Treatment and Cancer Control


Virus a

Cancer type b

Antiviral therapy c
 
HBV

HCC

Interferons

(Interferon-α and pegylated interferon-α2a)
  
Nucleos(t)ide analogues

(Lamivudine, adefovir, entecavir, telbivudine, and tenofovir)

HCV

HCC

Peglyated interferon plus ribavirin
   
Direct antiviral agents (protease inhibitor)

(Boceprevir and telaprevir)

KSHV

AIDS-KS

Antiviral herpesvirus drug

(Ganciclovir and valganciclovir)
  
HAART, PI based
   
HAART, NNRTI based
  
HIV-negative KS

HIV protease inhibitor

(Indinavir)

HTLV-1

ATL (acute, chronic, and smoldering forms)

Interferon-α plus zidovudine/zalcitabine alone or combined with chemotherapy
 
EBV

PTLD, NPC, HL

Immunotherapy

(EBV-specific cytotoxic T cells)
 
EBV-associate lymphoma

Virus directed

(Lytic replication inducer plus acyclovir/ganciclovir)

HPV

Cervical cancer

Therapeutic HPV vaccine (focus on HPV-16 and HPV-18)
   
RNA-interference-based therapy

(Antisense oligonucleotides, ribozymes, and siRNAs)

MCV

MCC

Interferon

(Interferon-α and interferon-β)


a HBV hepatitis B virus, HCV hepatitis C virus, EBV Epstein–Barr virus, HPV human papillomavirus, HTLV-1 human T-cell lymphotropic virus type 1, KSHV Kaposi’s associated sarcoma virus, MCV Merkel cell polyomavirus

b HCC hepatocellular carcinoma, AIDS-KS AIDS-Kaposi’s sarcoma, ATL adult T-cell lymphoma, PTLD post-transplant lymphoproliferative disorder, NPC nasopharyngeal carcinoma, HL Hodgkin’s lymphoma, MCC Merkel cell carcinoma

c HAART highly active antiretroviral treatment, PI protease inhibitor, NNRTI: non-nucleoside reverse transcriptase inhibitor



In this chapter, we reviewed the current evidences on the correlation between viral load and clinical outcomes (e.g., cancer risk and survival) and the current understandings on the effect of antiviral treatments for cancer-associated viruses.



2 Hepatitis B Virus and Hepatitis C Virus


Chronic HBV and HCV infections are major etiological factors in 75–80 % of HCC cases worldwide. Universal HBV vaccination on newborns in Taiwan since 1985 has lead to a 70 % reduction in HBV-related HCC in young-aged children (Chang et al. 2009). Nevertheless, millions of individuals who had already chronically infected by HBV are still under the risk of developing HCC. Unfortunately, no effective vaccine is available for preventing HCV infection now. The proportion of HCV-related HCC has progressively increased, especially in the developed countries (El-Serag 2012). For these people persistently infected by either HBV or HCV, antiviral therapies’ target on HBV and HCV can be an effective strategy to reduce risk of HCC.


2.1 Anti-HBV Therapies


HBV replication is the key force to drive the progression of HBV-related diseases (Liaw 2006). HBeAg is a well-known HBV replication marker (Chen et al. 2009; Fang et al. 2003). Epidemiological studies also consistently show that the elevated HBV viral load, which represented a higher HBV replication activity, is associated with high HCC risk, worse progression, and poor survival (Chen et al. 2009). Reducing HBV DNA to undetectable level or induction of HBeAg seroconversion has been the main therapeutic endpoints (Feld et al. 2009).

Conventional interferon-α, pegylated interferon-α2a, and nucleos(t)ide analogues (NUCs), including lamivudine, adefovir, entecavir, telbivudine, and tenofovir, are currently approved treatments for chronic HBV infection. Treatment with newer NUCs, including entecavir, telbivudine, and tenofovir, could suppress HBV DNA on average by 6.2–6.9 and 4.6–5.2 log10 IU/mL in HBeAg-positive (Chang et al. 2006; Lai et al. 2007; Marcellin et al. 2008) and HBeAg-negative patients (Lai et al. 2006, 2007; Marcellin et al. 2008), respectively. Undetectable levels of HBV DNA can be achieved in 60–80 % of HBeAg-positive patients (Chang et al. 2006; Dienstag 2009; Lai et al. 2007; Marcellin et al. 2008) and 88–95 % of HBeAg-negative patients (Dienstag 2009; Lai et al. 2006, 2007; Marcellin et al. 2008). Treatment with lamivudine and adefovir yields less reduction in HBV DNA and lower proportion of undetectable HBV DNA in both HBeAg-positive and HBeAg-negative patients. In addition, HBeAg seroconversion was observed in 12–23 % of NUC-treated patients (Chang et al. 2006; Dienstag 2009; Lai et al. 2007; Marcellin et al. 2008). After one-year course of NUC treatment, sustained virological response was maintained in relatively small proportion of initial responders (Dienstag 2009). Thus, NUCs were usually used as a long-term therapy. No optimal duration of NUC therapy is available now, but extending treatment for at least more than 6 months after HBeAg seroconversion to consolidate the sustained response is currently an acceptable practice.

Although NUCs are highly potent and safe, emergence of drug resistance after prolonged use of NUCs is a major concern. It has been reported that patients with lamivudine resistance had higher HCC risk than NUC-naïve patients (Papatheodoridis et al. 2010). Lamivudine resistance would accumulate rapidly to 15–25 % after 12 months and to 60–65 % after 4 years of treatment (Papatheodoridis et al. 2008). Resistance to adefovir and telbivudine can also reach 25–30 % after long-term treatment. Only treatment with entecavir and tenofovir showed negligible resistance. Thus, due to better resistance profile, excellent safety profile, and superior efficacy, entecavir and tenofovir have now been suggested as the first-line therapy (Dienstag 2009).

In contrast to NUCs, interferon-based therapy was less used for HBV infection due to side effects and poor tolerability. Nevertheless, recent studies have provided more supportive evidence for its role in anti-HBV therapy. Treatment of HBeAg-positive patients with pegylated interferon for 48–56 weeks achieved undetectable HBV DNA in 10–25 % of patients and <2–4.5 log10 copies/mL mean reduction in HBV DNA (Janssen et al. 2005; Lau et al. 2005). HBeAg loss and HBeAg seroconversion were durable in 81 and 70 % of initial responders with or without concomitant lamivudine therapy, respectively, for a mean of 3 years of follow-up after treatment (Buster et al. 2008). The proportion of HBV DNA undetectability in HBeAg-negative patients was 63 %, and average reduction of viral load was 4.1 log10 copies/mL after 48 weeks of therapy of pegylated interferon (Marcellin et al. 2004). After 24 weeks of follow-up (week 72), rate of HBV DNA undetectability decreased to 19 % and average reduction in viral load became 2.3 log10 copies/mL (Marcellin et al. 2004). A durable suppression of HBV DNA to undetectable level was observed in 46 % of HBeAg-negative initial responders at 3 years of follow-up after the end of treatment (Marcellin et al. 2009). Although pegylated interferon resulted in virological response in a small proportion of patients, it seemed to have higher probability to achieve sustained off-therapy response. Due to this advantage and the fixed duration of treatment, pegylated interferon still has a therapeutic role in selected patients.

The long-term effect of anti-HBV therapy with NUCs and interferon on survival and incidence of HCC has also been investigated. For interferon-based therapy, two studies reported that sustained virological responders showed significantly better survival and lower risk of developing HCC (Niederau et al. 1996; van Zonneveld et al. 2004). Compared with non-treated patients, interferon-treated patients had lower HCC incidence with RR of 0.23–0.66 (Table 2). For NUCs, two early, large randomized control trials of treatment for chronic HBV-infected patients with advanced liver diseases showed that lamivudine could reduce risk of disease progression including developing HCC in treated patients and in patients with persistent viral suppression (Di Marco et al. 2004; Liaw et al. 2004). Several meta-analyses consistently concluded that NUC treatment was associated with a lower HCC incidence (Table 2).


Table 2
Meta-analyses of nucleos(t)ide analogues and interferon-based therapy for HBV and HCV infections























































































































































Study

Design

No. of subjects

Therapy

Endpoint

Main findings

HBV

Mommeja-Marin et al. (2003)

26 prospective studies

N = 3,428

(2,524 HBeAg positive)

IFN vs. CN

&

NUCs vs. CN

Histological response

Biochemical response

Serological response

Treatment-induced HBV DNA reduction correlated with histological, biochemical, and serological responses

Sung et al. (2008)

12 studies

N = 2,742

1,292 treated

1,450 untreated

IFN vs. CN

HBV-related HCC

RR = 0.66 (0.48–0.89)
 
5 studies

N = 2,289

1,267 treated

1,022 untreated

NUCs vs. CN

HBV-related HCC

RR = 0.22 (0.10–0.50)

Yang et al. (2009)

11 clinical trials

N = 2,082

1,006 treated

1,076 untreated

IFN vs. CN

HBV-related HCC incidence

RR = 0.59 (0.43–0.81)
 
5 clinical trials

N = 935

516 treated

419 untreated

IFN vs. CN

HBV-related cirrhosis

RR = 0.65 (0.47–0.91)

Wong et al. (2010)

11 studies

N = 2,122

975 treated

1,147 untreated

IFN vs. CN

Overall hepatic events

RR = 0.55 (0.43–0.70)

Liver-related mortality

RR = 0.63 (0.42–0.96)

Papatheodoridis et al. (2010)

21 studies

N = 4,415

3,881 treated

534 untreated

NUCs vs. CN

HCC incidence

HCC incidence:

treated: 2.8 %

untreated: 6.4 %

Zhang et al. (2011)

2 RCTs

N = 1,58

95 treated

63 untreated

Non-maintenance IFN

treated vs. CN

HBV-related HCC incidence

RR = 0.23 (0.05–1.04)

HCV

Cammá et al. (2001)

14 studies

HCV-related cirrhosis

N = 3,109

IFN

treated vs. non-treated

HCC incidence

OR = 0.28 (0.22–0.36)

Papatheodoridis et al. (2001)

11 studies

HCV-related cirrhosis

N = 2178

CN vs. IFN

HCC incidence

OR = 3.02 (2.35–3.89)
 
5 studies

HCV-related cirrhosis

N = 683

Non-SVR vs. SVR

HCC incidence

OR = 3.65 (1.71–7.78)

Zhang et al. (2011)

4 RCTs

HCV patients

N = 378

Non-maintenance IFN

treated vs. non-treated

HCV-related HCC incidence

RR = 0.39 (0.26–0.59)
 
2 RCTs

HCV patients

N = 223

Non-maintenance IFN

treated vs. non-treated

HCV-SVR

RR = 0.30 (0.04–2.15)
 
2 RCTs

Initial non-responders of IFN therapy

N = 1,101

Maintenance IFN

treated vs. non-treated

HCV-related HCC incidence

RR = 0.96 (0.59–1.56)

Singal et al. (2010)

20 studies

HCV-related cirrhosis

N = 4,700

Treated vs. non-treated

HCC incidence

RR = 0.43 (0.33–0.56)
 
14 studies
 
SVR vs. non-SVR

HCC incidence

RR = 0.35 (0.26–0.46)

Qu et al (2012)

8 RCTs

HCV-related cirrhosis

N = 1505

Treated vs. non-treated

HCC incidence

OR = 0.29 (0.10–0.80)
 
3 RCTs

HCV-related cirrhosis

N = 1155

Maintenance IFN therapy

treated vs. non-treated

HCC incidence

OR = 0.54 (0.32–0.90)


CN control group (placebo or no treatment), RR relative risk, OR odds ratio, SVR sustained virological response

IFN interferon, NUCs nucleos(t)ide analogues

RCTs randomized controlled trials


2.2 Anti-HCV Therapies


For HCV, there is still no vaccine available. Fortunately, effective anti-HCV therapies are available to provide good clinical outcomes for HCV patients. Interferon-based treatments by using pegylated interferon with ribavirin are the current standard of care for HCV patients. This regimen resulted in sustained virological response (SVR) in about 50 % of HCV patients with genotype 1 and 80 % of HCV patients with genotypes 2 and 3 (Munir et al. 2010). The achievement of SVR is durable (Hofmann and Zeuzem 2011) and highly associated with good overall clinical outcome, including decreased risk of HCC (Table 2) and liver-related deaths (Masuzaki et al. 2010). Nevertheless, non-SVR patients did not show risk reduction in disease progression, and maintenance of anti-HCV treatment provided no further benefits on clinical outcomes in these patients (Table 2).

High viral load and genotype 1 were used to predict a lower response rate to anti-HCV therapies in chronic hepatitis C patients. But the percentage of non-responders has decreased in recent years because of the advance in therapy. Pegylated interferon combined with ribavirin increased SVR to about 40 % in HCV genotype 1b patients with high viral load (Masuzaki et al. 2010). In addition, these non-responders now could be identified by many clinical markers (e.g., obesity, IL-28 polymorphisms) before treatment to allow treatment optimization (Masarone and Persico 2011). Furthermore, upcoming new drugs (boceprevir and telaprevir, two HCV non-structural 3/4A protease inhibitors) may provide satisfactory HCV RNA suppression in patients with genotype 1 infection, which did not respond to interferon-based regimens (Hofmann and Zeuzem 2011). We are now at the dawn of a new era of more effective and friendlier anti-HCV therapies.


3 HBV–HCV Coinfection


Treatment of HBV–HCV coinfection is important in endemic area because of its fairly high prevalence due to shared routes of transmission and increased risk for liver diseases, including HCC. Although no specific treatment guidelines are established, individualized therapy according to hepatitis virology, history of antiviral treatment, and stage and grade of liver disease is recommended (Potthoff et al. 2010).

Before initiation of therapy, determination of dominant virus infection is required. Combination of interferon/pegylated interferon and ribavirin are current choice of treatment for patients with dominant HCV infection. There were 43–69 % of treated patients who showed HCV-SVR at the end of 24 weeks of therapy with interferon plus ribavirin (Potthoff et al. 2010). After 24 or 48 weeks of combination therapy in genotype 2/3 HCV and genotype 1 HCV, respectively, high HCV-SVR rate can be achieved in both genotypes (83 % for genotype 2/3 and 72 % for genotype 1) (Liu et al. 2009). At the end of 24–48 weeks of follow-up, undetectability of HBV DNA was obtained in 11–18 % for interferon plus ribavirin and 56 % for pegylated interferon plus ribavirin (Liu et al. 2009; Potthoff et al. 2010). Interestingly, the combination therapy seemed to have higher chance of HBsAg clearance, which is the optimal treatment goal. The proportion of HBsAg clearance was 11–19 % for these two types of regimens (Liu et al. 2009; Yeh et al. 2011). However, as severe hepatic flares and recurrence of HBV after therapy have been reported, using this combination therapy with caution and long-term virological monitoring in some coinfected patients are necessary. In HBV-dominant or dually active HBV–HCV-coinfected patients, the optimal therapeutic regimen remains unestablished.


4 HBV–HIV Coinfection and HCV–HIV Coinfection


Coinfection of HBV–HIV and HCV–HIV usually hastens the development of liver diseases, including fibrosis and HCC. Generally speaking, monotherapy of interferons, pegylated interferons, lamivudine, adefovir, entecavir, and telbivudine has yielded much less satisfactory responses in HBV–HIV-coinfected patients (Lacombe and Rockstroh 2012). Nevertheless, tenofovir monotherapy offered some benefits, such as effective virological suppression, good resistance profile, and histological improvement. However, interruption use must be avoided because of viral breakthrough-induced deleterious consequences.

According to the guidelines proposed by the European AIDS Clinical Society (Lacombe and Rockstroh 2012; Soriano et al. 2010), HBV–HIV-coinfected patients who do not need HIV treatment (>500 CD4 + cells per uL) are suggested to receive early combination therapy of tenofovir and lamivudine/emtricitabine or to receive NUC therapy (either adefovir or telbivudine) in “early add-on” strategy when their HBV DNA level was more than 2,000 IU/mL. In this group of patients who have favorable response to interferon therapy, a 48-week course of interferon is suggested. When both HBV and HIV treatments must be applied, the appropriate option for treatment depends on prior use of lamivudine. In lamivudine-naïve patients, combination of tenofovir plus lamivudine/emtricitabine is the therapeutic option. And for lamivudine-treated patients, adding tenofovir, entecavir, or other HIV nucleos(t)ide reverse transcriptase inhibitors is recommended. Besides, lamivudine-containing HAART has been shown to be able to suppress HBV replication in HBeAg-negative HBV–HIV-coinfected patients (Fang et al. 2003). Although the above guidelines have been proposed, more solid evidence is still needed to make evidence-based therapeutic decisions.

In regard to treatment of HCV–HIV-coinfected patients, SVR rates could be achieved to 27–50 % by using treatment of pegylated interferon with ribavirin, which is the current standard therapy. Higher SVR (44–73 %) was observed in genotypes 2 and 3. In contrast, only 17–35 % genotype 1 and genotype 4 patients reached SVR (Lacombe and Rockstroh 2012).

Regimens for HCV–HIV-coinfected patients would be different according to HCV genotype and virological response. For non-HCV genotype 1 patients, in patients with rapid virological response (undetectable HCV viral load at week 4), 24 and 48 weeks of pegylated interferon/ribavirin combination therapy are recommended for genotype 2/3 and genotype 4, respectively. If the virological response is not achieved at week 12, treatment should be stopped. Patients with virological response at week 12 and undetectable HCV RNA at week 24 are recommended to receive 48- and 72-week courses of treatment for genotype 2/3 and genotype 4, respectively (Lacombe and Rockstroh 2012; Soriano et al. 2010). For HCV genotype 1 patients, longer duration of treatment may provide benefits under the conditions of early virological response at week 12 and undetectable virus at week 24. However, patients with higher fibrosis stages are recommended to have therapy of interferon/ribavirin combination plus HCV protease inhibitors. Recent pilot trials showed that this triple therapy in coinfected patients increased the SVR rate when compared with patients receiving combined treatment (Ingiliz and Rockstroh 2012). In addition, based on the results from clinical studies, HAART may offer positive impact on control of prognosis of liver damage in HCV–HIV-coinfected patients, and most of first-line HAART has good fitness in these patients (Jones and Nunez 2011).


5 Kaposi’s Sarcoma-Associated Herpesvirus


Kaposi’s sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is a necessary factor for Kaposi’s sarcoma (KS), the most common malignancy in HIV patients who became immunocompromised. Elevated KSHV viral load was observed more frequently in KS patients than in asymptomatic KSHV-infected patients and was associated with a higher risk of AIDS-KS (Gantt and Casper 2011; Sunil et al. 2010). There is still no effective vaccine for KSHV. Nevertheless, several studies showed that anti-herpes virus drugs, such as ganciclovir and valganciclovir, not only reduced KSHV viral load but also prevented AIDS-KS (Gantt and Casper 2011). Though these small studies still need further confirmation (Gantt and Casper 2011), it will be interesting to examine the effects of these promising agents in combination with DNA synthesis blockers or lytic replication inducer in future clinical trials.

The replication of KSHV strongly depends on HIV-induced immunodeficiency (Mesri et al. 2010). Early use of highly active antiretroviral treatment (HAART) can restore host immunity and decrease the incidence and mortality of AIDS-KS in HAART-treated patients (Bower et al. 2006; Mesri et al. 2010). Compared with that in pre-HAART era, KS incidence in HAART era dropped by sixfold (Sunil et al. 2010). Even in resource-limited regions, early use of HAART can result in reduced KS incidence (Casper 2011). In the individual patient level, HAART is also significantly associated with control of KSHV viremia (Bourboulia et al. 2004). However, for late presenters who had already developed KS at the time of initial diagnosis, HAART alone induces complete remission in only half of these patients (Nguyen et al. 2008). For AIDS-KS patients, who did not reach complete remission with HAART alone, co-administration of HAART and chemotherapy could improve the response rate to 81.5 % (Bower et al. 2006). HAART can be classified to protease inhibitor (PI)-based and non-nucleoside reverse transcriptase inhibitor (NNRTI)-based regimens. No difference in KS incidence rate between these two types of regimens was observed in small observational studies (Gantt and Casper 2011), but PI-based HAART seemed to have better efficacy because more complete remission and relapse were reported when switching therapy from PI-based to NNRTI-based HAART (Gantt and Casper 2011). Nevertheless, whether PI-based HAART does have a therapeutic advantage for KSHV or not, this superiority still requires more convincing evidence to support, and controlled trials with large sample size and optimized HAART regimens are urgently needed.

For KS in HIV-negative patients, a 28-patient trial study showed that treatment with individual ART (indinavir) induced tumor regression and stabilization of disease progression in some patients (Monini et al. 2009). In addition to KS, KSHV was also linked to primary effusion lymphoma (PEL) and multicentric Castleman’s disease (MCD) (Chang et al. 2006), which are rare malignancies with very poor prognosis. Recently, few studies proved that treatment with HAART alone or with other therapy (e.g., monoclonal antibodies) for PEL and MCD may prolong resolution of symptoms (Sunil et al. 2010).


6 Human T-Cell Lymphotropic Virus Type 1


Human T-cell lymphotropic virus type 1 (HTLV-1) is the etiological agent of adult T-cell lymphoma (ATL) (Poiesz et al. 1980). Although only a small proportion (<6 %) of 15–20 million estimated infected people worldwide developed ATL after a 10- to 40-year latent period (Proietti et al. 2005), the prognosis for ATL patients is very poor. Median survivals of acute and lymphomatous ATLs are less than one year (Goncalves et al. 2010).

Compared with asymptomatic carriers, ATL patients had significantly higher level of HTLV-1 proviral DNA and antibody titer (Manns et al. 1999). The quantity of HTLV-1 proviral DNA is a predictive factor for the development of ATL and is correlated with clinical outcomes (Etoh et al. 1999; Iwanaga et al. 2010; Okayama et al. 2004). Based on the accumulated data, antiviral therapy now is one of the treatment options for ATL (Tsukasaki et al. 2009). Treatment for ATL with antiviral drugs was investigated in the early 1980s in Japan, but encouraging improvement was obtained from two trials that combined interferon-α and zidovudine (AZT) in treating ATL patients reported in 1995 (Gill et al. 1995; Hermine et al. 1995). These two studies described impressive high response rate (more than 50 %) and mild toxic effects, and the survival time was prolonged to more than one year. After that, many small studies (all less than 30 patients) using interferon-α and AZT/zalcitabine were performed in France (Hermine et al. 2002), United Kingdom (Matutes et al. 2001), Martinique (French West Indies) (Besson et al. 2002), and United States (Ratner et al. 2009). Overall, these studies showed consistent efficacy of antiviral therapy. Recently, a worldwide meta-analysis with 254 ATL patients treated with interferon-α and AZT combination and/or with chemotherapy provided further evidence that combination of interferon-α and AZT treatment resulted in high response and significantly prolonged survival (Bazarbachi et al. 2010). However, the survival advantage was limited to acute, chronic, and smoldering subtypes. For lymphomatous ATL, chemotherapy seemed to be more effective than antiviral therapy. Besides, treatment with combination of interferon-α/AZT and arsenic trioxide also resulted in promising outcome on 7 patients with relapsed/refractory acute or lymphomatous ATL (Hermine et al. 2004). Another study using 10 chronic ATL patients showed 100 % response to the treatment (Kchour et al. 2009). Although these were preliminary observations, the feasibility of this regimen has been considered.

Only gold members can continue reading. Log In or Register to continue

Stay updated, free articles. Join our Telegram channel

Feb 18, 2017 | Posted by in ONCOLOGY | Comments Off on Treatment and Cancer Control

Full access? Get Clinical Tree

Get Clinical Tree app for offline access