in HCV-induced lymphomagenesis (reviewed in Zignego et al).40  The presence of t(14;18) and the rearrangement of bcl-2 were reported to be present with increased frequency in patients with chronic HCV-related MC and to disappear after successful antiviral therapy, suggesting that the antiapoptotic function of Bcl-2 can be relevant in HCV-infected B cells. Different studies have identified genetic instability, increased aneuploidy, and reduced Rb protein expression in HCV-infected cells, potentially favoring a neoplastic transformation. In addition to the BLyS/BAFF contribution to an increased B-cell survival, other studies have suggested a role of cytokines and chemokines, particularly in MC, in which Th1 cytokines (IFNγ and TNFα) and some chemokines (MIP-1α, MIP-1β, CXCL10 and CXCR-3, CXCL13) levels have been found elevated in the vasculitic lesions, suggesting a potential pathogenic role.

HCV replication in B-lymphocytes is controversial and may be restricted to a subset of the B-cell population, those that are CD5-positive, which also expresses high CD81 levels. Additional events (eg, EBV coinfection) may be needed for the B cell to become permissive for viral replication.21  There is limited evidence of a direct role of intracellular viral proteins. HCV may be able to induce cellular transformation without intracellular viral replication with different putative, still poorly known oncogenes and tumor suppressor genes.34 

Marcucci and Mele have recently reviewed the potential lymphomagenic mechanisms of HCV and proposed a multifactorial model for HCV lymphomagenesis.9  According to this model, all the previously described oncogenic events may individually have a relatively low lymphomagenic potential, and the accumulation of at least 2 oncogenic signals is required for lymphoma occurrence. These required signals may derive from HCV itself or may result from the integration of an HCV signal with HCV-independent events generated by either genetic damage or coinfection with other viruses such as EBV.9 

The purpose of antiviral therapy in HCV-related chronic hepatitis is to prevent hepatic and extrahepatic complications. The recent approval of new direct-acting antiviral agents (boceprevir, telaprevir) that specifically inhibit viral replication has provided a tool to improve the virologic response rate in the resistant genotypes as well.41,42  The current first-line treatment of chronic HCV infection is the combination of pegylated IFNα (PegIFNα) and ribavirin for genotypes 2–6. The addiction of boceprevir or telaprevir is indicated for the treatment of chronic hepatitis C genotype 1. Several other novel compounds, orally available and clinically well tolerated, are being tested in clinical trials in various combinations with or without ribavirin or PegIFNα (Tables 6, 7); some of these will be registered soon.41,42  The indication to treat, as well as the timing and type of treatment, have thus far been based on viral genotype, patient conditions and comorbidities, and degree of liver fibrosis. The novel anti-HCV agents may, however, dramatically change the treatment scenario in the very next future.42 

Table 6.

Efficacy by genotype in recent randomized trials of standard treatments with pegylated interferon-α (PegIFNα) plus ribavirin and the novel regimens based on direct-acting antiviral agents

Efficacy by genotype in recent randomized trials of standard treatments with pegylated interferon-α (PegIFNα) plus ribavirin and the novel regimens based on direct-acting antiviral agents
Efficacy by genotype in recent randomized trials of standard treatments with pegylated interferon-α (PegIFNα) plus ribavirin and the novel regimens based on direct-acting antiviral agents

*Detailed references in Liang and Ghany58  and in the 2014 guidelines of the European Association for the Study of the Liver (EASL).41 

†Patients with cirrhosis were included in the trial.

Table 7.

Anti-HCV oral compounds in clinical development at the beginning of 201459 

Anti-HCV oral compounds in clinical development at the beginning of 201459
Anti-HCV oral compounds in clinical development at the beginning of 201459

In principle, HCV eradication is indicated in patients with lymphoma because it may induce, in certain subtypes, lymphoma regression and may remove the antigenic stimulation that may promote a relapse. However, there are specific management issues associated with presenting clinical features and histological type.

As already mentioned, the use of antiviral therapy to treat HCV-associated lymphomas was established after the demonstration by Hermine et al of SMZL regression after HCV eradication.35  Later, other reports demonstrated that antiviral therapy can be considered as front-line therapy for other HCV-associated indolent B-NHLs.30,43-45  This treatment approach has been more frequently explored in MZLs, in which the achievement of a virologic response can be followed by a lymphoma response in up to 75% of the cases. With respect to the other indolent histologies, hematologic responses to antiviral therapy have been shown in follicular lymphoma and lymphoplasmacytic lymphoma. Although most studies included very small numbers of patients (Table 5), a very recent large retrospective study of 134 patients has confirmed that antiviral therapy is effective in either MZL or non-MZL indolent entities and that HCV-RNA clearance is needed to attain lymphoma response.45  At present, in HCV-positive patients with MZL who do not need immediate conventional treatment for lymphoma, antiviral treatment with PegIFNα and ribavirin should be considered as first-line treatment.45,46  A similar approach may also be considered for the asymptomatic HCV-infected patients with lymphoplasmacytic and follicular lymphomas, but the evidence from published studies is less strong and further studies are needed to proper evaluate the role of front-line antiviral treatment in non-MZL indolent subtypes.

The management of HCV-associated DLBCL is still based on immunochemotherapy with anthracycline-containing regimens in combination with rituximab and there are no standard recommendations.47,48  In these patients, unlike patients with hepatitis B, the immunosuppression induced by rituximab does not usually cause HCV reactivation. Indeed, hepatic flare after rituximab seems to be rare49,50  and associated with a good outcome and low mortality.28,48,51  Nevertheless, it may lead to discontinuation of potentially life-saving immunochemotherapy.51  Although prophylactic antihepatitis B therapy during chemotherapy is the standard of care, concurrent administration of immunochemotherapy with IFN-based HCV treatments is usually not feasible because of the increased hematological and liver toxicity.52  In most cases, the treatment of HCV can be safely given only after the completion of the immunochemotherapy program and may result in prolonged disease-free survival.53  However, the utility of antiviral treatment in DLBCL in remission after cytotoxic chemotherapy has never been studied properly.

Liver disease is a well-known complication of hematopoietic stem cell transplantation (HSCT). HCV infection is not an absolute contraindication for HSCT, but is a cause of morbidity and late mortality. A prospective study of the European Group of Blood and Marrow Transplantation (EBMT) followed over time a cohort of 195 HCV-infected patients (134 had allogeneic HSCT and 61 autologous HSCT). Survival probability at 20 years from HSCT was 82%, with a cumulative incidence of severe liver complications (death from liver failure, cirrhosis, and liver transplantation) of 12%. Antiviral therapy with IFN was given to 85 patients (42 in combination with ribavirin), with a SVR rate of 40%. Toxicity was comparable to other patient populations; no patient developed significant exacerbations of GVHD. Patients receiving antiviral therapy had a trend toward a decreased risk of severe liver complications.54 

The current challenge is to identify the right combination of antiviral drugs with the highest potency and the best toxicity profile. The role of IFN-based treatments may change rapidly and IFN-free regimens may soon become a suitable option for patients with contraindications to this drug.42  This may be particularly advantageous for the treatment of chronic hepatitis C in lymphoma patients. However, whether the safety profile of novel regimens will allow the administration of antiviral treatment concomitantly with chemotherapy is still unknown. In the light of the changing therapeutic paradigm, and with very active oral regimens (eg, the combination of ledipasvir and sofosbuvir) that will become available in many countries, we strongly recommend that treatment modalities are chosen after consultation with an expert gastroenterologist. Ideally, HCV eradication is indicated in all HCV-infected lymphoma patients and the shortest and least toxic regimen should be used to achieve the highest possible SVR rate. Regrettably, the cost of novel HCV treatments will be extremely high and most HCV-infected patients live in areas where these drugs will not be available for many years.

The authors thank Luca Arcaini, Florian Bihl, and Davide Rossi for helpful comments and Sarah Jane Ortelli Giannakis for excellent editorial assistance.

Conflict of interest disclosures: E.Z. is on the board of directors or an advisory committee for Roche, Celgene, and Janssen; has received research funding from Roche and Mundipharma; has consulted for Roche, Mundipharma, Celgene, and Janssen; and has received advisory honoraria from Celgene, Janssen, Gilead, Mundipharma, Roche and Takeda. B.V. declares no competing financial interests. Off-label drug use: None disclosed.

Emanuele Zucca, MD, Oncology Institute of Southern Switzerland (IOSI), Ospedale San Giovanni, CH-6500 Bellinzona, Switzerland. Phone: +41-91-811-90-40; Fax: +41-91-811-91-82; e-mail: ielsg@ticino.com.

1
World Health Organization
Hepatitis C: fact sheet number 164
 
2
Hope
 
VD
Eramova
 
I
Capurro
 
D
Donoghoe
 
MC
Prevalence and estimation of hepatitis B and C infections in the WHO European Region: a review of data focusing on the countries outside the European Union and the European Free Trade Association
Epidemiol Infect
2014
, vol. 
142
 
2
(pg. 
270
-
286
)
3
Zaltron
 
S
Spinetti
 
A
Biasi
 
L
Baiguera
 
C
Castelli
 
F
Chronic HCV infection: epidemiological and clinical relevance
BMC Infect Dis
2012
, vol. 
12
 
suppl 2
pg. 
S2
 
4
Centers for Disease Control and Prevention
Hepatitis C information for health professionals
 
5
Sievert
 
W
Altraif
 
I
Razavi
 
HA
, et al. 
A systematic review of hepatitis C virus epidemiology in Asia, Australia and Egypt
Liver Int
2011
, vol. 
31
 
suppl 2
(pg. 
61
-
80
)
6
Agnello
 
V
Chung
 
RT
Kaplan
 
LM
A role for hepatitis C virus infection in type II cryoglobulinemia
N Engl J Med
1992
, vol. 
327
 
21
(pg. 
1490
-
1495
)
7
Mazzaro
 
C
Tirelli
 
U
Pozzato
 
G
Hepatitis C virus and non-Hodgkin's lymphoma 10 years later
Dig Liver Dis
2005
, vol. 
37
 
4
(pg. 
219
-
226
)
8
Viswanatha
 
DS
Dogan
 
A
Hepatitis C virus and lymphoma
J Clin Pathol
2007
, vol. 
60
 
12
(pg. 
1378
-
1383
)
9
Marcucci
 
F
Mele
 
A
Hepatitis viruses and non-Hodgkin lymphoma: epidemiology, mechanisms of tumorigenesis, and therapeutic opportunities
Blood
2011
, vol. 
117
 
6
(pg. 
1792
-
1798
)
10
Libra
 
M
Polesel
 
J
Russo
 
AE
, et al. 
Extrahepatic disorders of HCV infection: a distinct entity of B-cell neoplasia?
Int J Oncol
2010
, vol. 
36
 
6
(pg. 
1331
-
1340
)
11
Monti
 
G
Pioltelli
 
P
Saccardo
 
F
, et al. 
Incidence and characteristics of non-Hodgkin lymphomas in a multicenter case file of patients with hepatitis C virus-related symptomatic mixed cryoglobulinemias
Arch Intern Med
2005
, vol. 
165
 
1
(pg. 
101
-
105
)
12
Matsuo
 
K
Kusano
 
A
Sugumar
 
A
Nakamura
 
S
Tajima
 
K
Mueller
 
NE
Effect of hepatitis C virus infection on the risk of non-Hodgkin's lymphoma: a meta-analysis of epidemiological studies
Cancer Sci
2004
, vol. 
95
 
9
(pg. 
745
-
752
)
13
Dal Maso
 
L
Franceschi
 
S
Hepatitis C virus and risk of lymphoma and other lymphoid neoplasms: a meta-analysis of epidemiologic studies
Cancer Epidemiol Biomarkers Prev
2006
, vol. 
15
 
11
(pg. 
2078
-
2085
)
14
Negri
 
E
Little
 
D
Boiocchi
 
M
La Vecchia
 
C
Franceschi
 
S
B-cell non-Hodgkin's lymphoma and hepatitis C virus infection: a systematic review
Int J Cancer
2004
, vol. 
111
 
1
(pg. 
1
-
8
)
15
Musto
 
P
Hepatitis C virus infection and B-cell non-Hodgkin's lymphomas: more than a simple association
Clin Lymphoma
2002
, vol. 
3
 
3
(pg. 
150
-
160
)
16
Gisbert
 
JP
Garcia-Buey
 
L
Pajares
 
JM
Moreno-Otero
 
R
Prevalence of hepatitis C virus infection in B-cell non-Hodgkin's lymphoma: systematic review and meta-analysis
Gastroenterology
2003
, vol. 
125
 
6
(pg. 
1723
-
1732
)
17
de Sanjose
 
S
Benavente
 
Y
Vajdic
 
CM
, et al. 
Hepatitis C and non-Hodgkin lymphoma among 4784 cases and 6269 controls from the International Lymphoma Epidemiology Consortium
Clin Gastroenterol Hepatol
2008
, vol. 
6
 
4
(pg. 
451
-
458
)
18
Engels
 
EA
Chatterjee
 
N
Cerhan
 
JR
, et al. 
Hepatitis C virus infection and non-Hodgkin lymphoma: results of the NCI-SEER multi-center case-control study
Int J Cancer
2004
, vol. 
111
 
1
(pg. 
76
-
80
)
19
Giordano
 
TP
Henderson
 
L
Landgren
 
O
, et al. 
Risk of non-Hodgkin lymphoma and lymphoproliferative precursor diseases in US veterans with hepatitis C virus
JAMA
2007
, vol. 
297
 
18
(pg. 
2010
-
2017
)
20
Nieters
 
A
Kallinowski
 
B
Brennan
 
P
, et al. 
Hepatitis C and risk of lymphoma: results of the European multicenter case-control study EPILYMPH
Gastroenterology
2006
, vol. 
131
 
6
(pg. 
1879
-
1886
)
21
Peveling-Oberhag
 
J
Arcaini
 
L
Hansmann
 
ML
Zeuzem
 
S
Hepatitis C-associated B-cell non-Hodgkin lymphomas: epidemiology, molecular signature and clinical management
J Hepatol
2013
, vol. 
59
 
1
(pg. 
169
-
177
)
22
Kawamura
 
Y
Ikeda
 
K
Arase
 
Y
, et al. 
Viral elimination reduces incidence of malignant lymphoma in patients with hepatitis C
Am J Med
2007
, vol. 
120
 
12
(pg. 
1034
-
1041
)
23
Arcaini
 
L
Burcheri
 
S
Rossi
 
A
, et al. 
Prevalence of HCV infection in nongastric marginal zone B-cell lymphoma of MALT
Ann Oncol
2007
, vol. 
18
 
2
(pg. 
346
-
350
)
24
De Vita
 
S
Sacco
 
C
Sansonno
 
D
, et al. 
Characterization of Overt B-Cell Lymphomas in Patients With Hepatitis C Virus Infection
Blood
1997
, vol. 
90
 
2
(pg. 
776
-
782
)
25
Besson
 
C
Canioni
 
D
Lepage
 
E
, et al. 
Characteristics and outcome of diffuse large B-cell lymphoma in hepatitis C virus-positive patients in LNH 93 and LNH 98 Groupe d'Etude des Lymphomes de l'Adulte programs
J Clin Oncol
2006
, vol. 
24
 
6
(pg. 
953
-
960
)
26
Visco
 
C
Arcaini
 
L
Brusamolino
 
E
, et al. 
Distinctive natural history in hepatitis C virus positive diffuse large B-cell lymphoma: analysis of 156 patients from northern Italy
Ann Oncol
2006
, vol. 
17
 
9
(pg. 
1434
-
1440
)
27
Tomita
 
N
Kodama
 
F
Takabayashi
 
M
, et al. 
Clinical features and outcome in HCV-positive aggressive non-Hodgkin's lymphoma
Leuk Lymphoma
2003
, vol. 
44
 
7
(pg. 
1159
-
1164
)
28
Merli
 
M
Visco
 
C
Spina
 
M
, et al. 
Outcome prediction of diffuse large B-cell lymphomas associated with hepatitis C virus infection: a study on behalf of the Fondazione Italiana Linfomi
Haematologica
2014
, vol. 
99
 
3
(pg. 
489
-
496
)
29
Saadoun
 
D
Resche Rigon
 
M
Sene
 
D
, et al. 
Rituximab plus Peg-interferon-α/ribavirin compared with Peg-interferon-α/ribavirin in hepatitis C–related mixed cryoglobulinemia
Blood
2010
, vol. 
116
 
3
(pg. 
326
-
334
)
30
Paulli
 
M
Arcaini
 
L
Lucioni
 
M
, et al. 
Subcutaneous ‘lipoma-like’ B-cell lymphoma associated with HCV infection: a new presentation of primary extranodal marginal zone B-cell lymphoma of MALT
Ann Oncol
2010
, vol. 
21
 
6
(pg. 
1189
-
1195
)
31
Weng
 
WK
Levy
 
S
Hepatitis C virus (HCV) and lymphomagenesis
Leuk Lymphoma
2003
, vol. 
44
 
7
(pg. 
1113
-
1120
)
32
Pawelczyk
 
A
Kubisa
 
N
Jablonska
 
J
, et al. 
Detection of hepatitis C virus (HCV) negative strand RNA and NS3 protein in peripheral blood mononuclear cells (PBMC): CD3+, CD14+ and CD19+
Virol J
2013
, vol. 
10
 pg. 
346
 
33
Ito
 
M
Masumi
 
A
Mochida
 
K
, et al. 
Peripheral B cells may serve as a reservoir for persistent hepatitis C virus infection
J Innate Immun
2010
, vol. 
2
 
6
(pg. 
607
-
617
)
34
Tucci
 
FA
Broering
 
R
Johansson
 
P
Schlaak
 
JF
Kuppers
 
R
B cells in chronically hepatitis C virus-infected individuals lack a virus-induced mutation signature in the TP53, CTNNB1, and BCL6 genes
J Virol
2013
, vol. 
87
 
5
(pg. 
2956
-
2962
)
35
Hermine
 
O
Lefrere
 
F
Bronowicki
 
JP
, et al. 
Regression of splenic lymphoma with villous lymphocytes after treatment of hepatitis C virus infection
N Engl J Med
2002
, vol. 
347
 
2
(pg. 
89
-
94
)
36
Arcaini
 
L
Merli
 
M
Volpetti
 
S
Rattotti
 
S
Gotti
 
M
Zaja
 
F
Indolent B-cell lymphomas associated with HCV infection: clinical and virological features and role of antiviral therapy
Clin Dev Immunol
2012
, vol. 
2012
 pg. 
638185
 
37
Gisbert
 
JP
Garcia-Buey
 
L
Pajares
 
JM
Moreno-Otero
 
R
Systematic review: regression of lymphoproliferative disorders after treatment for hepatitis C infection
Aliment Pharmacol Ther
2005
, vol. 
21
 
6
(pg. 
653
-
662
)
38
Zucca
 
E
Copie-Bergman
 
C
Ricardi
 
U
Thieblemont
 
C
Raderer
 
M
Ladetto
 
M
Gastric marginal zone lymphoma of MALT type: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up
Ann Oncol
2013
, vol. 
24
 
suppl 6
(pg. 
vi144
-
vi148
)
39
Ng
 
PP
Kuo
 
CC
Wang
 
S
, et al. 
B-cell receptors expressed by lymphomas of hepatitis C virus (HCV)-infected patients rarely react with the viral proteins
Blood
2014
, vol. 
123
 
10
(pg. 
1512
-
1515
)
40
Zignego
 
AL
Giannini
 
C
Gragnani
 
L
HCV and lymphoproliferation
Clin Dev Immunol
2012
, vol. 
2012
 pg. 
980942
 
41
EASL Clinical Practice Guidelines: management of hepatitis C virus infection
J Hepatol
2014
, vol. 
60
 
2
(pg. 
392
-
420
)
42
Liang
 
TJ
Ghany
 
MG
Current and future therapies for hepatitis C virus infection
N Engl J Med
2013
, vol. 
368
 
20
(pg. 
1907
-
1917
)
43
Arcaini
 
L
Paulli
 
M
Boveri
 
E
, et al. 
Splenic and nodal marginal zone lymphomas are indolent disorders at high hepatitis C virus seroprevalence with distinct presenting features but similar morphologic and phenotypic profiles
Cancer
2004
, vol. 
100
 
1
(pg. 
107
-
115
)
44
Vallisa
 
D
Bernuzzi
 
P
Arcaini
 
L
, et al. 
Role of anti-hepatitis C virus (HCV) treatment in HCV-related, low-grade, B-cell, non-Hodgkin's lymphoma: a multicenter Italian experience
J Clin Oncol
2005
, vol. 
23
 
3
(pg. 
468
-
473
)
45
Arcaini
 
L
Vallisa
 
D
Rattotti
 
S
, et al. 
Antiviral treatment in patients with indolent B-cell lymphomas associated with HCV infection: a study of the Fondazione Italiana Linfomi
Ann Oncol
2014
, vol. 
25
 
7
(pg. 
1404
-
1410
)
46
Dreyling
 
M
Thieblemont
 
C
Gallamini
 
A
, et al. 
ESMO Consensus conferences: guidelines on malignant lymphoma. part 2: marginal zone lymphoma, mantle cell lymphoma, peripheral T-cell lymphoma
Ann Oncol
2013
, vol. 
24
 
4
(pg. 
857
-
877
)
47
Borchardt
 
RA
Torres
 
HA
Challenges in managing hepatitis C virus infection in cancer patients
World J Gastroenterol
2014
, vol. 
20
 
11
(pg. 
2771
-
2776
)
48
Pellicelli
 
AM
Marignani
 
M
Zoli
 
V
, et al. 
Hepatitis C virus-related B cell subtypes in non Hodgkin's lymphoma
World J Hepatol
2011
, vol. 
3
 
11
(pg. 
278
-
284
)
49
Ennishi
 
D
Maeda
 
Y
Niitsu
 
N
, et al. 
Hepatic toxicity and prognosis in hepatitis C virus-infected patients with diffuse large B-cell lymphoma treated with rituximab-containing chemotherapy regimens: a Japanese multicenter analysis
Blood
2010
, vol. 
116
 
24
(pg. 
5119
-
5125
)
50
Lake-Bakaar
 
G
Dustin
 
L
McKeating
 
J
Newton
 
K
Freeman
 
V
Frost
 
SD
Hepatitis C virus and alanine aminotransferase kinetics following B-lymphocyte depletion with rituximab: evidence for a significant role of humoral immunity in the control of viremia in chronic HCV liver disease
Blood
2007
, vol. 
109
 
2
(pg. 
845
-
846
)
51
Mahale
 
P
Kontoyiannis
 
DP
Chemaly
 
RF
, et al. 
Acute exacerbation and reactivation of chronic hepatitis C virus infection in cancer patients
J Hepatol
2012
, vol. 
57
 
6
(pg. 
1177
-
1185
)
52
Arcaini
 
L
Merli
 
M
Passamonti
 
F
, et al. 
Impact of treatment-related liver toxicity on the outcome of HCV-positive non-Hodgkin's lymphomas
Am J Hematol
2010
, vol. 
85
 
1
(pg. 
46
-
50
)
53
La Mura
 
V
De Renzo
 
A
Perna
 
F
, et al. 
Antiviral therapy after complete response to chemotherapy could be efficacious in HCV-positive non-Hodgkin's lymphoma
J Hepatol
2008
, vol. 
49
 
4
(pg. 
557
-
563
)
54
Ljungman
 
P
Locasciulli
 
A
de Soria
 
VG
, et al. 
Long-term follow-up of HCV-infected hematopoietic SCT patients and effects of antiviral therapy
Bone Marrow Transplant
2012
, vol. 
47
 
9
(pg. 
1217
-
1221
)
55
Kelaidi
 
C
Rollot
 
F
Park
 
S
, et al. 
Response to antiviral treatment in hepatitis C virus-associated marginal zone lymphomas
Leukemia
2004
, vol. 
18
 
10
(pg. 
1711
-
1716
)
56
Mazzaro
 
C
De Re
 
V
Spina
 
M
, et al. 
Pegylated-interferon plus ribavirin for HCV-positive indolent non-Hodgkin lymphomas
Br J Haematol
2009
, vol. 
145
 
2
(pg. 
255
-
257
)
57
Saadoun
 
D
Suarez
 
F
Lefrere
 
F
, et al. 
Splenic lymphoma with villous lymphocytes, associated with type II cryoglobulinemia and HCV infection: a new entity?
Blood
2005
, vol. 
105
 
1
(pg. 
74
-
76
)
58
Liang
 
TJ
Ghany
 
MG
Therapy of hepatitis C–back to the future
N Engl J Med
2014
, vol. 
370
 
21
(pg. 
2043
-
2047
)
59
Pawlotsky
 
JM
New hepatitis C therapies: the toolbox, strategies, and challenges
Gastroenterology
2014
, vol. 
146
 
5
(pg. 
1176
-
1192
)