Overview on Hepatitis C, Treatment Strategy, Instrumental Analysis of Anti-HCV drugs

Authors

Adel E. Ibrahim1, Magda Elhenawee2, Hanaa Saleh2, Mahmoud M. Sebaiy3*
1Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Port Said University, Port Said, 42511, Egypt.
2Department of Analytical Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt.
3Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.

Article Information

*Corresponding author: Mahmoud M. Sebaiy, Department of Medicinal Chemistry, Faculty of Pharmacy, Zagazig University, Zagazig, 44519, Egypt.
Received: April 03, 2021
Accepted: April 16, 2021
Published: April 20, 2021
Citation: Adel E. Ibrahim, Magda Elhenawee, Hanaa Saleh, Mahmoud M. Sebaiy. “Overview on Hepatitis C, Treatment Strategy, Instrumental Analysis of Anti-HCV drugs’’. J Pharmacy and Drug Innovations, 2(2); DOI: http;//doi.org/03.2020/1.1014.
Copyright: © 2021 Mahmoud M. Sebaiy. This is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

This literature review is dealing with Hepatitis C, treatment strategy, and different analytical techniques for determination of recently used anti-HCV drugs specifically Sofosbuvir, Simeprevir, Ledipasvir, Daclatasvir and Velpatasvir in different pharmaceutical and biological samples.


Keywords: hepatitis c; analytical techniques; sofosbuvir; simeprevir; ledipasvir; daclatasvir; velpatasvir

Hepatitis C
Liver is a very vital organ responsible for many important processes inside the body. Such processes include digestion of food, metabolism of carbohydrates, fats and proteins, storage of glucose,  iron and vitamins (A, D, E, K and B12), production of some plasma proteins, regulating blood clotting, detoxification of blood from harmful substances (eg. Alcohol and drugs), immunity by producing immune factors and removing bacteria from blood stream……..etc [
1, 2]. Hepatitis is defined as the inflammation of liver. Toxins, heavy alcohol consumption and even some medications can cause hepatitis. The most common cause for hepatitis is viral infection with viral Hepatitis A, Hepatitis B and Hepatitis C viruses. Hepatitis A is spread via foecal-oral route, while Hepatitis B and C are spread via contaminated blood and blood products. Viral Hepatitis A and B can be prevented by immunization. Acute hepatitis A and B infections resolve without treatment in the majority of adults. Medications are mostly given to relieve their symptoms [3].

Hepatitis C virus (HCV) is the most dangerous type of this group of viruses. HCV was first discovered in 1989 and is now affecting more than 170 million people worldwide [4]. Since then a lot of research had been made on HCV which led to decrease in its transmission. The highest prevalence is found in North Africa, Middle East and central Asia with mortality rate around 350 000 deaths per year [5].

Hepatitis C is called a silent disease because people can be infected without knowing it. Also symptoms from acute infections can include fever, gastric upset and feeling fatigue which are common with many other diseases. Only 15-25% of acute infections can be self limiting, while more than 80% develop chronic HCV infections which can take decades to develop its symptoms. But when symptoms appear, they are often indicating advanced liver disease. Chronic HCV infections are serious and can lead to liver damage, cirrhosis, liver failure and carcinoma [6].Acute HCV infections are asymptomatic and about 15-20% of patients can clear acute infections, while the remaining majority develops into chronic infections [7]. Chronic HCV infection affects liver cells leading to cirrhosis, end-stage liver disease, and hepatic cancer. Chronic HCV infections caused about 350000 deaths per year [8].

HCV is a single-stranded RNA virus. A single HCV polyprotein is translated, and then cleaved by cellular and viral proteases into three structural proteins (core, E1, and E2) and seven non-structural proteins (p7, NS2, NS3, NS4A, NS4B, NS5A and NS5B) [9].  There are at least 6 genotypes of HCV that have arisen from the fast viral replication enabling it to adapt to host immune response and antiviral drugs. Genotyping is used as treatment guide since some viral genotypes respond better than others to different therapies.

HCV transmission is mainly by blood; however rare cases are transmitted sexually especially when associated with human Immunodeficiency virus (HIV) or on having relationships with multiple partners.  Blood transfusion and reuse of injection needles are the major transmission routes globally. Mother to child transmission during pregnancy occurs in 2-8% from infected mothers [10].  Sexual transmission of HCV to partner is very rare [11].

HCV diagnosis relies on HCV antibodies detection and tests that detect HCV RNA. Presence of HCV antibodies without detection of viral RNA resolved or treated infections, but detecting both simultaneously indicates current infection. HCV is a single stranded RNA virus. HCV single poly-protein of 3011 amino acids is translated then cleaved by viral and cellular proteases into three structural proteins and seven non-structural proteins that are (P7, NS2, NS3, NS4A, NS4B, NS5A and NS5B) [9].

Seven genotypes of HCV were discovered. Genotype 1 is the most common worldwide genotype in about 46.2% of HCV infected population. Genotype 2 occurs to about 9.1% of HCV infections and was found to be the easiest for treatment. Genotype 3 occurs to about 30% of infected population globally and is the most difficult genotype in treatment. Genotype 4 is confined mostly to Africa. Egypt is considered the highest in prevalence of HCV in the world and 90% of infected Egyptians are infected with genotype 4. Genotypes 5, 6 and 7 occur in smaller prevalence [12].

HCV treatment

Early treatment of HCV was found to be much more effective than late stage treatments. Milestones in HCV treatment are summarized in (Figures 1 [13] and 2 [14]). In 1991, FDA approved the first interferon (Intron A) for treatment of HCV. Ribavirin was then approved in 1998 to be used in combination with interferon for treatment [15]. The leap forward in HCV treatment wasn’t until the discovery of directly acting antiviral drugs (DAADs).

In 2011, FDA approved triple therapy using two DAADs, boceprevir and telaprevir, together with pegylated interferon and ribavirin. Unfortunately, the safety profile of this treatment was poor. In 2013, FDA approved three DAADs; Simeprevir (SMP) as new NS3 protease inhibitor, Sofosbuvir (SFS) as NS5B RNA polymerase inhibitor and daclatasvir (DAC) as NS5A inhibitor. These drugs were approved as combination therapy with peg-interferon and ribavirin for 12-24 weeks [10]. Ledipasvir (LDS) and velpatasvir (VLP) were approved as NS5A inhibitors in 2014 and 2015, respectively. It wasn’t until late 2014 when interferon free regimen was approved. 

The new regimens comprise a NS5B nucleoside inhibitor (eg. SFS) together with a protease inhibitor (eg.SMP) or NS5A inhibitor (eg.LDS, DAC, VLP), with or without ribavirin. The first approved combined single dose tablet was Harvoni® (SFS and LDS) then in 2016 Epclusa® (SFS plus VLP) was approved by FDA.

hcv milestone

 

 

 

 

 

 

 

 

 

 

 

Figure 1: Milestones in HCV treatment.

Figure 2: DAADs discovery milestone

Sofosbuvir (SFS) [16, 17]
* Molecular formula:     C22H29FN3O9P.
* Molecular weight:       529.453 g/mol.
*IUPAC name: propan-2-yl (2S)-2-[[[(2R,3R,4R,5R)-5-(2,4-dioxopyrimidin-1-yl)-4-fluoro-3-hydroxy-4-methyloxolan-2-yl]methoxy-phenoxyphosphoryl]amino]propanoate (Figure 3). 

Figure 3: Sofosbuvir chemical structure

 Physical properties: 
SFS is white to off-white crystalline powder which is slightly soluble in water and soluble in methanol. It has pKa values of 9.38 and 10.30.
*Mechanism of action:
SFS is a prodrug which is metabolized by body into its active form that act as defective nucleotide analog for HCV NS5B (non-structural protein 5B) RNA-dependent RNA polymerase. SFS prevents HCV viral replication by binding to HCV NS5B polymerase's active site and preventing further replication of HCV genetic material. After oral administration of the drug, SFS reaches its maximum plasma concentration in about 0.5 to 2 hours with a maximal concentration (Cmax) of 567ng/mL.
*Literature review:
Reviewing literature revealed several methods for determination of SFS in pharmaceutical products and in

plasma using liquid chromatography Table 1 summarizes LC methods for determination of SFS.

 

Matrix

Column

Mobile phase

System

Ref. No

 

Plasma

C18

0.1% formic acid: ACN (50:50)

UPLC-MS/MS

[18]

Plasma

C18

0.1% formic acid: ACN (gradiant)

UPLC-MS/MS

[19]

Tablets

C18

Water:MeOH (70:30)

HPLC-MS/MS

[20]

Plasma

C18

0.5% formic acid: MeOH (30:70)

UPLC-MS/MS

[21]

Plasma

C18

0.1% Phosphoric acid: ACN (68:32)

HPLC-UV 228nm

[22]

Tablets

C18

Phosphate buffer pH 4.0: MeOH (50:50)

HPLC-UV 262nm

[23]

Tablets

C18

0.1% formic acid: ACN (60:40)

HPLC-UV 260nm

[24]

Tablets

C18

MeOH: ACN  (90:10)

HPLC-UV 260nm

[25]

Tablets

C18

0.2% Phosphoric acid: ACN (gradiant)

UPLC-UV & MS/MS

[26]

Serum

C18

0.05% Phosphoric acid: ACN (gradiant)

HPLC-UV

[27]

Table 1Table 1: Summary of reported LC methods for SFS determination

Other spectrophotometric methods were published [24, 25, 28]. The spectrophotometric methods were done at wavelength 260nm using methanol as organic solvent. The limits of quantitation for the reported spectrophotometric methods were higher than 2.5µg/mL and the determination ranges started from 5-100µg/mL .

Only one HPTLC and CE method for SFS determination was reported [24]. HPTLC method used range of 25-100µg/mL and had the lowest sensitivity that LOQ was 7µg/mL. The highest sensitivity was for CE method and LOQ was 0.3µg/mL

Simeprevir (SMP) [16, 17]

* Molecular formula:     C38H47N5O7S2.

* Molecular weight:       749.942 g/mol.

*IUPAC name: (1R,4R,6S,7Z,15R,17R)-N-(cyclopropanesulfonyl)-2-hydroxy-17-([7-methoxy-8-methyl-2-[4-(propan-2-yl)-1,3-thiazol-2-yl]quinolin-4-yl]oxy)-13-methyl-14-oxo-3,13-diazatricyclo[13.3.0.0] octadeca-2,7-diene-4-carboximidic acid (Figure 4).

smp

   Figure 4: Simeprevir chemical structure

* Physical properties:

 SMP is white to almost-white powder insoluble in water and slightly soluble in acetone and freely soluble in dichloromethane. It has pKa valueof 5.87. The drug is pharmaceutically available as water soluble sodium salt.

* Mechanism of action:

SMP inhibits HCV polyprotein cleavage via induced-fit binding to an NS3 catalytic site. After oral administration of the drug, SMP reaches its maximum plasma concentration in about 4 to 6 hours.

* Literature review:

Reviewing literature, A spectrophotometric method for determination of SMP in pharmaceutical dosage forms was published [29]. SMP was determined in presence of its oxidative degradation product at the range of 2.5-403µg/mL using different apectrophotometricmethods. Other LC determinations of SMP are summarized in Table 2.

 

Matrix

 

Column

 

Mobile phase

 

System

Ref. No.

Plasma

C18

Phosphate buffer: ACN (30:70)

HPLC-UV

[30]

Tablets

C18

ACN

HPLC-UV

[31]

Plasma

C18

AmmoniumFormate: ACN: MeOH (gradiant)

HPLC-MS/MS

[32]

Table 2: Summary of reported LC methods for SMP determination

Ledipasvir (LDS)  [16, 17]

* Molecular formula:     C49H54N8O6F2.

* Molecular weight:       889.0 g/mol.

*IUPAC name: methyl [(2S)-1-[(6S)-6-[4-(9,9-difluoro-7-[2-[(1R,3S,4S)-2-[(2S)-2-[(methoxycarbonyl)amino]-3-methylbutanoyl]-2-azabicyclo[2.2.1]hept-3-yl]-1H-benzimidazol-5-yl]-9H-fluoren-2-yl)-1H-imidazol-2-yl]-5-azaspiro[2.4]hept-5-yl]-3-methyl-1-oxobutan-2-yl]carbamate (Figure 5).

lds

Figure 5: Ledipasvir chemical structure

 

* Physical properties:

LDS is white to beige powder. LDS is Insoluble in water, but soluble in acetonitrile and in methanol. It has pKa values of 4.0 and 5.0. The drug is pharmaceutically available as water soluble acetone solvate and Ledipasvircopovidone mixture.

*Mechanism of action:

LDS prevent hyperphosphorylation of NS5A polymerase which is required for viral protein production. It was found to be effective against HCV genotypes (1, 4, and 5) and with a lesser activity against genotypes (2 and 3). After oral administration of the drug, LDS reaches its maximum plasma concentration in about 4 to 4.5 hours with Cmaxof 323 ng/mL.

*Literature review:

Only two papers described the determination of LDS alone in tablet dosage forms [33, 34]. However; being only formulated in combination with sofosbuvir, several papers described TLC [35], spectrophotometric [36-40] and spectrofluorimetric [41, 42] besides chromatographic determinations of both drugs in several matrices.  Table 3 summerizes some LC methods published for simultaneous determination of SFS and LDS.

 

Matrix

 

Column

 

Mobile phase

 

System

Ref. No.

Plasma

C18

0.1% formic acid: ACN (gradiant)

UPLC-MS/MS

[43]

Plasma

C18

0.1% formic acid: ACN (50:50)

UPLC-MS/MS

[44]

Plasma

C8

Ammonium formatebuffer:ACN:MeOH (gradiant)

HPLC-MS/MS

[45]

Plasma

C18

0.1% formic acid in MeOH:ACN: Acetic acid (63:25:12)

HPLC-MS/MS

[46]

Tablet

C18

Ammonium acetate buffer: ACN (35:65)

HPLC-UV 245nm

[47]

Tablets

C18

Phosphate buffer : ACN (50:50)

HPLC-UV 254nm

[48]

Tablets

C18

Trifluoroacetic in water: ACN:MeOH (30:50:20)

HPLC-UV 267nm

[49]

Table 3: Summary of reported LC methods for LDS determination

Daclatasvir (DAC) [16, 17]

* Molecular formula:     C40H50N8O6.

* Molecular weight:       738.89 g/mol.

* IUPAC name: Dimethyl N,N'-([1,1'-biphenyl]-4,4'-diylbis[1H-imidazole-5,2-diyl-[(2S)-pyrrolidine-2,1-diyl][(2S)-3-methyl-1-oxobutane-1,2-diyl]])dicarbamate (Figure 6).

DAC

Figure 6: Daclatasvir chemical structure

*Physical properties:

DAC is white powder freely soluble in water. It has pKa values of 6.0 and 11.1. The drug is pharmaceutically available as dihydrochloride salt.

*Mechanism of action:

DAC inhibits HCV viral RNA replication and protein translation by directly inhibiting HCV protein NS5A polymerase. NS5A is critical for HCV viral transcription and translation, and as of 2014 it appeared that resistance can arise to daclatasvir fairly swiftly 

After oral administration of the drug, DAC reaches its maximum plasma concentration in about 2 hours with bioavailability of 67%.

*Literature review:

A spectrophotometric and another spectrofluorimetric methods were published for determination of DAC [50, 51]. The spectrophotometric method used methanol as solvent for quantitation of DAC in range of 2-12µg/mL at wavelength 317nm. The spectrofluorimetric method was for simultaneous determination of DAC in presence of LDS.

Other LC methods for determination of DAC were reported either alone or in combination with sofosbuvir. Table 4 summarizes those reported chromatographic determinations.

Matrix

Column

Mobile phase

System

Ref. No.

Plasma

C18

Ammonium formate buffer: ACN (50:50)

UPLC-MS/MS

[52]

Tablets

C8

Phosphate buffer: ACN (75:25)

HPLC-UV 306nm

[53]

powder

Amylose

ACN:MeOH containing diethylamine (gradiant)

HPLC-UV 315nm

[54]

Tablets

C18

Phosphate buffer: ACN (60:40)

HPLC-UV 315nm

[55]

Plasma

C18

Ammonium acetate buffer: ACN (56:44)

HPLC-UV 318nm

[56]

Plasma

C18

0.1% formic acid: ACN

UPLC-MS/MS

[57]

Plasma

C18

Ammonium formate buffer: ACN (50:50)

UPLC-MS/MS

[58]

Tablets

C18

Phosphate buffer: ACN  (60:40)

HPLC-UV 265nm

[59]

Tablets

C18

Phosphate buffer: ACN  (50:50)

HPLC-UV 320nm

[60]

Table 4: Summary of reported LC methods for DAC determination
Velpatasvir (VLP) [
16, 17]

* Molecular formula:     C49H54N8O8.

* Molecular weight:       883.019 g/mol.

*IUPAC name: Methyl [(2S)-1-[(2S,5S)-2-(9-[2-[(2S,4S)-1-[(2R)-2-[(methoxycarbonyl)amino]-2-phenylacetyl]-4-(methoxymethyl)-2-pyrrolidinyl]-1H-imidazol-4-yl]-1,11-dihydroisochromeno[4',3':6,7] naphtho[1,2-d]imidazol-2-yl)-5-methyl-1-pyrrolidinyl]-3-methyl-1-oxo-2-butanyl]carbamate (Figure 7).

Velpatasvir

Figure 7: Velpatasvir chemical structure

*Physical properties:

VLP is off-white to yellow color powder insoluble in water and slightly soluble in methanol. It has pKa values of 3.8 and 6.0. The drug is pharmaceutically available as VLP copovidone mixture.

*Mechanism of action:

VLP is non-structural protein NS5A polymerase inhibitor. It inhibits HCV viral RNA replication and protein translation. After oral administration, VLP is 99.5% bound to plasma proteins with bioavailability of 25-30%.

*Literature review:

Only two LC methods were reported for determination of VLP in tablet dosage forms as listed in Table 5.

 

Matrix

 

Column

 

Mobile phase

 

System

Ref. No.

Tablets

C18

0.1% phosphoric acid in water:ACN (45:55)

HPLC-UV 240 nm

[61]

Tablets

C18

0.1% phosphoric acid in water:ACN (45:55)

HPLC-UV 260 nm

[62]

Table 5: Summary of reported LC methods for VLP determination

Although some papers described validated methods for simultaneous determination of two DAADs (e.g. SFS/LDS, SFS/VLP, SFS/DAC), only two LC methods were developed for the quantification of some of the new DAADs using UHPLC-MS/MS [63, 64].

Conclusion:

This literature review is introducing brief summary about Hepatitis C disease, its treatment strategy, and instrumental analysis of recently used anti-HCV drugs specifically Sofosbuvir, Simeprevir, Ledipasvir, Daclatasvir and Velpatasvir in different matrices.

 

References

  1. Tortora, G.J. and Derrickson, B.H., Principles of anatomy and physiology. (2008): John Wiley & Sons.
  2. Barrett, K., Chapter 10. Functional anatomy of the liver and biliary system. Gastrointestinal Physiology. 2nd ed. New York, NY: McGraw-Hill, 2014.
  3. Sweetman, S., Martindale: the complete drug reference. Vol. 36. (2009): London: Pharmaceutical Press.
  4. Mohd Hanafiah, K., Groeger, J., Flaxman, A.D., and Wiersma, S.T., Global epidemiology of hepatitis C virus infection: New estimates of age‐specific antibody to HCV seroprevalence. Hepatology, 2013. 57(4): p. 1333-1342.
  5. Zaltron, S., Spinetti, A., Biasi, L., Baiguera, C., and Castelli, F., Chronic HCV infection: epidemiological and clinical relevance. BMC infectious diseases, 2012. 12(2): p. S2.
  6. National Institutes of Health Consensus Development Conference Statement: Management of hepatitis C 2002. Gastroenterology. 123(6): p. 2082-2099.
  7. Thomson, E.C., Smith, J.A., and Klenerman, P., The natural history of early hepatitis C virus evolution; lessons from a global outbreak in human immunodeficiency virus-1-infected individuals. Journal of General Virology, 2011. 92(10): p. 2227-2236.
  8. Szabó, E., Lotz, G., Páska, C., Kiss, A., and Schaff, Z., Viral hepatitis: new data on hepatitis C infection. Pathology & Oncology Research, 2003. 9(4): p. 215.
  9. Halliday, J., Klenerman, P., and Barnes, E., Vaccination for hepatitis C virus: closing in on an evasive target. Expert review of vaccines, 2011. 10(5): p. 659-672.
  10. Floreani, A., Hepatitis C and pregnancy. World journal of gastroenterology: WJG, 2013. 19(40): p. 6714.
  11. Terrault, N.A., Dodge, J.L., Murphy, E.L., Tavis, J.E., Kiss, A., Levin, T., Gish, R.G., Busch, M.P., Reingold, A.L., and Alter, M.J., Sexual transmission of hepatitis C virus among monogamous heterosexual couples: the HCV partners study. Hepatology, 2013. 57(3): p. 881-889.
  12. Smith, D.B., Bukh, J., Kuiken, C., Muerhoff, A.S., Rice, C.M., Stapleton, J.T., and Simmonds, P., Expanded classification of hepatitis C virus into 7 genotypes and 67 subtypes: updated criteria and genotype assignment web resource. Hepatology, 2014. 59(1): p. 318-327.
  13. Heim, M.H., 25 years of interferon-based treatment of chronic hepatitis C: an epoch coming to an end. Nature reviews Immunology, 2013. 13(7): p. 535.
  14. Webster, D.P., Klenerman, P., and GM, D., Hepatitis C. The Lancet, 2015. 385(9973): p. 1124-1135.
  15. Strader, D.B. and Seeff, L.B., A brief history of the treatment of viral hepatitis C. Clinical Liver Disease, 2012. 1(1): p. 6-11.
  16. https://pubchem.ncbi.nlm.nih.gov.
  17. https://www.drugbank.ca.
  18. Rezk, M.R., Basalious, E.B., and Karim, I.A., Development of a sensitive UPLC-ESI-MS/MS method for quantification of sofosbuvir and its metabolite, GS-331007, in human plasma: application to a bioequivalence study. Journal of pharmaceutical and biomedical analysis, 2015. 114: p. 97-104.
  19. Shi, X., Zhu, D., Lou, J., Zhu, B., Hu, A.-r., and Gan, D., Evaluation of a rapid method for the simultaneous quantification of ribavirin, sofosbuvir and its metabolite in rat plasma by UPLC–MS/MS. Journal of Chromatography B, 2015. 1002: p. 353-357.
  20. Nebsen, M. and Elzanfaly, E.S., Stability-indicating method and LC–MS-MS characterization of forced degradation products of sofosbuvir. Journal of chromatographic science, 2016. 54(9): p. 1631-1640.
  21. Gandhi, B., Rao, A., and Rao, J. UPLC-MS/MS method for determination of sofosbuvir in human plasma. in Annales pharmaceutiques francaises. 2017. Elsevier.
  22. MADHAVI, S. and RANI, A.P., BIOANALYTICAL METHOD DEVELOPMENT AND VALIDATION FOR THE DETERMINATION OF SOFOSBUVIR FROM HUMAN PLASMA. 2016.
  23. Vikas, P.M., Vikas, P.M., Satyanarayana, D.T., Kumar, D.V., Mounika, E., Latha, M.S., Anusha, R., and Sathish, Y., Development and validation of new RP-HPLC method for the determination of sofosbuvir in pure form. World Journal of pharmacy and pharmaceutical Sciences, 2016. 5(5): p. 775-781.
  24. El-Yazbi, A.F., Comparative Validation of the Determination of Sofosbuvir in Pharmaceuticals by Several Inexpensive Ecofriendly Chromatographic, Electrophoretic, and Spectrophotometric Methods. Journal of AOAC International, 2017. 100(4): p. 1000-1007.
  25. Abdel-Gawad, S.A.-N., Simple chromatographic and spectrophotometric determination of sofosbuvir in pure and tablet forms. European Journal of Chemistry, 2016. 7(3): p. 375-379.
  26. Contreras, M.d.M., Morales-Soto, A., Segura-Carretero, A., and Valverde, J., Potential of RP-UHPLC-DAD-MS for the qualitative and quantitative analysis of sofosbuvir in film coated tablets and profiling degradants. Journal of Pharmaceutical Analysis, 2017. 7(4): p. 208-213.
  27. Miraghaei, S., Mohammadi, B., Babaei, A., Keshavarz, S., and Bahrami, G., Development and validation of a new HPLC-DAD method for quantification of sofosbuvir in human serum and its comparison with LC–MS/MS technique: Application to a bioequivalence study. Journal of Chromatography B, 2017. 1063: p. 118-122.
  28. El Hamd, M.A., Ali, R., Marzouk, A.A., and Abdelmageed, O.H., Validated Ultraviolet-Spectrometric Method for Determination of Sofosbuvir in Tablets Formulation. Journal of Applied Pharmaceutical Science Vol, 2017. 7(02): p. 114-119.
  29. Attia, K.A.M., El-Abasawi, N.M., El-Olemy, A., and Serag, A., Different spectrophotometric methods applied for the analysis of simeprevir in the presence of its oxidative degradation product: Acomparative study. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 2018. 190: p. 1-9.
  30. Nannetti, G., Pagni, S., Parisi, S.G., Alberti, A., Loregian, A., and Palù, G., Development of a simple HPLC–UV method for the determination of the hepatitis C virus inhibitor simeprevir in human plasma. Journal of Pharmaceutical and Biomedical Analysis, 2016. 121: p. 197-203.
  31. Attia, K.A., El-Abasawi, N.M., El-Olemy, A., and Serag, A., Stability-indicating HPLC-DAD Method for the Determination of Simeprevir. Analytical Chemistry Letters, 2017. 7(1): p. 43-51.
  32. Vanwelkenhuysen, I., de Vries, R., Timmerman, P., and Verhaeghe, T., Determination of simeprevir: A novel, hepatitis C protease inhibitor in human plasma by high-performance liquid chromatography–tandem mass spectrometry. Journal of Chromatography B, 2014. 958: p. 43-47.
  33. Swain, D. and Samanthula, G., Study on the forced degradation behaviour of ledipasvir: Identification of major degradation products using LC–QTOF–MS/MS and NMR. Journal of Pharmaceutical and Biomedical Analysis, 2017. 138: p. 29-42.
  34. Devilal, J., Durgaprasad, B., Pal, N., and Rao, A.S., New method development and validation for the determination of ledipasvir in bulk drug form by using reverse phase HPLC technique. World Journal of Pharmacy and Pharmaceutical Science, 2016. 5(8): p. 1312-1321.
  35. Salama, F.M., Attia, K.A., Abouserie, A.A., El-Olemy, A., and Abolmagd, E., Application of TLC Densitometric Method for Simultaneous Estimation of the Newly Co-formulated Antiviral Agents Ledipasvir and Sofosbuvir in Their Tablet Dosage Form. Analytical Chemistry Letters, 2017. 7(2): p. 241-247.
  36. Mansour, F.R., A new innovative spectrophotometric method for the simultaneous determination of sofosbuvir and ledipasvir. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 2018. 188: p. 626-632.
  37. Rai, S.P.Y., Prajapati, Y., and Patni, P., development and Validation of RP-HPLC and UV Spectroscopic methods for Simultaneous Estimation of Sofosbuvir and Ledipasvir in their combined tablet dosage forms. An International journal of Pharmaceutical Sciences, 2017. 8(2).
  38. SALAMA, F.M., ATTIA, K.A., ABOUSERIE, A.A., EL-OLEMY, A., and ABOLMAGD, E., DIFFERENT SPECTRAL DATA PROCESSING TECHNIQUES FOR DETERMINATION OF LEDIPASVIR AND SOFOSBUVIR IN THEIR PURE AND DOSAGE FORMS; A COMPARATIVE STUDY. 2017.
  39. Baker, M.M., El-Kafrawy, D.S., Mahrous, M.S., and Belal, T.S., Validated spectrophotometric and chromatographic methods for analysis of the recently approved hepatitis C antiviral combination ledipasvir and sofosbuvir. Annales Pharmaceutiques Françaises, 2018. 76(1): p. 16-31.
  40. Eissa, M.S., Simultaneous determination of the brand new two-drug combination for the treatment of hepatitis C: Sofosbuvir/ledipasvir using smart spectrophotometric methods manipulating ratio spectra. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 2017. 183: p. 362-370.
  41. Salama, F.M., Attia, K.A., Abouserie, A.A., El-Olemy, A., and Abolmagd, E., Spectroflurimetric estimation of the new antiviral agent ledipasvir in presence of sofosbuvir. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 2018. 190: p. 513-517.
  42. Khalili, M., Sohrabi, M.R., Mirzabeygi, V., and Torabi Ziaratgahi, N., Chemometric simultaneous determination of Sofosbuvir and Ledipasvir in pharmaceutical dosage form. Spectrochimica Acta Part A: Molecular and Biomolecular Spectroscopy, 2018. 194: p. 141-151.
  43. Pan, C., Chen, Y., Chen, W., Zhou, G., Jin, L., Zheng, Y., Lin, W., and Pan, Z., Simultaneous determination of ledipasvir, sofosbuvir and its metabolite in rat plasma by UPLC–MS/MS and its application to a pharmacokinetic study. Journal of Chromatography B, 2016. 1008: p. 255-259.
  44. Rezk, M.R., Bendas, E.R., Basalious, E.B., and Karim, I.A., Quantification of sofosbuvir and ledipasvir in human plasma by UPLC–MS/MS method: Application to fasting and fed bioequivalence studies. Journal of Chromatography B, 2016. 1028: p. 63-70.
  45. Abdallah, O.M., Abdel-Megied, A.M., and Gouda, A.S., Development a validated highly sensitive LC–MS/MS method for simultaneous quantification of Ledipasvir, sofosbuvir and its major metabolite GS-331007 in human plasma: Application to a human pharmacokinetic study. Journal of Pharmaceutical and Biomedical Analysis, 2017. 143: p. 305-310.
  46. Elkady, E.F. and Aboelwafa, A.A., A rapid and optimized LC-MS/MS method for the simultaneous extraction and determination of sofosbuvir and ledipasvir in human plasma. Journal of AOAC International, 2016. 99(5): p. 1252-1259.
  47. Zaman, B., Siddique, F., and Hassan, W., RP-HPLC method for simultaneous determination of sofosbuvir and ledipasvir in tablet dosage form and its application to in vitro dissolution studies. Chromatographia, 2016. 79(23-24): p. 1605-1613.
  48. Hassouna, M.E.-K.M., Abdelrahman, M.M., and Mohamed, M.A., Assay and Dissolution Methods development and validation for simultaneous determination of Sofosbuvir and Ledipasvir by RP-HPLC method in tablet dosage forms. Journal of Forensic Science & Criminal Investigations, 2017. 1(3): p. 555-562.
  49. Nagaraju, T., Vardhan, S., Kumar, D.R., and Ramachandran, D., A New RP-HPLC Method for the Simultaneous Assay of SOFOSBUVIR and LEDIPASVIR in Combined Dosage Form. International Journal of Chemtech Research, 2017. 10(7): p. 761-769.
  50. Kekan, V., Gholve, S., and Bhusnure, O., Development, Validation and Stability Study of UV Spectrophotometric Method for Determination of Daclatasvirin Bulk and Pharmaceutical Dosage Forms. Int J ChemTech Res, 2017. 10: p. 281-287.
  51. Abo-Zeid, M.N., Atia, N.N., El-Gizawy, S.M., and El-Shaboury, S.R., Ultrasensitive spectrofluorimetric method for rapid determination of daclatasvir and ledipasvir in human plasma and pharmaceutical formulations. Journal of Pharmaceutical and Biomedical Analysis, 2018.
  52. Rezk, M.R., Bendas, E.R., Basalious, E.B., and Karim, I.A., Development and validation of sensitive and rapid UPLC–MS/MS method for quantitative determination of daclatasvir in human plasma: Application to a bioequivalence study. Journal of pharmaceutical and biomedical analysis, 2016. 128: p. 61-66.
  53. Baker, M.M., El-Kafrawy, D.S., Mahrous, M.S., and Belal, T.S., Validated stability-indicating HPLC-DAD method for determination of the recently approved hepatitis C antiviral agent daclatasvir. Annales Pharmaceutiques Françaises, 2017. 75(3): p. 176-184.
  54. Srinivasu, G., Kumar, K.N., Thirupathi, C., Narayana, C.L., and Murthy, C.P., Development and validation of the chiral HPLC method for daclatasvir in gradient elution mode on amylose-based immobilized chiral stationary phase. Chromatographia, 2016. 79(21-22): p. 1457-1467.
  55. Hassib, S.T., Taha, E.A., Elkady, E.F., and Barakat, G.H., Reversed-Phase Liquid Chromatographic Method for Determination of Daclatasvir Dihydrochloride and Study of Its Degradation Behavior. Chromatographia, 2017. 80(7): p. 1101-1107.
  56. Nannetti, G., Messa, L., Celegato, M., Pagni, S., Basso, M., Parisi, S.G., Palù, G., and Loregian, A., Development and validation of a simple and robust HPLC method with UV detection for quantification of the hepatitis C virus inhibitor daclatasvir in human plasma. Journal of Pharmaceutical and Biomedical Analysis, 2017. 134: p. 275-281.
  57. Notari, S., Tempestilli, M., Fabbri, G., Libertone, R., Antinori, A., Ammassari, A., and Agrati, C., UPLC–MS/MS method for the simultaneous quantification of sofosbuvir, sofosbuvir metabolite (GS-331007) and daclatasvir in plasma of HIV/HCV co-infected patients. Journal of Chromatography B, 2018. 1073: p. 183-190.
  58. Abdallah, O.M., Abdel‐Megied, A.M., and Gouda, A.S., Development and validation of LC‐MS/MS method for simultaneous determination of sofosbuvir and daclatasvir in human Plasma: Application to pharmacokinetic study. Biomedical Chromatography, 2018.
  59. Eldin, A.S., Azab, S.M., Shalaby, A., and El-Maamly, M., The Development of A New Validated HPLC and Spectrophotometric Methods for the Simultaneous Determination of Daclatasvir and Sofosbuvir: Antiviral Drugs. Journal of Pharmacy and Pharmacology Research, 2017. 1(1): p. 28-42.
  60. Saleh, H., Ragab, G.H., and Othman, M.A., Stability indicating HPLC method development and validation for determination of daclatasvir in pure and tablet dosage forms. Indo Am. J. pharm. Sci, 2016. 3: p. 1565-1572.
  61. Nalla, S. and Seshagiri Rao, J., A Stability indicating RP-HPLC method for simultaneous estimation of Velpatasvir and Sofosbuvir in combined tablet dosage forms. World Journal of Pharmacy and Pharmaceutical Sciences, 2017. 6(9): p. 1596-1611.
  62. RANI, J.S. and DEVANNA, N., A New RP-HPLC Method Development and Validation for Simultaneous Estimation of Sofosbuvir and Velpatasvir in Pharmaceutical Dosage Form. 2017.
  63. Ariaudo, A., Favata, F., De Nicolò, A., Simiele, M., Paglietti, L., Boglione, L., Cardellino, C.S., Carcieri, C., Di Perri, G., and D’Avolio, A., A UHPLC–MS/MS method for the quantification of direct antiviral agents simeprevir, daclatasvir, ledipasvir, sofosbuvir/GS-331007, dasabuvir, ombitasvir and paritaprevir, together with ritonavir, in human plasma. Journal of pharmaceutical and biomedical analysis, 2016. 125: p. 369-375.
  64. Jiang, H., Kandoussi, H., Zeng, J., Wang, J., Demers, R., Eley, T., He, B., Burrell, R., Easter, J., and Kadiyala, P., Multiplexed LC-MS/MS method for the simultaneous quantitation of three novel hepatitis C antivirals, daclatasvir, asunaprevir, and beclabuvir in human plasma. Journal of pharmaceutical and biomedical analysis, 2015. 107: p. 409-418.