Overview of New Treatments for Hepatitis C Virus: Moving Towards a Public Health Agenda Tracy Swan Hepatitis/HIV Project Director, Treatment Action Group 1
Table of Contents 1. Hepatitis C: Natural History 2. Hepatitis C: Genotypes and Global Distribution 3. HCV Treatment: Goals and Evolution 3.1 HCV Protease Inhibitors 3.2 HCV Non‐nucleoside Polymerase Inhibitors 3.3 HCV Nucleoside/tide Polymerase Inhibitors 3.4 HCV NS5A Inhibitors 4. HCV Target Product/Regimen Profile 5. HCV Treatment Criteria and Strategies 5.1 DAA Pricing and Access 5.2. DAA implementation 6. State of the Field: HCV Drug Development and Research Gaps 6.1 Role of Ribavirin in DAA Regimens 6.2 Excluded and Under‐Represented Populations: Pregnancy, Nursing, Paediatrics, Comorbidities, People Who Inject Drugs, the Elderly 6.3 DAAs and Pharmacogenomics 6. 4 HCV Genotypes 5 and 6 6.5 HCV Drug Resistance and Treatment Sequencing 6.6 Research Coordination and Opportunities 7. The HCV Treatment Pipeline 7.1 Sofosbuvir‐based FDCs 2.2 Grazoprevir/Elbasvir‐Based Regimens 8. Conclusion 9. References 2
Figure 1. Global Distribution of HCV Genotypes Figure 2. Evolution of HCV Treatment Figure 3: The First Generic DAAs Figure 4. Minimum Cost for HCV Treatment, Diagnostics, Genotyping and Monitoring 

Table 1. Global Distribution of HCV Genotypes, By Income Classification Table 2. DAAs: Regulatory Approval in the European Union and the United States Table 3. Target Product Profile: Viekira Pak, Daklinza, Sovaldi, Harvoni and Olysio  Table 4. Current HCV Treatment Criteria Table 5. Sofosbuvir + GS‐5816* With or Without RBV in Phase 2 Table 6. SVR among Cirrhotic Participants in C‐WORTHY 3
AIDS acquired immune deficiency syndrome ART antiretroviral therapy ARVs antiretrovirals DAAs direct‐acting antivirals DCV daclatasvir EMA European Medicines Agency EU European Union FDA Food and Drug Administration FDC fixed‐dose combination G genotype; as in genotype 1 (G1), genotype 2 (G2), and genotypes 3, 4, 5 and 6 HCC hepatocellular carcinoma HCV hepatitis C virus HIC high‐income country HIV human immunodeficiency virus LIC low‐income country LMIC low‐and middle‐Income countries MIC middle‐income country MSM men who have sex with men NSP needle syringe programme OST opioid substitution PEG‐IFN pegylated interferon PK pharmacokinetic PWID people who inject drugs RAVs resistance‐associated variants RBV ribavirin RLS resource limited setting SOF sofosbuvir SVR sustained virologic response SVR‐12 sustained virologic response, 12 weeks after treatment completion; equivalent to cure US United States WHO World Health Organization 4
W
Worldwide,, an estimatted 185 million people have been iinfected witth hepatitis C virus (HCV); 170 million are chronically infected. TThe highest p
prevalence of hepatitiss C is found in ome countriies (MICs). middle‐inco
hrough unsccreened orggans, blood and blood Hepatitis C is blood borne. It is transmitted th
products; via injection drug use with shared n
needles, syrringes, filterrs and otherr where infecttion controll procedures are paraphernaalia; in medical or dentaal settings w
inadequate; by tattooing with shared equipm
ment, ink and
d inkwells aand sexuallyy, especially between HIIV‐positive m
men who haave sex with
h men. HCV can be vertically transmitted.. Vertical trransmission incidence rranges from
m 3% to 10%
%; HIV coinfecttion increasees the risk. Currently, tthere is no w
way to reduce the risk o
of, untreated H
o
or prevent vvertical HCV
V transmissiion. 1. Hepatittis C: Natu
ural Historry Heepatitis C beecomes chro
onic in 75% of adults, and 80% of cchildren. Otthers will sp
pontaneously cleear their infection, usuaally within m
months. Reinfection is possible in people who
o spontaneously cleared HCV, or weree successfully treated. Co
onsequences of untreatted chronic HCV infection include d
depression,, cardiovasccular and au
utoimmune disorders and liver scarring. Liver damage is accelerated
d in males, aand by HIV
V coinfectio
on, alcohol intake and other factors, especially duration of infection
n. In otherw
wise heealthy childrren, progresssion to cirrhosis rarelyy occurs during the firstt decade of infection. A
After 20
0 years of inffection, 16%
% of adults d
develop cirrrhosis; this iincreases exxponentiallyy to 40% aftter 30
0 years. Peo
ople with cirrrhosis are aat risk for heepatocellulaar carcinoma and liver ffailure. Each
h year, 700,000
0 people die from HCV‐related causses. Hepatitis C becomes ch
hronic in ~75% of infectted adults; tthe remainiing 25% usu
ually clear months afterr initial infecction, an ou
utcome know
wn as infection within the firsst 3 to 12 m
sspontaneou
us viral clearrance.1 Peo
ople can beccome reinfeected with h
hepatitis C aafter sspontaneou
us viral clearrance or succcessful treaatment. A
Although it is often inittially asymp
ptomatic, un
ntreated, ch
hronic hepattitis C infecttion can w
wreak havo
oc outside of the liver, aas well as caausing liver damage. Up
p to 74% of people with
h cchronic HCV
V suffer from
m extrahepaatic manifesstations—evven in the aabsence of sserious liverr d
damage, inccluding dep
pression, fatigue, anxietty, autoimm
mune and deermatologiccal disorderss, increased riisk for strokke and lymp
phoma, and cardiovascu
ular, neuroccognitive, ceentral 2,3,4,55,6,7
nervous sysstem, renal and bone damage.
5
Hepatitis C may contribute to non‐liver related mortality. People with hepatitis C are dying two decades earlier from non‐liver related illness (such as cardiovascular disease, respiratory failure, hypertension and diabetes) than their uninfected peers.8 Spontaneous viral clearance occurs in 20% of children with HCV infection.9,10 Chronic hepatitis C is usually asymptomatic in children. During the first decade of infection, progression to cirrhosis and end‐stage liver disease is rare among otherwise healthy children.11,12,13
Over time, chronic hepatitis C causes liver scarring. The progression of liver damage is not linear, and it varies, by host and viral factors that include male sex, HIV status, coinfection with hepatitis B virus, alcohol intake, obesity, type 2 diabetes, steatosis (fatty liver) and aging.Error! Bookmark not defined. Duration of infection accelerates liver damage: after 20 years of HCV infection, the estimated prevalence of cirrhosis is 16%; at 30 years, it increases to 40%.14 People with cirrhosis are at risk for hepatocellular carcinoma (HCC) at the rate of 1% to 5% per year, and liver failure at the rate of 3% to 6% per year.Error! Bookmark not defined. Each year, 700,000 people die from these HCV complications.15 As of 2012, cirrhosis of the liver was the tenth leading cause of death in lower‐middle‐income countries, and liver cancer was the ninth leading cause of death in upper‐middle‐income countries.16,17 In certain high‐income countries, where access to antiretroviral therapy (ART) for HIV is widespread, more people are now dying from HCV complications than AIDS.18,19,20 2. Hepatitis C Genotypes and Their Global Distribution There are seven known HCV genotypes—each with dozens of subtypes.21,22 The high genetic variability of hepatitis C virus (>30% nucleotide difference between genotypes, 15% to 30% nucleotide difference among subtypes) has made it difficult to develop universally effective vaccines and treatment.22 In fact, the type, duration and outcome of HCV treatment may vary according to genotype—and sometimes subtype. Globally, genotype 1 predominates (46.2%, or 83.4 million cases), followed by genotype 3 (30.1%, or 54.3 million cases), genotype 2 (9.1%, or 16.5 million cases), genotype 4, (8.3%, or 15 million cases), genotype 6 (5.4%, or 9.8 million cases), and genotype 5 (<1%, or 1.4 million cases); there is a single known case of genotype 7.21 Mixed infections, with more than one hepatitis C genotype or subtype have been reported in people with multiple exposures, such as transfusion and blood product recipients, dialysis patients and people who inject drugs (PWID).23,24 6
Figure 1. G
Global Disstribution of HCV Geenotypes
Used with peermission fro
om: Messina JP, Humphreeys I, Flaxman A, et al. Global distribu
ution and prevalence o
of hepatitis C
C virus genotyypes. Hepato
ology. 2015 Jan;61(1):77‐‐87. doi: 10.1002/hep
p.27259. Pan‐genotyypic hepatitis C treatmeent regimen
ns will simpliify scale‐up,, procuremeent and d
delivery of HCV treatm
ment. Many direct‐actin
ng antiviralss (DAAs) havve been devveloped primarily fo
or genotype 1; some aree effective aagainst multtiple HCV geenotypes. D
DAAs are lesss eeffective for people with genotypee 3 and cirrh
hosis. Safetty and efficaacy data in ggenotypes 4
4, 5 and 6 are limited to in vitro stud
5
dies and smaall numberss of study paarticipants. TTable 1. G
Global Disttribution o
of HCV Geenotypes, By Income Classificcation HCV genotype %, Low
w‐Income Countrries %, Low
wer Middlee‐Income Counttries %, Uppeer Middle‐In
ncome Countrie
es 1 40.3 30.7 60.6 2 8.3 6.7 10.8 3 25.4 44.4 15.9 4 18.5 17.7 0.6 5 5.9 0.2 1.5 6 1.7 0.4 10.7 A
Adapted and
d used with p
permission frrom: Messinaa JP, Humphrreys I, Flaxmaan A, et al. G
Global d
distribution aand prevalen
nce of hepatiitis C virus geenotypes. Heepatology. 20
015 Jan;61(1)):77‐87. doi: 10.1002/hep
p.27259. 7
V Treatmen
nt: Goals aand Evolu
ution 3. HCV
The goal of HCV
V treatmentt is cure (also referred tto as sustain
ned virologic response [SVR]). Bein
ng cured redu
uces the riskk for liver‐reelated and aall‐cause mo
orbidity and
d mortality. CV has imprroved dramatically. Peggylated interferon and ribavirin‐‐ aa partially The standard off care for HC
bilitating sid
de effects— is being rep
placed by saafe, tolerablle oral effecctive regimeen with potentially deb
direcct‐acting an
ntivirals (DAAs). These d
drugs have ccured >90%
% of people iin clinical trrials, in only 12 weeks. DAA
A regimens aare suitable for resourcce‐limited seettings—wh
here HCV is rampant, siince these d
drugs simp
plify treatment procureement and d
delivery, improve HCV ttreatment o
outcomes, aand could saave millionss of livves. Therre are four cclasses of diirect‐acting antivirals: p
protease inh
hibitors, non‐nucleosid
de polymeraase inhib
bitors, nucleeoside/tide polymerasee inhibitors and NS5A inhibitors, each with diffferent charracteristics.
Currrently, two ffixed‐dose ccombination
ns (sofosbuvvir/ledipasvvir and Omb
bitasvir/paritaprevir/rito
onavir plus dasaabuvir), and three DAAss (sofosbuvir, a nucleottide polymeerase inhibittor, daclatassvir , an NS5
5A inhibitorr and simeprevir,, an HCV pro
otease inhib
bitor) have b
been approved in high‐‐income cou
untries. TThe goal of hepatitis C treatment iis to cure th
he virus, an outcome reeferred to ass sustained vvirologic ressponse (SVR
R‐12; no hep
patitis C viru
us can be deetected in aa blood sample 12 weekks aafter finishing treatment). SVR is d
durable; aftter being cu
ured, some p
patients havve been 25
ffollowed for two decad
des and rem
main virus‐frree. Being cured reduces the riskk for liver‐
related and all‐cause m
morbidity an
nd mortalityy, including ffrom cardio
ovascular dissease, at all 26,27,228,29,30,31
sstages of livver disease, and regardless of HIV status.
Until recenttly, hepatitis C was treaated with 6 to 12 montths of weeklly pegylated
d interferon (PEG‐IFN) in
njections, an
nd twice‐daaily ribavirin (RBV) tableets—a partially effectivve regimen, w
with side efffects that ccould be deb
bilitating an
nd even life tthreateningg. The toxicity, ccomplexity,, monitoringg requiremeents, and lim
mited efficaccy of PEG‐IFFN‐based heepatitis C ttreatment m
make it undesirable—eespecially fo
or resource‐limited setttings (RLS). TThe interferron‐free HC
CV treatmen
nt era began
n in 2011. Reegulatory aggencies app
proved the ffirst HCV prrotease inhibitors (initiaally used with PEG‐IFN and RBV), aand researchers d
demonstratted proof‐off‐concept fo
or curative o
oral therapyy.32 Since th
hen, numero
ous trials of interferon‐ffree, oral direct‐acting antiviral (DA
AA) regimen
ns have rep
ported cure rates ≥ 85%
% 33,34
4,35,36,37
regardless o
of HCV geno
otype‐‐manyy in only 12 weeks. By 20
014, several options fo
or 8
HCV treatment became available‐‐ primarily in HICs‐‐where hepatitis C treatment is shifting to DAA combinations. Figure 2. Evolution of HCV Treatment Source: http://pennstatehersheygireport.org/2013/06/27/new‐developments‐in‐the‐rapidly‐
evolving‐landscape‐of‐hepatitis‐c‐therapy/ (did not request/obtain permission since I found it while searching for copyright‐free images) 9
Table 2. DAAs: Regulatory Approval in the European Union and the United States DAA or Regimen
Class, Genotypes
Approval formulation Sofosbuvir Used with PEG‐IFN and RBV or RBV alone; simeprevir or daclatasvir, with or without RBV, for 12 or 24 weeks Simeprevir Used with: PEG‐IFN and RBV, or sofosbuvir, with or without RBV, for 12 or 24 weeks Daclatasvir Used with PEG‐IFN and RBV orsofosbuvir, with or without RBV, for 12 or 24 weeks Sofosbuvir/ledipasvir Used with or without RBV, for 8, 12 or 24 weeks Nucleotide polymerase inhibitor 1,2,3,4
(less data in genotypes 5 and 6) 2013 (EU and US)
Protease inhibitor 1 and 4 2013 (US) 2014 (EU) NS5A inhibitor
2014 (EU) Submitted in 2015 (US) 1,2,3,4 (no data in genotypes 5 and 6) FDC: nucleotide 1,3,4 and 6 2014 (EU and US) polymerase inhibitor and NS5A inhibitor 1 and 4 2014 Ombitasvir/paritaprevir/ritonavir FDC: NS5A plus dasabuvir inhibitor, Used with or without RBV, for 12 ritonavir‐boosted or 24 weeks HCV protease inhibitor used with a non‐
nucleoside polymerase inhibitor DAA regimens are shorter, safer, better tolerated and significantly more effective across host and viral factors than PEG‐IFN and RBV. These regimens require minimal monitoring during and after treatment, so they are far simpler to administer and undergo than interferon‐based treatment. Although other approaches to treating hepatitis C virus—such as micro‐RNA and host‐
targeting agents—are under study, none have been approved. There are four classes of 10
DAAs: protease inhibitors, non‐nucleoside polymerase inhibitors, nucleoside/tide polymerase inhibitors and NS5A inhibitors. 3.1 HCV Protease Inhibitors In 2002, BILN‐2061, the first HCV protease inhibitor entered human trials; it was discontinued for cardiotoxicity shortly thereafter.38 The first approved DAAs were HCV protease inhibitors (boceprevir and telaprevir). These drugs were used as part of response‐
guided therapy with PEG‐IFN and RBV. Although they increased SVR (from approximately 45% to approximately 70%) in genotype 1, boceprevir and telaprevir were associated with serious adverse events and added to cost and complexity of treatment. Now, once‐daily HCV protease inhibitors (simeprevir and paritaprevir/ritonavir) are part of all‐oral DAA combinations for HCV genotypes 1 and 4. HCV protease inhibitors are less effective for genotype 1a, due to a lower resistance barrier. Some protease inhibitor‐based regimens require ribavirin for genotype 1a, or are used only for genotype 1b. Most HCV protease inhibitors are not pan‐genotypic—they are not sufficiently effective against genotype 3. But it may be possible to overcome this Achilles heel by combining them with other DAAs . Merck is exploring this strategy by adding sofosbuvir to their grazoprevir/elbasvir (HCV protease inhibitor and NS5A inhibitor fixed‐dose combination (FDC). HCV protease inhibitors have a propensity for drug‐drug interactions and cannot be co‐
administered with several commonly used medications, including some HIV antiretrovirals (ARVs). Newer protease inhibitors may be more potent overall, have a higher genetic barrier to drug resistance, and activity against drug‐resistant strains of hepatitis C. The protease inhibitors currently in phase 2, ABT‐493 (under study as an FDC with ABT‐530, AbbVie’s NS5A inhibitor) and Gilead’s GS 9857 (under study sofosbuvir/velpatasvir [nucleotide polymerase inhibitor/ NS5A inhibitor] FDC), may be more effective against genotype 3. 3.2 HCV Non‐Nucleoside Polymerase Inhibitors The potency and resistance barrier of drugs in this class range from low to moderate; many have been discontinued for toxicity or lack of efficacy. Most non‐nucleoside polymerase inhibitors are twice‐daily drugs, primarily active against genotype 1. Although non‐
nucleosides contribute to regimen efficacy for HCV genotype 1, the need for, and future of drugs from this class is unclear.39,40 3.3 HCV Nucleoside/tide Polymerase Inhibitors Sofosbuvir, the only approved nucleotide polymerase inhibitor, has the best features of this class: multi or pan genotypic activity, potency and high resistance barrier, low propensity 11
ffor drug‐dru
ug interactio
ons, and on
nce daily dossing. Althou
ugh HCV nuccleotides haave great potential, to
oxicity or lack of efficaccy has hindeered their developmen
nt in the passt. Fortunatelyy there are o
other nucleo
otide polym
merase inhib
bitors in early‐stage devvelopment: Merck’s uridine analogg MK‐3682 (formerly ID
DX 21437) is in phase 2. Achillion’s uridine aanalog polyymerase inhibitor, ACH‐‐3422, is in phase I. Jan
nssen’s AL‐3
335, a uridin
ne analog, iss in phase I, aand AL‐516, a purine an
nalog in pre‐clinical devvelopment. 3
3.4 HCV NS5A Inhibito
ors HCV NS5A inhibitors arre a completely novel d
drug class. M
Many—but n
not all‐‐NS5
5A inhibitorss aare pan‐gen
notypic (alth
hough they have been studied in o
only a handfful of peoplee with HCV ggenotypes 5
5 and 6). Th
hese drugs aare once daiily; their pro
opensity forr drug‐drug interactionss vvaries. NS5A inhibiitors are pottent, yet mo
ost have a lo
ow barrier tto resistance (although
h some ccandidates in early devvelop may h
have a higheer barrier to
o, or activity against, ressistant viruss). Pretreatmeent NS5A ressistance doees not alwayys preclude being cured, especiallyy when thesse d
drugs are co
ombined wiith potent D
DAAs. But fo
or people w
who are not cured, longgevity and impact of N
NS5A resistance on HCV
V treatment options aree unclear. W
With this classs, additional research is needed to fforestall or o
overcome d
drug resistan
nce. 4
4. HCV: Taarget Prod
duct/Regim
men Profile Th
he characterristics of an ideal regim
men include ssafety and u
universal effficacy (curee rate of ≥85
5% across all pop
pulations, including during pregnan
ncy and nurssing, in paediatrics and
d people witth n; and otherr common comorbiditiees). Treatmeent must bee simple, witth cirrrhosis; HIV coinfection
miinimal safetty and efficaacy monitorring, and con
nvenient—p
preferably o
once daily‐‐sso it is easy to ad
dminister an
nd undergo. Daata is needeed on DAAs d
during preggnancy and n
nursing; thee first paediatric studies are un
nderway. Deespite gaps iin knowledgge and limittations of cu
urrently available DAAss, they have maany of the ccharacteristiics needed tto address H
HCV globallyy. nce of HCV ggenotype 1—especiallyy in high‐inccome countries—DAA Due to the predominan
development has been
d
n focused on
n it. All DAA
A classes work against ggenotype 1—
—although mon strateggy most are more effectivve against geenotype 1b than genottype 1a. Thee most comm
12
for treating HCV genotype 1 is using DAAs from multiple classes, to shorten treatment and increase cure rates. Although cure rates in trials combining DAAs from 3 classes have topped 95%, those who are not cured may be left with few options. Pan genotypic strategies are becoming more desirable, due to the global focus on HCV. Currently, sofosbuvir is the optimal backbone for a first‐line regimen, since it is pan‐
genotypic, can be used in cirrhosis, has a low propensity for drug‐drug interactions, and a high resistance barrier. Pairing it with a pan‐genotypic NS5A inhibitor obviates pre‐
treatment genotypic testing, and will minimize safety and efficacy monitoring during and after treatment. But the weakness of NS5A inhibitors is baseline or emergent drug resistance that may limit their effectiveness.41,42,43,44,45 Candidates in phase II development (AbbVie’s ABT‐530, Achillion’s ACH‐3102, and Merck’s MK‐8408) are purported to be more potent, and active against drug resistant virus. The ideal DAA regimen for HCV treatment scale up does not exist—yet (especially in regard to affordability an use in pregnancy, nursing and paediatrics). These characteristics are: Safe and Tolerable; preferably ribavirin‐free; minimal pre‐treatment assessment or safety/efficacy monitoring needed; 





Effective and Durable: potent, with high genetic barrier and SVR ≥85%, regardless of age, treatment experience, HIV status, HCV genotype, liver disease severity, kidney disease; Universal: pan‐genotypic; safe for use during pregnancy and nursing, and in pediatrics, HIV/HCV, and cirrhosis; o In paediatrics, age appropriate formulation, scored, dispersible tablets, usable across broad weight bands Simple and convenient: manageable drug‐drug interactions with ARVs, opioid substitution treatment (OST) and other commonly‐used medications; fixed duration (≤ 12 weeks); minimal requirements for pre‐treatment assessment and safety/efficacy monitoring during and after treatment; once‐daily (FDC preferred), without food requirement; Stable at high and low temperatures Affordable. Table 3. Target Product Profile: Ombitasvir/paritaprevir/ritonavir plus dasabuvir, Daclatasvir, Sofosbuvir, Sofosbuvir/ledipasvir and Simeprevir Error! Bookmark not defined.,Error! Bookmark not defined.,Error! Bookmark not defined.,Error! Bookmark not defined.,Error! Bookmark not defined. 
13
REGIMEN, STATUS, SPONSOR (s) UNIVERSAL and SAFE SIMPLE
EFFECTIVE
Assumptions: DAAs or regimens are well tolerated, as evidenced by low discontinuation rates in clinical trials (<5%). They are safe‐‐but less effective‐‐ for people with compensated cirrhosis, especially in G3. No data during pregnancy and nursing, or pediatrics SVR >85% COMMENTS
Pan‐
Data in genotypic HIV/HCV Hepatic impairment Renal ≤12 Propensity QD impairment weeks for DDIs for all G1 and G4 only SVR is comparable Not recommended for Child‐Pugh B; contraindicated in Child‐Pugh C No dose adjustment for mild, moderate or severe renal impairment NO HIGH NO YES Complex, subtyping required, RBV needed in some populations SVR is comparable No dose adjustment for mild, moderate or severe hepatic impairment No dose adjustment for mild, moderate or severe renal impairment ? MODERATE YES YES, except in G3 and cirrhosis BMS Pan‐
genotypic in vitro; studied in G1, G2, G3 and G4 Optimal duration with sofosbuvir unclear for G3 and cirrhosis; RBV may be needed; contraindicated in pregnancy; no in vivo data in G5 and G6 Sofosbuvir YES YES, SVR is comparable No dose adjustment for mild, moderate or severe hepatic impairment No dose adjustment for mild or moderate renal impairment; no data or dose for severe renal impairment Varies LOW YES YES, except in G3 and cirrhosis Used with RBV in G2 and G3; very limited in vivo data in G5 and G6; duration depends on genotype and regimen NO; G1,3, 4 and 6 YES, SVR is comparable No dose adjustment for mild, moderate or severe hepatic impairment No dose adjustment for mild or moderate renal impairment; no data or dose for severe renal Varies MODERATE YES YES, except in G3 RBV required in G3; under study in G2; no in vivo data in G5; limited in vivo data in G6 Ombitasvir/ paritaprevir/ ritonavir and dasabuvir APPROVED in EU and US AbbVie Daclatasvir APPROVED in EU; submitted in US Gilead Sciences APPROVED in EU and US Sofosbuvir/ ledipasvir) APPROVED in EU and US Gilead Sciences 14
impairment Simeprevvir APPROVED in EU and US Janssen NO; G1 and G4 SSVR with P
PEG‐IFN and R
RBV is c
comparable; n
no i
interferon‐
f
free trials in H
HIV/HCV No dose adjustment forr mild hepatic o impairment; no
data or dose in
n moderate or severe hepatic impairment No dose adjustmentt for mild, or moderate o
severe renaal impairment; no data in severe renaal impairment Varies HIGH YES YES, witth Sovaldi RBV may be needed;; sun protectiion needed during use to aavoid photoseensitivity SSource: Adap
pted and useed with perm
mission, from UNITAID 201
15 Hepatitis C Medicines / Technologgy aand Market Landscape. namics/publiications/HCV
V_Meds_Land
dscape_Feb2
2015.pdf http://unitaiid.org/imagees/marketdyn
TThe currentt pan genotyypic optionss are sofosb
buvir, with d
daclatasvir , or sofosbuvvir with ribavirin—aalthough curre rates in p
people with genotype 3
3 and cirrho
osis with theese regimens .37
have rarely topped 60%
% With so
ofosbuvir an
nd daclatasvvir, extendin
ng treatmen
nt to 24 w
weeks, and//or adding rribavirin maay do the triick; ongoingg trials are eexploring 12
2, 16 or 24 5. HCV Trreatment Criteria an
nd Strateggies Each year, 7
700,000 peo
ople die from complicaations of HCV, a curablee virus. HCV
V treatment has individual and public heealth benefits. Currentt HCV treatm
ment rates aare low, ran
nging from 1
1% to <<5%, in partt due to the high price o
of DAAs. Th
his has led tto restrictions, and prio
oritizing peo
ople with tthe most ad
dvanced liveer disease—
—a strategy tthat will red
duce HCV‐asssociated illn
ness and deeath, but w
won’t stop tthe epidemic from spreeading. W
When comb
bined with p
primary prevvention, DA
AAs increasee feasibility of HCV treaatment‐as‐p
prevention ((TasP). Mod
dest increasees in HCV trreatment raates among people who
o inject druggs could draamatically rreduce prevvalence in th
his population. TThe same stteps that haave led to drrastic reducctions in thee price of HIV
V antiretrovvirals can bee used w
with DAAs. In fact, theyy can be maass‐produced profitablyy, and remain affordablle. D
Diagnostics and monito
oring can alsso be simpliified, leadin
ng to a total cost of lesss than US$ 5
500 to d
diagnose an
nd cure hepaatitis C. A
A public heaalth approacch can be used for glob
bal HCV treaatment scalee up: selecting a univerrsal first‐
line regimen
n that minim
mizes safetyy and efficaccy monitorin
ng. 15
To stem an annual death toll of 700,000 people, global HCV treatment scale‐up must be prioritized.15 HCV treatment has the potential to provide vast individual and public health benefits. But high prices have limited access to HCV treatment‐‐even in high‐income countries‐‐to people with advanced liver disease (METAVIR F3 and F4). In the United States, public and private payers have imposed non‐evidence based restrictions (such as requiring 3 to 12 months of abstinence from drugs and alcohol), and forged exclusivity agreements with pharmaceutical companies that limit clinical decision‐making.46,47,48,49 In Europe, high DAA prices have led France to threaten taxation for costly HCV drugs, delayed their approval in the United Kingdom, or limited access in Spain.50,51,52 If DAAs are to stem HCV‐related morbidity and mortality in the coming years, and reduce the global burden of HCV infection, current HCV treatment rates of 1% to <5%‐‐ must increase.53,54,55 Treating people with fibrosis (METAVIR SCORE: >F2) will reduce HCV‐
associated morbidity and mortality—but unless people with less liver damage (METAVIR SCORE: F0 and F1) are treated, the epidemic will continue to spread.56 Table 4. Current HCV Treatment Criteria57,58,59 Source Recommendation All adults and children with chronic HCV infection, including Guidelines for the Screening, Care and PWID, should be assessed for antiviral treatment...patients with Treatment of Persons with Hepatitis C advanced fibrosis and cirrhosis (METAVIR stages F3 and F4) Infection should be prioritized for treatment as they are at higher risk of developing cirrhosis and hepatocellular carcinoma. If resources World Health Organization permit, then persons with less advanced fibrosis (METAVIR April 2014 stages F1 and F2) could also be considered for treatment. EASL Recommendations on Treatment of All treatment‐naïve and –experienced patients with Hepatitis C 2014 compensated disease due to HCV should be considered for European Association for the Study of the therapy. Treatment should be prioritized for patients with Liver (EASL) significant fibrosis (METAVIR Score F3 to F4). Treatment is April 2014 justified in patients with moderate fibrosis (METAVIR Score F2). In patients with no or mild disease (METAVIR Score F0‐F1), the indication for, and timing of therapy can be individualized. Recommendations for Testing, Managing Treatment is recommended for patients with chronic HCV infection. Immediate treatment is assigned the highest priority and Treating Hepatitis C American Association for the Study of Liver for those patients with advanced fibrosis (Metavir F3), those Diseases/ Infectious Diseases Society of with compensated cirrhosis (Metavir F4), liver transplant recipients, and patients with severe extrahepatic hepatitis C. America (AASLD/IDSA) March 2015 16
In Egypt, home of the world’s highest HCV prevalence (14.7%), modeling the impact of different eligibility criteria on life‐years saved found that limiting treatment to people with advanced liver disease (METAVIR SCORE: F3 and F4) was most effective—but the model did not consider the annual infection rate, or the impact of these restrictions on transmission.60 Affordable DAAs increase the appeal and feasibility of HCV treatment‐as‐prevention (TasP). Primary prevention measures must be scaled up in tandem with TasP if it is to be fully effective. In Egypt, HCV is still spreading despite a national treatment program; scaling up infection control to prevent healthcare‐ and household‐associated exposures is as important as providing widespread HCV treatment.61 For people who inject drugs—the highest‐prevalence population‐‐ DAAs and high coverage opioid substitution treatment (OST) and needle and syringe programmes (NSP) — are essential for reducing HCV prevalence (PWID).62 Scaling up HCV treatment access ‐‐and rates‐‐among PWID could dramatically reduce prevalence in this population (estimated at 67%, or at least 10 million people).63 Martin and colleagues modeled the impact of DAA treatment scale‐up, based on a 90% SVR rate, HCV prevalence and current treatment rates among people who inject drugs in three cities. In Edinburgh (where prevalence among PWID is currently 25%, and 7/1000 are treated annually) and Melbourne (where prevalence is 50%, and 3/1000 are treated annually). In just 15 years, prevalence among people who inject drugs could be halved, by scaling up the annual treatment rate in these cities to 22/1000. In Vancouver, where HCV prevalence among PWID is 65% and 5/1000 are treated annually; treating 98/1000 would halve it in 15 years. Prevalence could be reduced to 30% in Vancouver and less than 5% in Edinburgh and Melbourne by treating 80/1000 annually.64 5.1 DAA Pricing and Access Although there are several barriers to universal HCV treatment access, high DAA prices are the greatest obstacle. The same steps that have led to a 99% drop in the price of antiretroviral therapy for HIV‐demand creation, community mobilization, treatment literacy, generic competition, economies of scale, and improved procurement of raw materials manufacturing processes are possible for HCV treatment.65,66 17
Figure 3: The First Generic DAAs Source: MSF Access Campaign, India. Used with permission. DAAs that simplify diagnostics and limit monitoring requirements can be mass‐produced affordably, and sustainably, according to Hill and colleagues, who compared chemical structure and synthesis complexity, molecular weight and dosing between antiretroviral agents and certain DAAs, and added margins for formulation and profit. DAAs were prioritized by safety and efficacy; these were ribavirin, sofosbuvir (nucleotide polymerase inhibitor), the NS5A inhibitors daclatasvir, ledipasvir and elbasvir (formerly MK‐8742; currently in phase 3) and the protease inhibitor grazoprevir (formerly MK‐5172; currently in phase 3). Regimens were priced at: US $118 (12 weeks of grazoprevir/elbasvir); US $121 (12 weeks of sofosbuvir/daclatasvir); US $129 to $193 (8 or 12 weeks of sofosbuvir/ledipasvir), US$ 149 to $298 (12 or 24 weeks of sofosbuvir and ribavirin). 67,68 As a next step, the researchers determined need for, type, and frequency of monitoring based on safety data from clinical trials (a complete blood count and other routine clinical chemistry tests, such as liver enzyme and creatinine levels, once before and once during treatment). They selected an antigen test (used for diagnosing HCV infection and determining treatment outcome), and included genotyping (in the absence of a pan genotypic regimen). The cost of diagnostics and monitoring was estimated based, on pricing in developing countries. Thus, a “package” of HCV treatment and diagnostics could be available without genotyping for US $174 to $354; with genotyping, from US $264 to $444.67,68 18
Figure 4. M
Minimum Cost for H
HCV Treattment, Diaagnostics, Genotypiing and Monitorin
ng SSource: van d
de Ven N, Fo
ortunak J, Sim
mmons B, et aal. Minimum
m target pricees for producction of direct‐
aacting antivirrals and asso
ociated diagn
nostics to com
mbat hepatittis C virus. Heepatology 20
015 A
Apr;61(4):11
174‐82. doi: 1
10.1002/hep.27641. Used
d with permission. 5
5.2 Implem
mentation .
Using a pub
blic‐health aapproach to HCV treatm
ment—one rregimen forr everyone‐‐‐will simpliffy procuremen
nt and delivvery of treattment, espeecially if durration does not vary by genotype, ssubtype or cirrhosis. So
ome regimeens do not require geno
otyping and subtyping, which are eexpensive aand not alwaays available. TThe compleexity and cost of HCV diiagnostics are often cited as a barrrier to treattment. SSimplifying diagnosticss by replacin
ng anti‐HCV and HCV RN
NA testing w
with a singlee antigen ttest; using aa pan genottypic DAA reegimen, and
d using anotther antigen
n test 12 or 24 weeks aafter treatm
ment compleetion will raadically simp
plify pretreaatment asseessment and
d minimize 68
ssafety and eefficacy monitoring. W
With DAAs, on‐treatmeent efficacy monitoringg is no longeer necessaryy to determine HCV ttreatment o
outcome. Sttopping rulees for treatm
ment failuree are no longger relevantt, since presence orr absence off detectablee HCV RNA aat week 4 does not preedict treatm
ment o
outcome; end‐of‐treatment HCV R
RNA may no
ot be reliable, since som
me people w
with detectable vviremia mayy ultimatelyy achieve SV
d
VR‐12. 69,70 ,771 19
6
6. State o
of the Field
d: HCV Drrug Develo
opment an
nd Researrch Gaps HCV
V drug development haas been done primarily by pharmacceutical com
mpanies, and driven byy com
mmercial inteerest. Companies havee developed
d incestuouss combinatio
ons instead of collaborrating to id
dentify optim
mal DAA reggimens with
h best‐in‐claass drugs. Th
his approach has delayed development of, aand access to promisingg regimens, and leaves many questions aboutt DAA safetyy and efficaccy in certaain populations (althou
ugh registriees will proviide addition
nal informattion on DAA
A use and outccomes outside of clinicaal trials). Som
me of the rem
maining queestions inclu
ude: need fo
or ribavirin in certain populations; treatment safety and efficacy in u
understudieed or exclud
ded populations, clinicaal relevance of drug ressistance, treatment sequ
uencing, pharmacogenomics, and genotypes 5
5 and 6. In HIV/AIDSS research, information
n from trials run by pharmaceutical companiess has been aaugmented—and optim
mized‐‐by reesults from cohort stud
dies, govern
nment‐fundeed research
h networks, p
public‐privatte partnersh
hips and invvestigator‐in
nitiated trials. These triials often ccombine drrugs from different com
mpanies, to aassess HIV ttreatment sstrategies su
uch as sstructured iinterruption
ns, simplification, or inttensification
n in differen
nt populatio
ons. Genericc d
drug produccers were th
he first to co
ombine anttiretroviral aagents from
m different pharmaceutical compaanies into fixxed‐dose co
ombinationss (FDCs) for resource‐lim
mited ssettings. Orriginator phaarmaceuticaal companiees eventuallly followed suit with Attripla, C
Complera/EEviplera, Evo
otaz and oth
hers. In contrast, HCV drug d
developmen
nt has been almost exclusively drivven by the pharmaceutical industrry. Companies have priioritized devvelopment o
of incestuou
us ccombinations rather th
han seeking to optimizee regimens tthrough collaboration. Commerciaal A regimens, delayed acccess to interests haave prevented explorattion of best‐‐in‐class DAA
promising rregimens, an
nd largely excluded peo
ople with co
ommon and
d serious com
morbidities o
or urgent need for treaatment from
m participation in registtration trialss. Thus, info
ormation on
n ttreatment ssafety, efficaacy, tolerab
bility and strrategy comees from regiistration triaals in ccarefully selected partiicipants. Altthough some real‐life d
data is becom
ming available from registries an
nd small post‐approval studies in h
high‐incomee countries, regimens aand strategies aare the sam
me as those used in clinical trials. 2
20
DAA mono therapy trials are short; for certain classes, regulatory agencies recommend limiting duration to three days, to forestall drug resistance.72 It is challenging to identify best‐in‐class drugs—or develop optimal regimens, since most regimens are comprised of drugs from one sponsor. As the market for DAAs becomes saturated pharmaceutical companies will move from HCV into new areas, abandoning trials in key populations, and development and assessment of new treatment strategies or exploration of the relevance of pharmacogenomics. 6.1 Role of Ribavirin in DAA Regimens The prevailing commercial strategies for DAA drug development are to shorten treatment, and remove ribavirin use. These may be at odds, since both longer treatment and ribavirin may be needed for people with the most advanced liver disease. Although ribavirin has significant limitations (teratogenicity, and side effects including anemia; insomnia; irritability; anxiety; depression; insomnia; nausea; and muscle and join pain), it is pan genotypic and it may bolster DAA treatment efficacy for certain regimens, genotypes and patient populations. In addition, it is no longer under patent protection and can be manufactured inexpensively. In the DAA era, the role of ribavirin remains unclear. Although RBV use has not increased SVR in several phase II trials, the number of participants has been too small to determine if, and which patients will benefit from it. Indeed, ribavirin remains a mainstay of many trials in “harder‐to‐treat” populations (compensated and more advanced‐stage cirrhosis, especially in genotype 3, and HIV/HCV). In particular, RBV may be needed to cure genotype 3, especially in people with cirrhosis. In the ALLY‐3 trial, 12 weeks of sofosbuvir and daclatasvir cured 96% of non‐cirrhotic participants‐‐‐but only 63% of cirrhotic participants.Error! Bookmark not defined. In the ELECTRON‐2 trial, adding RBV to 12 weeks of sofosbuvir/ledipasvir increased SVR from 64% to 100% among non‐cirrhotic, treatment‐naïve people with genotype 3; in treatment‐experienced cirrhotics, sofosbuvir/ledipasvir plus RBV cured 73%.73,74 6.2 Excluded and Under‐Represented Populations: Pregnancy, Nursing, Paediatrics, Comorbidities, People Who Use and Inject Drugs, the Elderly Despite the prevalence of HCV among women of childbearing age, there are not data on safety and efficacy of ribavirin‐free DAA regimens during pregnancy and nursing. Information on safety of HCV treatment during pregnancy and nursing is particularly important, given that drug‐drug interactions between DAAs and hormonal contraceptives may limit HCV treatment options or lead women to use less effective birth control during HCV treatment. 21
Paediatric trials (ages 3 to 17) of sofosbuvir and ribavirin and sofosbuvir/ledipasvir are underway. More research—especially of pan genotypic regimens—and optimal formulations are needed. People 65 and over are often excluded from DAA clinical trials. To date, safety, efficacy and tolerability of DAAs in elderly patients—albeit without common comorbidities—do not differ from younger people. More research is needed. Early stage or post‐transplant DAA clinical trials often limit enrollment to people with mild liver disease (METAVIR SCORE F0 to F2). DAAs are likely to be more effective in people with mild liver damage; exploration of shortened treatment duration in this population is needed. Although people with compensated cirrhosis are eligible for most DAA clinical trials, research in people with more advanced liver damage (Child Pugh stages B or C) is delayed until after approval. In the absence of compassionate use programmes (or those with limited eligibility), people with the most urgent need must wait until approval to gain access to potentially lifesaving DAAs in the absence of adequate safety and efficacy information. HCV treatment is a priority for people with hepatic or renal impairment, and people with other significant, common comorbidities (including cardiovascular disease, type 2 diabetes, HBV coinfection, COPD, and depression). Typically, small, often single‐dose pharmacokinetic (PK) studies are performed in people with renal or hepatic impairment before DAAs are approved, but actual trials do not occur until after approval. Often, people with other comorbidities are not eligible for clinical trials—and information on drug safety and efficacy in people with other illnesses does not emerge until drugs have been on the market for years. Failure to include people who inject drugs, the highest‐prevalence population, in clinical trials creates a vicious cycle, where treatment is withheld due to lack of evidence. After years of pressure from people who use and inject drugs and their advocates, people on opioid substitution treatment have not been completely excluded from clinical trials; a few HCV treatment trials are being conducted in active drug users. 6.3 DAAs and Pharmacogenomics Researchers finally identified part of the reason for poorer response to interferon among African Americans: the IL‐28B gene; this was only possible because of adequate representation in clinical trials. Some DAA regimens appear to be less effective in people with the IL28B TT genotype (most common among African Americans), especially when duration of treatment is shortened.75 With ombitasvir/paritaprevir/ritonavir and dasabuvir, ribavirin increased SVR in African Americans who had genotype 1a.76 Simeprevir dose may need to be adjusted in people of East Asian ancestry.Error! Bookmark not defined. 22
Other, unidentified pharmacogenomics factors may lessen or increase DAA treatment efficacy. 6.4 HCV Genotypes 5 and 6 DAAs are often characterized as pan genotypic, using data from in vitro studies. Safety and efficacy of HCV treatment across genotypes—and in people who are infected with multiple genotype (which occurs among people who inject drugs, dialysis recipients and others with multiple exposures to HCV) must be characterized in clinical trials.23,24 Data in genotypes 5 and 6 are limited. Although over a million people are infected with HCV genotype 5, only a handful them have been included in clinical trials of sofosbuvir‐based regimens.21,77,78,79 Although DAAs have demonstrated efficacy in a small group of people with G5 or G6, more data are needed to inform regimen selection and global treatment scale‐up. 6.5 HCV Drug Resistance and Treatment Sequencing Mutations that confer resistance to one or more DAA classes may be present at baseline or after treatment failure. The prevalence of baseline resistance‐associated variants (RAVs) varies by HCV genotype and subtype.80 For example, baseline RAVs associated with certain HCV protease inhibitors and non‐nucleoside polymerase inhibitors have been observed in genotype 3a.81 In genotype 1, baseline NS5A RAVS have been found in 6% to 12%, although these do not always preclude successful treatment.80 In clinical trials of sofosbuvir/ledipasvir, prevalence of baseline NS5A RAVs ranged from 14% to 18%; although 90% of people with these baseline RAVs achieved SVR, post‐treatment NS5A RAVs were found in most people who were not cured.33,82 Sofosbuvir has a high resistance barrier. Although the S282T mutation has been selected in all genotypes during in vitro studies, the clinical significance of this RAV is unclear; it has been found in in 1 of 1545 trial participants, who was not cured.83 Other mutations have been associated with reduced efficacy and sofosbuvir treatment failure. The L159F mutation has been found at baseline and post‐treatment in genotypes 1b and 3a; the V321A mutation has been detected after treatment failure in genotype 3a; C316N, and S282R may also reduce efficacy of sofosbuvir in genotype 1a.84,85 The selection of first‐line treatment will set the stage for a second‐line regimen. Until the longevity and significance of DAA drug resistance is well understood, avoiding retreatment with DAAs from the same class—unless they have demonstrated sufficient activity against drug‐resistant virus—is the preferred strategy. More information is needed on the relationship between baseline or post‐treatment RAVs and HCV treatment—or retreatment outcomes. Yet successful re‐treatment with sofosbuvir may be possible, even for people who were not cured by a sofosbuvir‐containing regimen. Lengthening treatment duration, adding ribavirin or use of additional DAAs may do the trick.43,86 In ELECTRON‐2, 100% of 19 sofosbuvir‐
experienced participants were cured by retreatment with 12 weeks of sofosbuvir/ledipasvir 23
aand ribavirin.73,74 Addittional real‐w
world experrience is neeeded, to sup
pport recycling ssofosbuvir. For second‐‐line regimeens, the pan
n genotypic HCV proteaase inhibitorrs in late‐staage d
development may provve essential for those w
who are not cured by an
n NS5A/nuccleotide ccombination. Research
h of pan gen
notypic‐baseed PI regimeens must incclude peoplle who weree unsuccessfu
ully treated with a nucleotide/NS5
5A regimen.
6
6. 6 Researcch Coordinaation and O
Opportunitiees SScaling up gglobal HCV ttreatment aaccess is chaallenging, bu
ut offers thee opportunity to leverage the lessons learned from
m HIV, wheree research h
has been invvaluable. W
With HIV, prevention,, treatment and testingg programm
mes were esttablished yeears before effective ttreatment w
was availablle. With HCV
V, we have the cure—b
but lack an iindependen
nt research aagenda for DAAs and their implem
mentation. A
A coordinateed plan to p
provide caree and ttreatment w
while seekin
ng to optimiize it‐‐ and aassess the im
mpact of do
oing so will m
make the most of resources. DAAs offer an unprecedented opp
portunity: to
o nest a clinical researcch agenda w
within HCV prevention,, testing, care and treattment programmes, an
nd monitor o
outcomes. FFor examplee, sscreening in
nitiatives co
ould be used
d to validatee diagnosticc tools and p
provide survveillance d
data—which can be used to forecaast treatmeent need, po
ool procurem
ment, and aallocate aadequate reesources, an
nd inform th
he design an
nd reach of prevention
n programm
mes. TTreatment programs caan embed q
questions on
n DAA safety and efficaacy, explore ideal regimens an
nd treatmen
nt duration through facctorial trialss, collect datta on treatm
ment o
outcomes aand reinfection rates—aand assess d
different mo
odels of carre. Data from
m these programmees can be ussed to devellop models to further in
nform resou
urce needs aand programmaatic planningg. 7
7. The HCV
V Treatmeent Pipelin
ne B
By 2016, two
o fixed‐dosee combinatiions are likeely to be app
proved. Botth appear prromising, bu
ut data are currently llimited to reesults from small phasee 2 trials. Some DAA
As in the neext batch wiill be pan genotypic, w
with activityy against som
me of the m
mutations known to ccause drug rresistance. Sponsors are hoping to
o avoid ribaavirin, and shorten treatment by combining DAAs from 3 classes. By 2016, there will be aadditional H
HCV treatmeent options. Gilead and
d Merck havve two‐DAA
A FDCs in phaase 3, and both compan
nies are exp
ploring triplee DAA treatment. 2
24
7.1 Sofosbuvir‐based FDCs Gilead’s sofosbuvir/velpatasvir (NS5A inhibitor; formerly GS‐5816) is a once‐daily FDC in phase 3. Phase 2 results are promising, but more data are needed, especially in genotype 3 and cirrhosis, and genotypes 4, 5 and 6. In phase 2, after 12 weeks of treatment, SVR in genotype 1 ranged from 96% to 100%, regardless of ribavirin use, treatment history and cirrhosis; shortening treatment to 8 weeks—albeit in non‐cirrhotic, treatment naïve study participants—did not appear to be a viable strategy for this regimen, even with ribavirin. In genotype 3, SVR in treatment‐experienced, cirrhotic study participants was 88%; adding RBV to the mix increased SVR to 96%.87 The sofosbuvir/velpatasvir FDC is currently being studied in people who are treatment‐
naïve or treatment experienced (PEG‐IFN and ribavirin, with or without an HCV protease inhibitor) , in HCV genotypes 1,2,4,5 and 6. The FDC is also being compared to 12 weeks (in G2) or 24 weeks (in G3) of sofosbuvir + RBV, and being studied in advanced liver disease: with or without ribavirin, in Child‐Pugh Class B for 12 weeks, and for 24 weeks, without ribavirin, in Child‐Pugh class C. A phase 2 trial is assessing sofosbuvir, GS‐5816 and GS‐9857, a pan genotypic protease inhibitor in genotypes 1, 2, 3, 4, 5 and 6, treatment‐naïve or –experienced people, with or without cirrhosis, for 6, 8 or 12 weeks. Table 5. Sofosbuvir + GS‐5816* With or Without RBV in Phase 279,87 Genotype Duration Treatment‐ Cirrhosis
experienced
SVR 1 8 weeks NO NO 12 weeks NO NO 12 weeks YES YES: Some participants were cirrhotic; data not broken out by cirrhosis status 8 weeks NO NO 12 weeks NO NO 81% + RBV (25/31) 90% no RBV (26/29) 100% no RBV (28/28) 100% no RBV (28/28) 96% + RBV (27/28) relapse occurred in a cirrhotic participant 100% no RBV (27/27) 88% no RBV (23/26) 88% + RBV (23/26) 100% no RBV (10/10) 2 25
3 12 weeks NO NO 93% no RBV (23/27) YES NO YES YES 4 12 weeks NO NO 5 (25 mg GS‐
5816) 6 12 weeks NO NO 100% ± RBV (53/53) 88% no RBV (23/26) 96% + RBV (25/26) 86% no RBV (6/7) 100% no RBV (1/1) 12 weeks NO NO 100% no RBV (5/5) 7.2 Grazoprevir/Elbasvir‐Based Regimens Merck’s grazoprevir/elbasvir FDC (formerly MK‐5172, an HCV protease inhibitor, and MK‐
8742, an NS5A inhibitor) has entered phase 3. In the phase 2 C‐WORTHY trial of grazoprevir/elbasvir, with or without ribavirin, cure rates in genotype 1 ranged from 93% (with RBV) to 98% (no RBV) in HCV monoinfection, and 87% (no RBV) to 97% (with RBV) in HIV/HCV.88 Grazoprevir and elbasvir also performed well in another genotype 1 trial, in treatment naïve people with cirrhosis, and treatment ‐experienced people with or without cirrhosis. An 8‐
arm trial compared 12 or 18 weeks of treatment, with or without RBV, in treatment‐naïve people with cirrhosis, and treatment‐experienced people (with and without cirrhosis). There were no significant differences in SVR by ribavirin use, subtype, cirrhosis, treatment duration or experience.89 Table 6. SVR among Cirrhotic Participants in C‐WORTHY89 Population SVR, 12 weeks, + RBV SVR, 12 weeks, no RBV SVR, 18 weeks, + RBV SVR, 18 weeks, no RBV 94% (29/31) Treatment‐naïve, 90% (28/31) (97%) 28/29 97% (31/32) cirrhosis Treatment‐
92% (23/25) 100% (22/23)
experienced, no significant difference for RBV no significant difference for RBV cirrhosis vs. no RBV vs. no RBV Merck’s FDC may become the backbone of an abbreviated, pan genotypic regimen. It is currently being studied in HCV genotypes 1, 2, 4, and 6, and in C‐SWIFT, a phase 2 trial of the FDC and sofosbuvir, in genotypes 1 and 3. Interim data from C‐SWIFT are available in genotype 1. Although the 4‐week regimen yielded an SVR‐4 of only 38% in non‐cirrhotic 26
study participants, extending treatment to 6 weeks increased SVR‐4 to 86% in non‐cirrhotic participants (vs. 80% in cirrhotics); and 8 weeks of treatment cured 94% of cirrhotic participants.90 Merck is developing MK‐3682, a nucleotide polymerase inhibitor and MK‐8408, a pan genotypic NS5A inhibitor with activity against common NS5A RAVs.91 With these DAAs, the company has the potential to construct pan genotypic, once daily regimens. They are studying MK‐3682 with the grazoprevir/elbasvir FDC, and comparing efficacy of MK‐8404 versus elbasvir with grazoprevir and MK‐3682 in phase 2 trials. 8. Conclusion Perfectovir should not become the enemy of Goodovir. ‐Jennifer Cohn, Medical Director, MSF Access Campaign Other promising DAA candidates are in early‐phase development. With such high cure rates, improvements in DAA regimens are likely to be incremental and do not warrant delaying access until “perfectovir” appears—instead, the focus should be directed towards working with affected communities, scaling up and linking prevention and treatment programmes, building capacity among non‐specialist providers to deliver these simplified regimens,92 The real challenge is no longer curing hepatitis C—it is getting treatment to the millions of people who need it, as soon as possible. References 1
Wang CC, Krantz E, Klarquist J, et al. Acute hepatitis C in a contemporary US cohort: modes of acquisition and factors influencing viral clearance. J Infect Dis. 2007 Nov 15;196(10):1474‐82. Skip to main content 2
Adinolfi LE, Nevola R, Lus G, et al. Chronic hepatitis C virus infection and neurological and psychiatric disorders: An overview. World J Gastroenterol. 2015 Feb 28;21(8):2269‐2280. 3
Bladowska J, Zimny A, Knysz B, et al. Evaluation of early cerebral metabolic, perfusion and microstructural changes in HCV‐positive patients: a pilot study. J Hepatol. 2013 Oct;59(4):651‐7. doi: 10.1016/j.jhep.2013.05.008. 4
Cacoub P, Gragnani L, Comarmond C, Zignego AL. Extrahepatic manifestations of chronic hepatitis C virus infection. Dig Liver Dis. 2014 Dec 15;46 Suppl 5:S165‐73. doi: 10.1016/j.dld.2014.10.005. 5
Thames AD, Castellon SA, Singer EJ, et al. Neuroimaging abnormalities, neurocognitive function, and fatigue in patients with hepatitis C. Neurol Neuroimmunol Neuroinflamm. 2015 Jan 14;2(1):e59. doi: 10.1212/NXI.0000000000000059. 27
6
Torres HA, Mahale P. Most patients with HCV‐associated lymphoma present with mild liver disease: A call to revise antiviral treatment prioritization. Liver Int. 2015 Mar 16. doi: 10.1111/liv.12825. 7
Zignego AL, Gragnani L, Giannini C, Laffi G. The hepatitis C virus infection as a systemic disease. Intern Emerg Med. 2012 Oct;7 Suppl 3:S201‐8. doi: 10.1007/s11739‐012‐0825‐6. 8
Ly KN, Xing J, Klevens RM, Jiles RB, Holmberg SD. Causes of death and characteristics of decedents with viral hepatitis, United States, 2010. Clin Infect Dis. 2014 Jan;58(1):40‐9. doi: 10.1093/cid/cit642. 9
European Paediatric Hepatitis C Virus Network. Three broad modalities in the natural history of vertically acquired hepatitis C virus infection. Clin Infect Dis. 2005 Jul 1;41(1):45‐51. 10
Resti M, Jara P, Hierro L, et al. Clinical features and progression of perinatally acquired hepatitis C virus infection. J Med Virol. 2003 Jul;70(3):373‐7. 11
Abdel‐Hady M, Bunn SK, Sira J, et al. Chronic hepatitis C in children‐‐review of natural history at a National Centre. J Viral Hepat. 2011 Oct;18(10):e535‐40. doi: 10.1111/j.1365‐2893.2011.01456.x. 12
Bortolotti F, Verucchi G, Cammà C, et al; Italian Observatory for HCV Infection and Hepatitis C in Children. Long‐term course of chronic hepatitis C in children: from viral clearance to end‐stage liver disease. Gastroenterology. 2008 Jun;134(7):1900‐7. doi: 10.1053/j. gastro.2008.02.082. 13
Rerksuppaphol S, Hardikar W, Dore GJ. Long‐term outcome of vertically acquired and post‐
transfusion hepatitis C infection in children. J Gastroenterol Hepatol. 2004 Dec;19(12):1357‐62. 14
Thein HH, Yi Q, Dore GJ, Krahn MD. Estimation of stage‐specific fibrosis progression rates in chronic hepatitis C virus infection: a meta‐analysis and meta‐regression. Hepatology. 2008 Aug;48(2):418‐31. doi: 10.1002/hep.22375. 15
Global Burden of Disease 2013 Mortality and Causes of Death Collaborators. Global, regional, and national age–sex specific all‐cause and cause‐specific mortality for 240 causes of death, 1990–2013: a systematic analysis for the Global Burden of Disease Study 2013. The Lancet, Vol. 385, No. 9963, p117–171. 16
World Health Organization. Media Centre. The top 10 causes of death in lower‐ middle‐income countries 2012. http://www.who.int/mediacentre/factsheets/fs310/en/index1.html 17
World Health Organization. Media Centre. The top 10 causes of death in upper‐middle‐income countries 2012. http://www.who.int/mediacentre/factsheets/fs310/en/index1.html 18
García‐Fulgueiras A, García‐Pina R, Morant C, García‐Ortuzar V, Génova R, Alvarez E. Hepatitis C and hepatitis B‐related mortality in Spain. Eur J Gastroenterol Hepatol. 2009 Aug;21(8):895‐901. doi: 10.1097/MEG.0b013e328313139d 19
Ly KN, Xing J, Klevens RM, Jiles RB, Ward JW, Holmberg SD. The increasing burden of mortality 28
from viral hepatitis in the United States between 1999 and 2007. Ann Intern Med. 2012 Feb 21;156(4):271‐8. doi: 10.7326/0003‐4819‐156‐4‐201202210‐00004. 20
Palmateer NE, Hutchinson SJ, McLeod A, Codere G, Goldberg DJ. Comparison of deaths related to Hepatitis C and AIDS in Scotland. J Viral Hepat. 2007 Dec;14(12):870‐4. 21
Messina JP, Humphreys I, Flaxman A, et al. Global distribution and prevalence of hepatitis C virus genotypes. Hepatology. 2015 Jan;61(1):77‐87. doi: 10.1002/hep.27259. 22
Smith DB, Bukh J, Kuiken C, et al. Expanded classification of hepatitis C virus into 7 genotypes and 67 subtypes: updated criteria and genotype assignment web resource. Hepatology. 2014 Jan;59(1):318‐27. doi: 10.1002/hep.26744. 23
Cunningham EB, Applegate TL, Lloyd AR, Dore GJ, Grebely J. Mixed HCV infection and reinfection in people who inject drugs‐impact on therapy. Nat Rev Gastroenterol Hepatol. 2015 Mar 17. doi: 10.1038/nrgastro.2015.36. 24
Qian KP, Natov SN, Pereira BJ, Lau JY. Hepatitis C virus mixed genotype infection in patients on haemodialysis. J Viral Hepat. 2000 Mar;7(2):153‐60. 25
Morisco F, Granata R, Stroffolini T, et al. Sustained virological response: a milestone in the treatment of chronic hepatitis C. World J Gastroenterol. 2013 May 14;19(18):2793‐8. doi: 10.3748/wjg.v19.i18.2793. 26
Berenguer J, Rodríguez E, Miralles P, et al; GESIDA HIV/HCV Cohort Study Group Sustained virological response to interferon plus ribavirin reduces non‐liver‐related mortality in patients coinfected with HIV and Hepatitis C virus. Clin Infect Dis. 2012 Sep;55(5):728‐36. 27
Innes HA, McDonald SA, Dillon JF, et al. Towards a more complete understanding of the association between a hepatitis C sustained viral response and cause‐specific outcomes. Hepatology. 2015 Feb 26. doi: 10.1002/hep.27766 28
Mira JA, Rivero‐Juárez A, López‐Cortés LF, et al; Grupo Andaluz para el Estudio de las Hepatitis Víricas de la Sociedad Andaluza de Enfermedades Infecciosas. Benefits from sustained virologic response to pegylated interferon plus ribavirin in HIV/hepatitis C virus‐coinfected patients with compensated cirrhosis. Clin Infect Dis. 2013 Jun;56(11):1646‐53. doi: 10.1093/cid/cit103. 29
Morgan RL, Baack B, Smith BD, Yartel A, Pitasi M, Falck‐Ytter Y. Eradication of hepatitis C virus infection and the development of hepatocellular carcinoma: a meta‐analysis of observational studies. Ann Intern Med. 2013 Mar 5;158(5 Pt 1):329‐37. doi: 10.7326/0003‐4819‐158‐5‐201303050‐
00005. 30
Morgan TR, Ghany MG, Kim HY et al; HALT‐C Trial Group. Outcome of sustained virological responders with histologically advanced chronic hepatitis C. Hepatology. 2010 Sep;52(3):833‐44. doi: 10.1002/hep.23744. 29
31
van der Meer AJ, Veldt BJ, Feld JJ, et al. Association between sustained virological response and all‐cause mortality among patients with chronic hepatitis C and advanced hepatic fibrosis. JAMA. 2012 Dec 26;308(24):2584‐93. doi: 10.1001/jama.2012.144878. 32
Lok AS, Gardiner DF, Lawitz E, et al. Preliminary study of two antiviral agents for hepatitis C genotype 1. N Engl J Med. 2012 Jan 19;366(3):216‐24. doi: 10.1056/NEJMoa1104430 33
Kowdley KV, Gordon SC, Reddy KR, et al; ION‐3 Investigators. Ledipasvir and sofosbuvir for 8 or 12 weeks for chronic HCV without cirrhosis. N Engl J Med. 2014 May 15;370(20):1879‐88. doi: 10.1056/NEJMoa1402355 34
Lawitz E, Sulkowski MS, Ghalib R, et al. Simeprevir plus sofosbuvir, with or without ribavirin, to treat chronic infection with hepatitis C virus genotype 1 in non‐responders to pegylated interferon and ribavirin and treatment‐naive patients: the COSMOS randomised study. Lancet. 2014 Nov 15;384(9956):1756‐65. doi: 10.1016/S0140‐6736(14)61036‐9. 35
Poordad F, Hezode C, Trinh R, et al. ABT‐450/r‐ombitasvir and dasabuvir with ribavirin for hepatitis C with cirrhosis. N Engl J Med. 2014 May 22;370(21):1973‐82. doi: 10.1056/NEJMoa1402869. 36
Sulkowski MS, Gardiner DF, Rodriguez‐Torres M, et al; AI444040 Study Group. Daclatasvir plus sofosbuvir for previously treated or untreated chronic HCV infection. N Engl J Med. 2014 Jan 16;370(3):211‐21. doi: 10.1056/NEJMoa1306218. 37
Zeuzem S, Dusheiko GM, Salupere R, et al; VALENCE Investigators. Sofosbuvir and ribavirin in HCV genotypes 2 and 3. N Engl J Med. 2014 May 22;370(21):1993‐2001. doi: 10.1056/NEJMoa1316145. 38
Hinrichsen H, Benhamou Y, Wedemeyer H, et al. Short‐term antiviral efficacy of BILN 2061, a hepatitis C virus serine protease inhibitor, in hepatitis C genotype 1 patients. Gastroenterology. 2004 Nov;127(5):1347‐55. 39
Everson GT, Sims KD, Rodriguez‐Torres M, et al. Efficacy of an interferon‐ and ribavirin‐free regimen of daclatasvir, asunaprevir, and BMS‐791325 in treatment‐naive patients with HCV genotype 1 infection. Gastroenterology. 2014 Feb;146(2):420‐9. doi: 10.1053/j.gastro.2013.10.057. 40
Kowdley KV, Lawitz E, Poordad F, et al. Phase 2b trial of interferon‐free therapy for hepatitis C virus genotype 1. N Engl J Med. 2014 Jan 16;370(3):222‐32. doi: 10.1056/NEJMoa1306227. 41
Doehle B, Gontcharova V, Chodavarapu et al. Resistance analysis of treatment‐naïve HCV genotype 1‐6 infected patients treated wit sofosbuvir in combination with GS‐5816 for 12 weeks (Abstract 1942). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7 – 11, 2014. Boston, MA. 42
Krishnan P, Tripathi R, Schnell G, et al. Pooled analysis of resistance in patients treated with ombitasvir/ABT‐450/r and dasabuvir with or without ribavirin in phase 2 and phase 3 clinical trials (Abstract 1936). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7 – 11, 2014. Boston, MA. 30
43
Lawitz E, Poordad FF, Pang PS et al. Sofosbuvir and ledipasvir fixed‐dose combination with and without ribavirin in treatment‐naïve and previously treated patients with genotype 1 hepatitis C: the LONESTAR study (Abstract 215). 64th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 1‐5, 2013. Washington DC. 44
Sarrazin C, Dvory‐Sobol H, Svarovskaia E, et al. Baseline and post‐baseline resistance analyses of phase 2/3 studies of ledipasvir/sofosbuvir ± RBV (Abstract 1926). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7‐ 11, 2014. Boston, MA. 45
Schneider MD, Sarrazin C.Antiviral therapy of hepatitis C in 2014: do we need resistance testing? Antiviral Res. 2014 May;105:64‐71. doi: 10.1016/j.antiviral.2014.02.011. 46
Helfand C. Hello, hep C pricing war Gilead hits back at AbbVie with exclusive CVS deal. Fierce Pharma. January 5, 2015. http://www.fiercepharma.com/story/gilead‐hits‐back‐abbvie‐exclusive‐
cvs‐hep‐c‐deal/2015‐01‐05 47
Hirst EJ. AbbVie lowers price of hepatitis C drugs to win deal with Express Scripts. Chicago Tribune. December 22, 2014. http://www.chicagotribune.com/business/breaking/ct‐abbvie‐hepatitis‐c‐drug‐
1223‐biz‐20141222‐story.html 48
Jerzyk E. R.I. Medicaid limits supply of hepatitis C drug due to cost. The Brown Daily. December 4, 2014. http://www.browndailyherald.com/2014/12/04/r‐medicaid‐limits‐supply‐hepatitis‐c‐drug‐
due‐cost/ 49
Venteicher W. Most Illinois Medicaid Patients Denied New Hepatitis C Drugs. Chicago Tribune. November 19th, 2014. http://kaiserhealthnews.org/news/most‐illinois‐medicaid‐patients‐denied‐
new‐hepatitis‐c‐drugs/ 50
Huet N. France uses tax to put pressure on hepatitis C drug prices. Reuters. September 30, 2014. http://www.reuters.com/article/2014/09/30/france‐deficit‐gilead‐idUSL6N0RV27X20140930. 51
Sevillano EG. Spanish hepatitis C patients to march for access to expensive new drugs. January 6, 2015. El Pais. http://elpais.com/elpais/2015/01/06/inenglish/1420544396_017257.html 52
Whalen, J. Patients blame cost for coverage delay in England of Gilead Drug. January 15, 2015. Wall Street Journal Pharmalot. http://blogs.wsj.com/pharmalot/2015/01/22/coverage‐for‐gileads‐
sovaldi‐is‐delayed‐in‐england‐over‐cost/ 53
Dore GJ, Ward J, Thursz M. Hepatitis C disease burden and strategies to manage the burden. J Viral Hepat 2014; 21(Suppl 1):1‐4. 54
Dore GJ, Feld JJ. Hepatitis C virus therapeutic development: in pursuit of perfectovir. Clin Infect Dis. 2015 Mar 11. pii: civ197. 55
Hatzakis A, Chulanov V, Gadano AC, et al, The present and future disease burden of hepatitis C virus (HCV) infections with today's treatment paradigm – volume 2. J Viral Hepat. 2015 Jan;22 Suppl 31
1:26‐45. doi: 10.1111/jvh.12351. 56
Gane E, Kershenobich, D Seguin‐Devaux C, et al. Strategies to manage hepatitis C virus (HCV) infection disease burden – volume 2. J Viral Hepat. 2015 Jan;22 Suppl 1:46‐73. doi: 10.1111/jvh.12352. 57
American Association for the Study of Liver Diseases/ Infectious Diseases Society of America (AASLD/IDSA) Recommendations for Testing, Managing and Treating Hepatitis C. March 2015. http://www.hcvguidelines.org 58
European Association for the Study of the Liver (EASL). EASL Recommendations on Treatment of Hepatitis C 2014. April 2014. http://www.easl.eu/assets/application/files/easl_recommendations_hcv_2014_full.pdf 59
World Health Organization. Guidelines for the Screening, Care and Treatment of Persons with Hepatitis C Infection. April 2014. http://apps.who.int/iris/bitstream/10665/111747/1/9789241548755_eng.pdf?ua=1 60
Obach D, Yazdanpanah Y, Esmat G, et al. How to optimize hepatitis C virus treatment impact on life years saved in resource‐constrained countries. Hepatology. 2015 Jan 10. doi: 10.1002/hep.27691. 61
Mohamoud YA, Mumtaz GR, Riome S, Miller D, Abu‐Raddad LJ. The epidemiology of hepatitis C virus in Egypt: a systematic review and data synthesis. BMC Infect Dis. 2013 Jun 24;13:288. doi: 10.1186/1471‐2334‐13‐288. 62
Martin NK, Hickman M, Hutchinson SJ, Goldberg DJ, Vickerman P.Combination interventions to prevent HCV transmission among people who inject drugs: modeling the impact of antiviral treatment, needle and syringe programs, and opiate substitution therapy. Clin Infect Dis. 2013 Aug;57 Suppl 2:S39‐45. doi: 10.1093/cid/cit296. 63
Nelson PK, Mathers BM, Cowie B, et al. Global epidemiology of hepatitis B and hepatitis C in people who inject drugs: results of systematic reviews. Lancet. 2011 Aug 13;378(9791):571‐83. doi: 10.1016/S0140‐6736(11)61097‐0. 64
Martin NK, Vickerman P, Grebely J, et al. Hepatitis C virus treatment for prevention among people who inject drugs: Modeling treatment scale‐up in the age of direct‐acting antivirals. Hepatology. 2013 Nov;58(5):1598‐609. doi: 10.1002/hep.26431. 65
Hill A, Cooke G.. Medicine. Hepatitis C can be cured globally, but at what cost? Science. 2014 Jul 11;345(6193):141‐2. doi: 10.1126/science.1257737. 66
MSF. Untangling the web of antiretroviral price reductions. 17th Edition. July 2014. http://www.msfaccess.org/sites/default/files/MSF_UTW_17th_Edition_4_b.pdf 67
Hill A, Khoo S, Fortunak J, Simmons B, Ford N Minimum costs for producing hepatitis C direct‐
acting antivirals for use in large‐scale treatment access programs in developing countries. Clin Infect 32
Dis. 2014 Apr;58(7):928‐36. doi: 10.1093/cid/ciu0 68
van de Ven N, Fortunak J, Simmons B, et al. Minimum target prices for production of direct‐acting antivirals and associated diagnostics to combat hepatitis C virus. Hepatology. 2014 Dec 6. doi: 10.1002/hep.27641. 69
Nguyen T, Guedj J, Canini L, et al. The paradox of highly effective sofosbuvir combo therapy despite slow viral decline (Abstract 148). 2015 Conference on Retroviruses and Opportunistic Infections (CROI) February 23‐26th, Seattle, WA. 70
Sidharthani S, Kohli A, Osinusi A, et al. Utility of hepatitis C viral load monitoring with ledipasvir and sofosbuvir therapy. (Abstract 689) 2015 Conference on Retroviruses and Opportunistic Infections (CROI) February 23‐26th, Seattle, WA. 71
Wyles D, Eron JJ, Trinh, R, et al. High SVR regardless of time to suppression with ombitasvir/paritaprevir/r and dasabuvir + RBV (Abstract 147). 2015 Conference on Retroviruses and Opportunistic Infections (CROI) February 23‐26th, Seattle, WA. 72
U.S. Department of Health and Human Services Food and Drug Administration Center for Drug Evaluation and Research (CDER). Guidance for Industry. Chronic hepatitis C infection: developing direct‐acting antiviral drugs for treatment. Draft Guidance. Revision 1. October 2013. http://www.fda.gov/downloads/Drugs/GuidanceComplianceRegulatoryInformation/Guidances/UC
M225333.pdf 73
Gane EJ, Hyland RH, An D, et al. Ledipasvir/sofodbuvir fixed‐dose combination is safe and effective in difficult‐to‐treat populations including GT 3 patients, decompensated GT 1 patients, and GT1 patients with prior sofosbuvir experience (Abstract 06). 49th Annual Meeting of the European Association for the Study of the Liver (EASL), April 9‐13, 2014, London UK. 74
Gane EJ, Hyland RH, An D, et al. High efficacy of LDV/SOF regimens for 12 weeks for patients with HCV genotype 3 or 6 infection (Abstract LB‐11). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD), November 7‐11, 2014, Boston, MA. 75
Jacobson IM, Kwo PY, Kowdley KV, et al. Virologic response rates to all‐oral fixed‐dose combination ledipasvir/sofosbuvir regimens are similar in patients with and without traditional negative predictive factors in phase 3 clinical trials (Abstract 1945). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). Nov 7 ‐ 11, 2014. Boston, MA 76
Vierling J, Puoti M, Bernstein D, et al. Efficacy by race or geographic region in HCV genotype 1‐
infected patients treated with ABT‐450/ritonavir/ombitasvir and dasabuvir, with or without ribavirin (Abstract 1968). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7 – 11, 2014. Boston, MA. 77
Everson G, Tran TT, Towner WJ, et al. Safety and efficacy of treatment with the interferon‐free, ribavirin‐free combination of sofosbuvir + GS‐5816 for 12 weeks in treatment naive patients with 33
genotype 1‐6 HCV infection. (Abstract 011) 49th Annual Meeting of the European Association for the Study of the Liver (EASL), April 9‐13, 2014, London UK. 78
Lawitz E, Mangia A, Wyles D, et al. Sofosbuvir for previously untreated chronic hepatitis C infection. N Engl J Med. 2013 May 16;368(20):1878‐87. doi: 10.1056/NEJMoa1214853. 79
Tran TT, Morgan TR, Thulyvath PJ, et al. Safety and efficacy of treatment with sofosbuvir + GS‐
5816 ± ribavirin for 8 or 12 weeks in treatment‐naïve patients with genotype 1‐6 infection (Abstract 80). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7‐ 11, 2014. Boston, MA. 80
Poveda E, Wyles DL, Mena A, Pedreira JD, Castro‐Iglesias A, Cachay E. Update on hepatitis C virus resistance to direct‐acting antiviral agents. Antiviral Res. 2014 Aug;108:181‐91. doi: 10.1016/j.antiviral.2014.05.015. 81
Alves R, Queiroz AT, Pessoa MG, et al. The presence of resistance mutations to protease and polymerase inhibitors in Hepatitis C virus sequences from the Los Alamos databank. J Viral Hepat. 2013 Jun;20(6):414‐21. doi: 10.1111/jvh.12051. 82
Afdhal N, Zeuzem S, Kwo P, et al; ION‐1 Investigators. Ledipasvir and sofosbuvir for untreated HCV genotype 1 infection N Engl J Med. 2014 May 15;370(20):1889‐98. doi: 10.1056/NEJMoa1402454. 83
Svarovskaia ES, Dvory‐Sobol H, Parkin N, et al. Infrequent development of resistance in genotype 1‐6 hepatitis C virus‐infected subjects treated with sofosbuvir in phase 2 and 3 clinical trials. Clin Infect Dis. 2014 Dec 15;59(12):1666‐74. doi: 10.1093/cid/ciu697. 84
Curry MP, Forns X, Chung RT, et al. Sofosbuvir and ribavirin prevent recurrence of HCV infection after liver transplantation: an open‐label study. Gastroenterology. 2015 Jan;148(1):100‐107.e1. doi: 10.1053/j.gastro.2014.09.023 85
Donaldson EF, Harrington PR, O'Rear JJ, Naeger LK. Clinical evidence and bioinformatics characterization of potential hepatitis C virus resistance pathways for sofosbuvir. Hepatology. 2015 Jan;61(1):56‐65. doi: 10.1002/hep.27375 86
Wyles D, Pockros P, Zhu Y, et al. Retreatment of patients who failed prior sofosbuvir‐based regimens with all oral fixed‐dose combination ledipasvir/sofosbuvir plus ribavirin for 12 weeks (Abstract 235). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7 – 11, 2014. Boston, MA. 87
Pianko S, Flamm SL, Shiffman ML, et al. High efficacy of treatment with sofosbuvir+GS‐5816 ± ribavirin for 12 weeks in treatment experienced patients with genotype 1 or 3 HCV infection (Abstract 197). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7 – 11, 2014. Boston, MA. 34
88
Sulkowski M, Hezode C, Gerstoft J, et al. Efficacy and safety of 8 weeks versus 12 weeks of treatment with grazoprevir (MK‐5172) and elbasvir (MK‐8742) with or without ribavirin in patients with hepatitis C virus genotype 1 mono‐infection and HIV/hepatitis C virus co‐infection (C‐WORTHY): a randomised, open‐label phase 2 trial. Lancet. 2015 Mar 21;385(9973):1087‐97. doi: 10.1016/S0140‐6736(14)61793‐1. 89
Lawitz E, Gane E, Pearlman B, et al. Efficacy and safety of 12 weeks versus 18 weeks of treatment with grazoprevir (MK‐5172) and elbasvir (MK‐8742) with or without ribavirin for hepatitis C virus genotype 1 infection in previously untreated patients with cirrhosis and patients with previous null response with or without cirrhosis (C‐WORTHY): a randomised, open‐label phase 2 trial. Lancet. 2015 Mar 21;385(9973):1075‐86. doi: 10.1016/S0140‐6736(14)61795‐5. 90
Lawitz E, Poordad F, Gutierrez JA, et al. C‐SWIFT: grazoprevir (MK‐5127) + elbasvir (MK‐8742) + sofosbuvir in treatment‐naïve patients with hepatitis C virus genotype 1 infection, with and without cirrhosis, for durations of 4, 6, or 8 weeks (interim results) (Abstract 2014). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7 – 11, 2014. Boston, MA. 91
Asante‐Appiah E, Liu R, Curry S, et al. MK‐8408, a potent and selective NS5A inhibitor with a high genetic barrier to resistance and activity against HCV genotypes 1‐6 (Abstract 1979). 65th Annual Meeting of the American Association for the Study of Liver Diseases (AASLD). November 7 – 11, 2014. Boston, MA. 92
Dore GJ, Feld JJ. Hepatitis C virus therapeutic development: in pursuit of perfectovir. Clin Infect Dis. 2015 Mar 11. pii: civ197 35
Scarica

Overview of New Treatments for Hepatitis C Virus