SlideShare une entreprise Scribd logo
1  sur  7
Télécharger pour lire hors ligne
1


2010


    Discovery & Development of
     a First in Class Anti-Ebola
         Drug- EBONAVIR®
   Summary:
   Ebola hemorrhagic fever (Ebola HF) caused by the Ebola virus
   is a severe, often-fatal viral hemorrhagic disease in humans and
   nonhuman primates with a fatality rate of 50-83%. The present
   document details the discovery and development of a novel
   small molecule organo-pharmaceutical that acts as a potent
   inhibitor of VP35 - a critical protein involved in EBOV
   pathogenesis through early preclinical studies as well as
   clinical studies. It provides an insight into the research,
   documentation and regulatory requirements pertaining to the
   commercial launch of the above mentioned small molecule
   drug.




                                      GAYATHRI VIJAYAKUMAR
                                    MS Biotechnology, NYU-POLY
                                                       5/1/2010
2




                                             CONTENTS



1) Introduction                                                                          5
   a) Ebola Virus                                                                        5
   b) Transmission                                                                       5
   c) Symptoms                                                                           5
   d) Diagnosis                                                                          6
   e) Treatment                                                                          6
   f) History                                                                            6
2) Current therapeutic advances for Ebola Hemorrhagic Fever                              6
3) Promising targets for development of therapeutics                                     7
4) Target for drug development                                                           8
   a) Reason for selection of VP35 as a therapeutic target                               8
   b) Impairment of innate immunity                                                     10
5) Medical hypothesis for developing an anti-Ebola therapeutic                          12
6) Conclusion regarding medical hypothesis                                              14
7) Assays for HTS screening of potential anti-Ebola therapeutic                         15
   a) Luciferase Budding Assay                                                          15
   b) Immunocapture Assay                                                               16
   c) Cell based assay using recombinant GFP-ZEBOV                                      16
8) Luciferase Reporter based gene assay selected for HTS screening                      18
   a) Illustration of principle                                                         21
9) Lead Compound                                                                        22
   a) Selected Lead Compound candidate                                                  24
10) Lead Optimization                                                                   25
   a) Lead Optimization results                                                         26
11) In vitro safety pharmacology profiling                                              28
   a) In vitro SPP Profile results                                                      33
12) Patent Application                                                                  35
13) Selection of appropriate animal model for efficacy evaluation of the selected NCE   36
   a) Mouse model used for preclinical studies                                          38
   b) Non-human primate model used for preclinical studies                              40
3

14) Biomarkers                                                       42
   a) Disease Biomarkers                                             43
   b) Surrogate Biomarker                                            45
   c) Toxicity Biomarker                                             45
   d) Target Biomarker                                               46
   e) Mechanism Biomarker                                            46
   f) Efficacy Biomarker                                             47
   g) Translational Biomarker                                        47
   h) Stratification Biomarker                                       47
15) Efficacy studies for Minimum Effective Dose                      48
   a) Minimum Effective Dose                                         49
16) Principal studies in animal models with NCT1087                  49
   a) Animal models in safety studies                                50
   b) Route of Administration of NCT1087                             51
   c) Safety Pharmacology Studies                                    51
   d) Acute Toxicity Study                                           53
   e) Repeated Dose Toxicity Study                                   53
   f) Genetoxicity Study                                             55
   g) Carcinogenicity Study                                          57
   h) Reproduction Toxicity Study                                    57
   i) Immunotoxicity Study                                           58
   j) Phototoxicty Study                                             59
17) Maximum Tolerated Dose                                           60
   a) Toxicity studies for determining the MTD                       60
18) Repeated dose toxicity studies for 2 months                      62
19) Estimation of the first human dose                               64
20) IND Application                                                  65
21) Phase I Clinical Trials                                          67
22) Phase II Clinical Trials                                         71
23) Phase III Clinical Trials                                        77
24) Conclusion                                                       83
25) Route of Delivery, Administration Regimen & Dose Concentration   83
26) NDA Application                                                  84
27) Life Cycle Management of NCT1087 or Ebonavir®                    85
28) The Drug Development process                                     86
4

29) Preclinical & Requirements for IND, NDA &Clinical studies   88
30) CMC Requirements for IND, NDA & Clinical studies            89
31) Possible Emergence of Resistance to Ebonavir®               90
32) References                                                  91
5


                                  EBOLA HEMORRHAGIC FEVER

       Ebola hemorrhagic fever (Ebola HF) is a severe, often-fatal viral hemorrhagic
                                                                                                          Figure 1. [3]
disease in humans and nonhuman primates (monkeys, gorillas, and chimpanzees)
that has appeared sporadically since its initial recognition in the Democratic Republic
of Congo in 1976 [1]. It is caused by the Ebola Virus which interferes with the
interior endothelial cell lining of blood vessels and coagulation which leads to
hypovolemic shock [2]. Destruction of endothelial surfaces is associated with
disseminated intravascular coagulation, and this may contribute to the
hemorrhagic manifestations [5]. Fatality rate differs from 50-83% [2].                                              [3]

EBOLA VIRUS (shown in image [6])
                                                                                                          Figure 2. [6]

       The Ebola virus belongs to the Filoviridae family (filovirus) and is
comprised of five distinct species: Zaire, Sudan, Côte d’Ivoire, Bundibugyo and
Reston. [4] The Zaire virus (ZEBOV) is the most lethal with an average case
fatality rate of 83% [2]. Ebola virus has a non-segmented, negative-stranded, RNA
genome containing 7 structural and regulatory genes [5]. The exact origin,
locations, and natural habitat (known as the "natural reservoir") of Ebola virus
remain unknown. Researchers believe that the virus is zoonotic (animal-borne) [4].
Electron microscopic image of an Ebola infected cell in [9] (Figure 3).

TRANSMISSION

   1) Direct contact with the blood and/or secretions of an infected person/ animal
       [1].
   2) Contact with objects, such as needles, that have been contaminated with
       infected secretions [1].
   3) Nosocomial transmission refers to the spread of a disease within a health-
       care setting, due to incorrect infection control precautions and adequate
       barrier nursing procedures [1, 4]
                                                                                          Figure 3: [9]

SYMPTOMS

       The incubation period for Ebola HF ranges from 2 to 21 days. The onset of illness is abrupt and
is characterized by fever, headache, joint and muscle aches, sore throat, and weakness, followed by diarrhea,
vomiting, and stomach pain. A rash, red eyes, hiccups and internal and external bleeding may be seen [1].
6

 DIAGNOSIS

           Diagnosis is usually done with inactivated blood or saliva specimens to detect the viral antigen/antibody or
 genetic material [4]. They can also be isolated in cell cultures. Diagnostic laboratory tests such as ELISA, RT-PCR,
 IgM ELISA, Immunohistochemistry testing, Coagulation studies; Complete Blood Count, etc are usually performed
 [1,7].

 TREATMENT

           There is no standard treatment for Ebola hemorrhagic
 fever. Treatment is primarily supportive and includes minimizing
 invasive procedures, balancing electrolytes, and, since patients
 are frequently dehydrated, replacing lost coagulation factors to
 help stop bleeding, maintaining oxygen and blood levels, and
 treating any complicating infections. Convalescent plasma,
 administration     of   an   inhibitor   of   coagulation   (rNAPc2),
 Morpholino antisense drugs have shown some promise in
 treatment for Ebola HF [2].

 HISTORY

           History of Ebola virus outbreaks have been shown in the
 table to the side (Figure 4). Confirmed cases of Ebola HF have
 been reported in the Democratic Republic of the Congo, Gabon,
 Sudan, the Ivory Coast, Uganda, and the Republic of the
 Congo. [1] Because of its high morbidity, it is a potential agent
 for bioterrorism and since no approved vaccine or drugs are
 available it is classified as a biosafety level 4 agents as well as
 Category A bioterrorism agent by the CDC [2]. The last
 reported outbreak was in December 2008 in the Western Kasai
 province of the Democratic Republic of Congo [2].
                                                                                   Figure 4: From [8]



          CURRENT THERAPEUTIC ADVANCES FOR EBOLA HEMORRHAGIC FEVER

           To date, beyond supportive care, no effective treatments, therapies, or vaccines are approved to treat or
prevent Ebola virus infections because natural immunity to this infection is difficult to find, plus there are no immune
correlates in humans [10]. However, several drugs are under development as summarized:
7

•   USAMRID investigated a drug called recombinant nematode anticoagulant protein C2 (rNAPC2) which blocks
    the harmful effects of tissue factors and targets the disease process. It showed reasonable success in infected
    rhesus monkeys [11].
•   AVI BioPharma’s proprietary NEUGENE drug targets a key Ebola gene providing complete protection in mice
    when administered either before or after an otherwise lethal infection with Ebola virus. NEUGENE antisense
    compounds are synthetic polymers designed to match up perfectly with a specific gene or viral sequence,
    blocking the function of the target gene or virus [12].
•   Phosphorodiamidate morpholino oligomers (PMO) are a class of uncharged single-stranded DNA analogs
    modified such that each subunit includes a phosphorodiamidate linkage and morpholinering. Data suggest that
    antisense PMO and P-PMO have the potential to control EBOV infection and are promising therapeutic
    candidates [13]. A combination of EBOV-specific PMOs targeting sequences of viral mRNAs for the viral
    proteins (VPs) VP24, VP35 has been used as both pre- and post- exposure therapeutic regimens in non-human
    primates [14].
•   NanoViricides, Inc. has come out with broad-spectrum nanoviricides(TM) drug candidates that have been found
    to be highly effective in cell culture studies by USAMRID. These antivirals use biomimetic technology (i.e. they
    mimic the host cell features) [15].
•   3-Deazaneplanocin A, an analog of adenosine, is a potent inhibitor of Ebola virus replication. A single dose early
    in infection prevents progression of the disease in EBOV infected mice. Protective effect of the drug results from
    massively increased production of interferon-α [17].
•   Cyanovirin-N (CV-N) which is an HIV inactivating protein has been found to have both in vitro and in vivo
    antiviral activity against the Zaire strain of the Ebola Virus by inhibiting the development of viral cytopathic
    effects (CPEs). It has been found to delay fatality in EBOV infected mice. It binds with considerable affinity to the
    Ebola envelope glycoprotein GP1,2 . [19].
•   EBOV infected immunocompetent mice treated with the adenosine analogue carbocyclic 3-deazaadenosine
    protects it against fatality by blocking the cellular enzyme S-adenosyl-L -homocysteine hydrolase (SAH) [20].
•   A vaccine based on recombinant vesicular stomatitis virus expressing the ZEBOV glycoprotein has been found
    to confer immunity against aerosol challenge in cynomolgus macaques which is likely to be the port of entry in a
    bioterrorist attack [21].
•   Intranasal vaccine based on replication-competent human parainfluenza virus type 3 (HPIV3) expressing EBOV
    glycoprotein GP (HPIV3/EboGP) and showed that it is immunogenic and protective against a high dose
    parenteral EBOV challenge [22].

               PROMISING TARGETS FOR DEVELOPMENT OF THERAPEUTICS

     An eight amino acid sequence (TELRTFSI) present in the carboxy terminal end (aa 577–584) of membrane-
anchored GP, the major structural protein of Ebola virus, was identified as an H-2k-specific murine cytotoxic T cell
epitope. Immunity can be stimulated by injecting irradiated Ebola virus encapsulated in liposomes containing lipid A
[16].

Contenu connexe

Tendances

Cell based assays (2010)
Cell based assays (2010)Cell based assays (2010)
Cell based assays (2010)jaayboy69
 
Pet mr poster-4-15-2012_v2[1]
Pet mr poster-4-15-2012_v2[1]Pet mr poster-4-15-2012_v2[1]
Pet mr poster-4-15-2012_v2[1]declen007
 
Pet mr poster-4-15-2012_v2[1]
Pet mr poster-4-15-2012_v2[1]Pet mr poster-4-15-2012_v2[1]
Pet mr poster-4-15-2012_v2[1]declen007
 
Ts111 Biomarker Solutions
Ts111 Biomarker SolutionsTs111 Biomarker Solutions
Ts111 Biomarker Solutionsgressero
 
2010 NTCA Conferece Submission w/ Abstract
2010 NTCA Conferece Submission w/ Abstract2010 NTCA Conferece Submission w/ Abstract
2010 NTCA Conferece Submission w/ Abstractecortes01
 
Evolution of biochemical diagnosis
Evolution of biochemical diagnosisEvolution of biochemical diagnosis
Evolution of biochemical diagnosisDr Heena Sharma
 
Asco 2012 abstract_45010_oral_final
Asco 2012 abstract_45010_oral_finalAsco 2012 abstract_45010_oral_final
Asco 2012 abstract_45010_oral_finalJames Hilbert
 
Breaking the Status Quo: Using Mass Spectrometry to detect Host Cell Proteins
Breaking the Status Quo: Using Mass Spectrometry to detect Host Cell ProteinsBreaking the Status Quo: Using Mass Spectrometry to detect Host Cell Proteins
Breaking the Status Quo: Using Mass Spectrometry to detect Host Cell ProteinsMerck Life Sciences
 

Tendances (11)

Plos
PlosPlos
Plos
 
Cell based assays (2010)
Cell based assays (2010)Cell based assays (2010)
Cell based assays (2010)
 
Pet mr poster-4-15-2012_v2[1]
Pet mr poster-4-15-2012_v2[1]Pet mr poster-4-15-2012_v2[1]
Pet mr poster-4-15-2012_v2[1]
 
Pet mr poster-4-15-2012_v2[1]
Pet mr poster-4-15-2012_v2[1]Pet mr poster-4-15-2012_v2[1]
Pet mr poster-4-15-2012_v2[1]
 
Ts111 Biomarker Solutions
Ts111 Biomarker SolutionsTs111 Biomarker Solutions
Ts111 Biomarker Solutions
 
2010 NTCA Conferece Submission w/ Abstract
2010 NTCA Conferece Submission w/ Abstract2010 NTCA Conferece Submission w/ Abstract
2010 NTCA Conferece Submission w/ Abstract
 
Evolution of biochemical diagnosis
Evolution of biochemical diagnosisEvolution of biochemical diagnosis
Evolution of biochemical diagnosis
 
Eccb
EccbEccb
Eccb
 
Asco 2012 abstract_45010_oral_final
Asco 2012 abstract_45010_oral_finalAsco 2012 abstract_45010_oral_final
Asco 2012 abstract_45010_oral_final
 
Getman Apr29 F I N A L
Getman  Apr29  F I N A LGetman  Apr29  F I N A L
Getman Apr29 F I N A L
 
Breaking the Status Quo: Using Mass Spectrometry to detect Host Cell Proteins
Breaking the Status Quo: Using Mass Spectrometry to detect Host Cell ProteinsBreaking the Status Quo: Using Mass Spectrometry to detect Host Cell Proteins
Breaking the Status Quo: Using Mass Spectrometry to detect Host Cell Proteins
 

En vedette

Najdobrijat email sa_godinata
Najdobrijat email sa_godinataNajdobrijat email sa_godinata
Najdobrijat email sa_godinataHristo Dundev
 
NPNH TIS Scenario Presentation
NPNH TIS Scenario PresentationNPNH TIS Scenario Presentation
NPNH TIS Scenario PresentationJeff Granger
 
Algonquin with notes
Algonquin with notesAlgonquin with notes
Algonquin with notessteevedoubt
 
Wireless industrial alarm adapter solution overview - q42015
Wireless industrial alarm adapter   solution overview - q42015Wireless industrial alarm adapter   solution overview - q42015
Wireless industrial alarm adapter solution overview - q42015stumanley
 
CS Education Event - University of Wollongong
CS Education Event - University of WollongongCS Education Event - University of Wollongong
CS Education Event - University of WollongongCollaborative Solutions
 
NOTAS PRIMER BIMESTRE 2012
NOTAS PRIMER BIMESTRE 2012NOTAS PRIMER BIMESTRE 2012
NOTAS PRIMER BIMESTRE 2012ipbingles
 
My personal learning network
My personal learning networkMy personal learning network
My personal learning networkHeli Vuorela
 
Create a stunning, mobile friendly business website with the divi theme
Create a stunning, mobile friendly business website with the divi themeCreate a stunning, mobile friendly business website with the divi theme
Create a stunning, mobile friendly business website with the divi themeMichelle Castillo
 
Design Mini Portfolio
Design Mini PortfolioDesign Mini Portfolio
Design Mini PortfolioEric Niehoff
 

En vedette (10)

Najdobrijat email sa_godinata
Najdobrijat email sa_godinataNajdobrijat email sa_godinata
Najdobrijat email sa_godinata
 
NPNH TIS Scenario Presentation
NPNH TIS Scenario PresentationNPNH TIS Scenario Presentation
NPNH TIS Scenario Presentation
 
10 spain
10 spain10 spain
10 spain
 
Algonquin with notes
Algonquin with notesAlgonquin with notes
Algonquin with notes
 
Wireless industrial alarm adapter solution overview - q42015
Wireless industrial alarm adapter   solution overview - q42015Wireless industrial alarm adapter   solution overview - q42015
Wireless industrial alarm adapter solution overview - q42015
 
CS Education Event - University of Wollongong
CS Education Event - University of WollongongCS Education Event - University of Wollongong
CS Education Event - University of Wollongong
 
NOTAS PRIMER BIMESTRE 2012
NOTAS PRIMER BIMESTRE 2012NOTAS PRIMER BIMESTRE 2012
NOTAS PRIMER BIMESTRE 2012
 
My personal learning network
My personal learning networkMy personal learning network
My personal learning network
 
Create a stunning, mobile friendly business website with the divi theme
Create a stunning, mobile friendly business website with the divi themeCreate a stunning, mobile friendly business website with the divi theme
Create a stunning, mobile friendly business website with the divi theme
 
Design Mini Portfolio
Design Mini PortfolioDesign Mini Portfolio
Design Mini Portfolio
 

Similaire à A theoretical discovery and development of an anti-ebola drug

Presentation on ICH guidelines Q5A (R1) and Q4B Annex 2 (R1)
Presentation on ICH guidelines Q5A (R1) and Q4B Annex 2 (R1)Presentation on ICH guidelines Q5A (R1) and Q4B Annex 2 (R1)
Presentation on ICH guidelines Q5A (R1) and Q4B Annex 2 (R1)HadiaNaz1
 
Rapid Methodologies for Biosafety Testing of Biologic Therapeutics
Rapid Methodologies for Biosafety Testing of Biologic TherapeuticsRapid Methodologies for Biosafety Testing of Biologic Therapeutics
Rapid Methodologies for Biosafety Testing of Biologic TherapeuticsMilliporeSigma
 
Rapid Methodologies for Biosafety Testing of Biologic Therapeutics
Rapid Methodologies for Biosafety Testing of Biologic TherapeuticsRapid Methodologies for Biosafety Testing of Biologic Therapeutics
Rapid Methodologies for Biosafety Testing of Biologic TherapeuticsMerck Life Sciences
 
Applications of monoclonal Antibodies.pdf
Applications of monoclonal Antibodies.pdfApplications of monoclonal Antibodies.pdf
Applications of monoclonal Antibodies.pdfgeneralinformation6
 
Tacking the menage of gram negative inf with novel bl bli.pptx
Tacking the menage of gram negative inf with novel bl bli.pptxTacking the menage of gram negative inf with novel bl bli.pptx
Tacking the menage of gram negative inf with novel bl bli.pptxaceforum
 
kansas_foodborne_illness_manual
kansas_foodborne_illness_manualkansas_foodborne_illness_manual
kansas_foodborne_illness_manualSiti Mastura
 
Ph.D. Defense 2021.pptx
Ph.D. Defense 2021.pptxPh.D. Defense 2021.pptx
Ph.D. Defense 2021.pptxHinaKhalid38
 
Dancey Clinical Trials Vancouver Dancey 20110302 Final.Ppt [Compatibility Mode]
Dancey Clinical Trials Vancouver Dancey 20110302 Final.Ppt [Compatibility Mode]Dancey Clinical Trials Vancouver Dancey 20110302 Final.Ppt [Compatibility Mode]
Dancey Clinical Trials Vancouver Dancey 20110302 Final.Ppt [Compatibility Mode]Warren Hamilton
 
Guideline on development, production, characterisation and specifications for...
Guideline on development, production, characterisation and specifications for...Guideline on development, production, characterisation and specifications for...
Guideline on development, production, characterisation and specifications for...National Institute of Biologics
 
viralsafetyevaluationofbiotechnologyproductsderivedfrom-210804180136.pptx
viralsafetyevaluationofbiotechnologyproductsderivedfrom-210804180136.pptxviralsafetyevaluationofbiotechnologyproductsderivedfrom-210804180136.pptx
viralsafetyevaluationofbiotechnologyproductsderivedfrom-210804180136.pptxRajakumari Rajendran
 
Viral Risk Mitigation - A Global Regulatory Perspective
Viral Risk Mitigation - A Global Regulatory PerspectiveViral Risk Mitigation - A Global Regulatory Perspective
Viral Risk Mitigation - A Global Regulatory PerspectiveMerck Life Sciences
 
Viral Risk Mitigation - A Global Regulatory Perspective
Viral Risk Mitigation - A Global Regulatory PerspectiveViral Risk Mitigation - A Global Regulatory Perspective
Viral Risk Mitigation - A Global Regulatory PerspectiveMilliporeSigma
 
Identification of antibiotic resistance genes in Klebsiella pneumoniae isolat...
Identification of antibiotic resistance genes in Klebsiella pneumoniae isolat...Identification of antibiotic resistance genes in Klebsiella pneumoniae isolat...
Identification of antibiotic resistance genes in Klebsiella pneumoniae isolat...QIAGEN
 
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...MilliporeSigma
 
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...Merck Life Sciences
 

Similaire à A theoretical discovery and development of an anti-ebola drug (20)

Presentation on ICH guidelines Q5A (R1) and Q4B Annex 2 (R1)
Presentation on ICH guidelines Q5A (R1) and Q4B Annex 2 (R1)Presentation on ICH guidelines Q5A (R1) and Q4B Annex 2 (R1)
Presentation on ICH guidelines Q5A (R1) and Q4B Annex 2 (R1)
 
Rapid Methodologies for Biosafety Testing of Biologic Therapeutics
Rapid Methodologies for Biosafety Testing of Biologic TherapeuticsRapid Methodologies for Biosafety Testing of Biologic Therapeutics
Rapid Methodologies for Biosafety Testing of Biologic Therapeutics
 
Rapid Methodologies for Biosafety Testing of Biologic Therapeutics
Rapid Methodologies for Biosafety Testing of Biologic TherapeuticsRapid Methodologies for Biosafety Testing of Biologic Therapeutics
Rapid Methodologies for Biosafety Testing of Biologic Therapeutics
 
Applications of monoclonal Antibodies.pdf
Applications of monoclonal Antibodies.pdfApplications of monoclonal Antibodies.pdf
Applications of monoclonal Antibodies.pdf
 
Tacking the menage of gram negative inf with novel bl bli.pptx
Tacking the menage of gram negative inf with novel bl bli.pptxTacking the menage of gram negative inf with novel bl bli.pptx
Tacking the menage of gram negative inf with novel bl bli.pptx
 
Evaluation of antibody drugs quality safety
Evaluation of antibody drugs quality safetyEvaluation of antibody drugs quality safety
Evaluation of antibody drugs quality safety
 
kansas_foodborne_illness_manual
kansas_foodborne_illness_manualkansas_foodborne_illness_manual
kansas_foodborne_illness_manual
 
Ph.D. Defense 2021.pptx
Ph.D. Defense 2021.pptxPh.D. Defense 2021.pptx
Ph.D. Defense 2021.pptx
 
abbas morovvati
abbas morovvatiabbas morovvati
abbas morovvati
 
Dancey Clinical Trials Vancouver Dancey 20110302 Final.Ppt [Compatibility Mode]
Dancey Clinical Trials Vancouver Dancey 20110302 Final.Ppt [Compatibility Mode]Dancey Clinical Trials Vancouver Dancey 20110302 Final.Ppt [Compatibility Mode]
Dancey Clinical Trials Vancouver Dancey 20110302 Final.Ppt [Compatibility Mode]
 
13593757-2.ppt
13593757-2.ppt13593757-2.ppt
13593757-2.ppt
 
Nanobodies
NanobodiesNanobodies
Nanobodies
 
Guideline on development, production, characterisation and specifications for...
Guideline on development, production, characterisation and specifications for...Guideline on development, production, characterisation and specifications for...
Guideline on development, production, characterisation and specifications for...
 
viralsafetyevaluationofbiotechnologyproductsderivedfrom-210804180136.pptx
viralsafetyevaluationofbiotechnologyproductsderivedfrom-210804180136.pptxviralsafetyevaluationofbiotechnologyproductsderivedfrom-210804180136.pptx
viralsafetyevaluationofbiotechnologyproductsderivedfrom-210804180136.pptx
 
Viral Risk Mitigation - A Global Regulatory Perspective
Viral Risk Mitigation - A Global Regulatory PerspectiveViral Risk Mitigation - A Global Regulatory Perspective
Viral Risk Mitigation - A Global Regulatory Perspective
 
Viral Risk Mitigation - A Global Regulatory Perspective
Viral Risk Mitigation - A Global Regulatory PerspectiveViral Risk Mitigation - A Global Regulatory Perspective
Viral Risk Mitigation - A Global Regulatory Perspective
 
Bio outsource hcp
Bio outsource hcpBio outsource hcp
Bio outsource hcp
 
Identification of antibiotic resistance genes in Klebsiella pneumoniae isolat...
Identification of antibiotic resistance genes in Klebsiella pneumoniae isolat...Identification of antibiotic resistance genes in Klebsiella pneumoniae isolat...
Identification of antibiotic resistance genes in Klebsiella pneumoniae isolat...
 
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
 
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
Viral Risk Mitigation Strategies: Key Considerations in the Prevention and De...
 

A theoretical discovery and development of an anti-ebola drug

  • 1. 1 2010 Discovery & Development of a First in Class Anti-Ebola Drug- EBONAVIR® Summary: Ebola hemorrhagic fever (Ebola HF) caused by the Ebola virus is a severe, often-fatal viral hemorrhagic disease in humans and nonhuman primates with a fatality rate of 50-83%. The present document details the discovery and development of a novel small molecule organo-pharmaceutical that acts as a potent inhibitor of VP35 - a critical protein involved in EBOV pathogenesis through early preclinical studies as well as clinical studies. It provides an insight into the research, documentation and regulatory requirements pertaining to the commercial launch of the above mentioned small molecule drug. GAYATHRI VIJAYAKUMAR MS Biotechnology, NYU-POLY 5/1/2010
  • 2. 2 CONTENTS 1) Introduction 5 a) Ebola Virus 5 b) Transmission 5 c) Symptoms 5 d) Diagnosis 6 e) Treatment 6 f) History 6 2) Current therapeutic advances for Ebola Hemorrhagic Fever 6 3) Promising targets for development of therapeutics 7 4) Target for drug development 8 a) Reason for selection of VP35 as a therapeutic target 8 b) Impairment of innate immunity 10 5) Medical hypothesis for developing an anti-Ebola therapeutic 12 6) Conclusion regarding medical hypothesis 14 7) Assays for HTS screening of potential anti-Ebola therapeutic 15 a) Luciferase Budding Assay 15 b) Immunocapture Assay 16 c) Cell based assay using recombinant GFP-ZEBOV 16 8) Luciferase Reporter based gene assay selected for HTS screening 18 a) Illustration of principle 21 9) Lead Compound 22 a) Selected Lead Compound candidate 24 10) Lead Optimization 25 a) Lead Optimization results 26 11) In vitro safety pharmacology profiling 28 a) In vitro SPP Profile results 33 12) Patent Application 35 13) Selection of appropriate animal model for efficacy evaluation of the selected NCE 36 a) Mouse model used for preclinical studies 38 b) Non-human primate model used for preclinical studies 40
  • 3. 3 14) Biomarkers 42 a) Disease Biomarkers 43 b) Surrogate Biomarker 45 c) Toxicity Biomarker 45 d) Target Biomarker 46 e) Mechanism Biomarker 46 f) Efficacy Biomarker 47 g) Translational Biomarker 47 h) Stratification Biomarker 47 15) Efficacy studies for Minimum Effective Dose 48 a) Minimum Effective Dose 49 16) Principal studies in animal models with NCT1087 49 a) Animal models in safety studies 50 b) Route of Administration of NCT1087 51 c) Safety Pharmacology Studies 51 d) Acute Toxicity Study 53 e) Repeated Dose Toxicity Study 53 f) Genetoxicity Study 55 g) Carcinogenicity Study 57 h) Reproduction Toxicity Study 57 i) Immunotoxicity Study 58 j) Phototoxicty Study 59 17) Maximum Tolerated Dose 60 a) Toxicity studies for determining the MTD 60 18) Repeated dose toxicity studies for 2 months 62 19) Estimation of the first human dose 64 20) IND Application 65 21) Phase I Clinical Trials 67 22) Phase II Clinical Trials 71 23) Phase III Clinical Trials 77 24) Conclusion 83 25) Route of Delivery, Administration Regimen & Dose Concentration 83 26) NDA Application 84 27) Life Cycle Management of NCT1087 or Ebonavir® 85 28) The Drug Development process 86
  • 4. 4 29) Preclinical & Requirements for IND, NDA &Clinical studies 88 30) CMC Requirements for IND, NDA & Clinical studies 89 31) Possible Emergence of Resistance to Ebonavir® 90 32) References 91
  • 5. 5 EBOLA HEMORRHAGIC FEVER Ebola hemorrhagic fever (Ebola HF) is a severe, often-fatal viral hemorrhagic Figure 1. [3] disease in humans and nonhuman primates (monkeys, gorillas, and chimpanzees) that has appeared sporadically since its initial recognition in the Democratic Republic of Congo in 1976 [1]. It is caused by the Ebola Virus which interferes with the interior endothelial cell lining of blood vessels and coagulation which leads to hypovolemic shock [2]. Destruction of endothelial surfaces is associated with disseminated intravascular coagulation, and this may contribute to the hemorrhagic manifestations [5]. Fatality rate differs from 50-83% [2]. [3] EBOLA VIRUS (shown in image [6]) Figure 2. [6] The Ebola virus belongs to the Filoviridae family (filovirus) and is comprised of five distinct species: Zaire, Sudan, Côte d’Ivoire, Bundibugyo and Reston. [4] The Zaire virus (ZEBOV) is the most lethal with an average case fatality rate of 83% [2]. Ebola virus has a non-segmented, negative-stranded, RNA genome containing 7 structural and regulatory genes [5]. The exact origin, locations, and natural habitat (known as the "natural reservoir") of Ebola virus remain unknown. Researchers believe that the virus is zoonotic (animal-borne) [4]. Electron microscopic image of an Ebola infected cell in [9] (Figure 3). TRANSMISSION 1) Direct contact with the blood and/or secretions of an infected person/ animal [1]. 2) Contact with objects, such as needles, that have been contaminated with infected secretions [1]. 3) Nosocomial transmission refers to the spread of a disease within a health- care setting, due to incorrect infection control precautions and adequate barrier nursing procedures [1, 4] Figure 3: [9] SYMPTOMS The incubation period for Ebola HF ranges from 2 to 21 days. The onset of illness is abrupt and is characterized by fever, headache, joint and muscle aches, sore throat, and weakness, followed by diarrhea, vomiting, and stomach pain. A rash, red eyes, hiccups and internal and external bleeding may be seen [1].
  • 6. 6 DIAGNOSIS Diagnosis is usually done with inactivated blood or saliva specimens to detect the viral antigen/antibody or genetic material [4]. They can also be isolated in cell cultures. Diagnostic laboratory tests such as ELISA, RT-PCR, IgM ELISA, Immunohistochemistry testing, Coagulation studies; Complete Blood Count, etc are usually performed [1,7]. TREATMENT There is no standard treatment for Ebola hemorrhagic fever. Treatment is primarily supportive and includes minimizing invasive procedures, balancing electrolytes, and, since patients are frequently dehydrated, replacing lost coagulation factors to help stop bleeding, maintaining oxygen and blood levels, and treating any complicating infections. Convalescent plasma, administration of an inhibitor of coagulation (rNAPc2), Morpholino antisense drugs have shown some promise in treatment for Ebola HF [2]. HISTORY History of Ebola virus outbreaks have been shown in the table to the side (Figure 4). Confirmed cases of Ebola HF have been reported in the Democratic Republic of the Congo, Gabon, Sudan, the Ivory Coast, Uganda, and the Republic of the Congo. [1] Because of its high morbidity, it is a potential agent for bioterrorism and since no approved vaccine or drugs are available it is classified as a biosafety level 4 agents as well as Category A bioterrorism agent by the CDC [2]. The last reported outbreak was in December 2008 in the Western Kasai province of the Democratic Republic of Congo [2]. Figure 4: From [8] CURRENT THERAPEUTIC ADVANCES FOR EBOLA HEMORRHAGIC FEVER To date, beyond supportive care, no effective treatments, therapies, or vaccines are approved to treat or prevent Ebola virus infections because natural immunity to this infection is difficult to find, plus there are no immune correlates in humans [10]. However, several drugs are under development as summarized:
  • 7. 7 • USAMRID investigated a drug called recombinant nematode anticoagulant protein C2 (rNAPC2) which blocks the harmful effects of tissue factors and targets the disease process. It showed reasonable success in infected rhesus monkeys [11]. • AVI BioPharma’s proprietary NEUGENE drug targets a key Ebola gene providing complete protection in mice when administered either before or after an otherwise lethal infection with Ebola virus. NEUGENE antisense compounds are synthetic polymers designed to match up perfectly with a specific gene or viral sequence, blocking the function of the target gene or virus [12]. • Phosphorodiamidate morpholino oligomers (PMO) are a class of uncharged single-stranded DNA analogs modified such that each subunit includes a phosphorodiamidate linkage and morpholinering. Data suggest that antisense PMO and P-PMO have the potential to control EBOV infection and are promising therapeutic candidates [13]. A combination of EBOV-specific PMOs targeting sequences of viral mRNAs for the viral proteins (VPs) VP24, VP35 has been used as both pre- and post- exposure therapeutic regimens in non-human primates [14]. • NanoViricides, Inc. has come out with broad-spectrum nanoviricides(TM) drug candidates that have been found to be highly effective in cell culture studies by USAMRID. These antivirals use biomimetic technology (i.e. they mimic the host cell features) [15]. • 3-Deazaneplanocin A, an analog of adenosine, is a potent inhibitor of Ebola virus replication. A single dose early in infection prevents progression of the disease in EBOV infected mice. Protective effect of the drug results from massively increased production of interferon-α [17]. • Cyanovirin-N (CV-N) which is an HIV inactivating protein has been found to have both in vitro and in vivo antiviral activity against the Zaire strain of the Ebola Virus by inhibiting the development of viral cytopathic effects (CPEs). It has been found to delay fatality in EBOV infected mice. It binds with considerable affinity to the Ebola envelope glycoprotein GP1,2 . [19]. • EBOV infected immunocompetent mice treated with the adenosine analogue carbocyclic 3-deazaadenosine protects it against fatality by blocking the cellular enzyme S-adenosyl-L -homocysteine hydrolase (SAH) [20]. • A vaccine based on recombinant vesicular stomatitis virus expressing the ZEBOV glycoprotein has been found to confer immunity against aerosol challenge in cynomolgus macaques which is likely to be the port of entry in a bioterrorist attack [21]. • Intranasal vaccine based on replication-competent human parainfluenza virus type 3 (HPIV3) expressing EBOV glycoprotein GP (HPIV3/EboGP) and showed that it is immunogenic and protective against a high dose parenteral EBOV challenge [22]. PROMISING TARGETS FOR DEVELOPMENT OF THERAPEUTICS An eight amino acid sequence (TELRTFSI) present in the carboxy terminal end (aa 577–584) of membrane- anchored GP, the major structural protein of Ebola virus, was identified as an H-2k-specific murine cytotoxic T cell epitope. Immunity can be stimulated by injecting irradiated Ebola virus encapsulated in liposomes containing lipid A [16].
  • 8. 8 Figure 5. Ebola Virus Potential targets • RNA is blocked by inhibiting the cellular enzyme SAH, indirectly [10]. resulting in reduced methylation of the 5′ cap of viral mRNA [17]. • NSecretary glycoproteins can be targeted by neutralizing monoclonal antibodies [18]. • VP24, VP30 and VP40 can induce protective immune responses [23]. • SCripps Research Institute found that VP35 is critical in allowing uncontrolled replication by fooling the human immune system [24]. In my opinion there definitely exists a medical need as 1) there are no approved therapies yet in the market, 2) it has a fatality of 50-80%, 3) it is a potential bioterrorism agent, but it is definitely not an impossible task which is clearly observable from all the latest developments in the field and all the possible targets available for developing a potential therapy. TARGET FOR DRUG DEVELOPEMENT The target that I have selected for development of a therapy against EBOV infection is the virion protein VP35. Reason for selection of VP35 as a therapeutic target The 18.9-kb RNA genome of EBOV is non-infectious and encodes seven structural proteins and one non-structural protein in the following order within the genome: 3′ non-coding region (leader), nucleoprotein (NP), virion protein 35 (VP35), VP40, glycoprotein (sGP and GP), VP30, VP24, RNA- dependant RNA-polymerase (L) protein and 5′ non-coding region (trailer) [27]. Mature EBOV particles form long filamentous rods with a uniform diameter of 80 nm and a mean length of 1250nm. Virus particles possess a central core, known as the ribonucleoprotein (RNP) complex that consists of NP, VP35, VP30, L and the viral RNA. This RNP complex is surrounded by a lipid envelope, with which the remaining proteins GP1,2, VP40 and VP24 are associated; these three proteins function as surface glycoprotein, major matrix protein and minor matrix protein, respectively as shown in the figure on the side. (Figure 6; [27]) Figure 6. [27] Three pathogenic mechanisms that underlie EBOV infection are vascular instability, coagulopathy and immunosupression (Figure 7; [27]. Besides the induction of cytopathic cytotoxicity in epithelial and endothelial cells (which perhaps explains the hemorrhagic manifestations characteristic of filovirus infection), destruction of the immune system is another significant feature of EBOV infection. The EBOV infects the mononuclear phagocyte and fibroblastic reticular systems associated with lymph nodes which maximizes immune
  • 9. 9 responses by activation of appropriate cytokine production and antigen trafficking. This results in inadequate development of immunity, thus playing an important role in pathogenesis [25]. Recent data shows that the EBOV virus VP35 protein, which plays an essential role in viral RNA synthesis, acts as a type 1 IFN antagonist in much the same way as the NS1 protein, another IFN antagonist, enhances the replicative ability of influenza virus, indicating that the presence of such a protein could be required for full expression of virulence [25]. Nucleoproteins VP-35 and -30 and RNA polymerase are required for RNA transcription and replication [25]. Another study found that the formation of nucleocapsid structure doesn’t occur in the absence of VP35, VP24 or NP [26]. Figure 7: Overview of the mechanisms that are involved in EBOV pathogenesis. taken from [27].
  • 10. 10 Impairment of innate immunity EBOV infection attacks the innate and adaptive immunity by • Inflammatory responses that are accompanied by substantial cytokine production [27]. • DCs, which have a crucial role in both innate and adaptive immunity, fail to fulfill their function after infection by not producing proinflammatory cytokines or express co-stimulatory molecules such as CD80 or CD86, are impaired in their ability to support T-cell proliferation and undergo anomalous maturation [27]. • Depletion of NK cells and lymphocytes [27]. • EBOV selectively suppresses responses to IFN-α and IFN-γ and the production of IFN-α in response to double-stranded RNA [27]. • Blockade of IFN signaling has an important role in the pathogenesis of EBOV infection in vivo [27]. VP35 blocks phosphorylation of the IFN-regulatory factor 3 (IRF3), which acts as a transcription factor for IFN production, whereas VP24 seems to block IFN signaling indicating that the block of IFN signaling might be due to inhibition of p38 phosphorylation, which is central in the mitogen-activated-protein-kinase (MAPK) p38 IFN-signaling pathway [27]. During virus infection, RNAi against the virus is activated by the production of virus-specific double-stranded RNAs (RNA interference (RNAi) mechanism). Similar to RNAi, the IFN pathway is triggered by cytoplasmic viral dsRNAs and acts as a sensitive and potent antiviral response that is involved in innate and subsequent adaptive immunity. To overcome this antiviral RNAi response, viruses encode RNA silencing suppressors (RSSs) and these include the influenza A virus NS1 (NS1), vaccinia virus E3L (E3L), hepatitis C virus Core, primate foamy virus type 1 (PFV-1) Tas, and the HIV-1 Tat proteins, as well as the adenovirus virus-associated RNAs I and II (VAI and VAII). Figure 8. RNA Interference Mechanism [28] Strikingly, all RSS proteins from mammalian viruses possess IFN or protein kinase R (PKR) antagonistic properties, suggesting that RNAi and other innate antiviral responses are interrelated. A study showed that Ebola virus (EBOV) VP35 protein is a potent RNAi suppressor that is functionally equivalent to the HIV-1 Tat RSS function [28].
  • 11. 11 Although the importance of antiviral RNAi responses in plants, nematodes, and insects is firmly established, it is still under debate whether RNAi has a similar function in mammalian cells. But all the same the above study demonstrates the importance of VP35 in helping EBOV in avoiding detection by the immune system [28]. Another study showed that the Ebola virus VP35 protein is a virus-encoded inhibitor of the type I IFN response. VP35 subsequently was shown to block double-stranded RNA- and virus-mediated induction of an IFN-stimulated response and to block double-stranded RNA- and virus-mediated induction of the IFN-β promoter. Type I IFN is synthesized in response to viral infection; double-stranded RNA (dsRNA) or viral infection activates latent transcription factors, including IRF-3 and NF-κB, resulting in the transcriptional up- regulation of type I IFN, IFN-α, and IFN-β, genes. Secreted type I IFNs signal through a common receptor, activating the JAK/STAT signaling pathway. This signaling stimulates transcription of IFN-sensitive genes, including a number that encode antiviral proteins, and leads to the induction of an antiviral state. Among the antiviral proteins induced in response to type I IFN are dsRNA-dependent protein kinase R (PKR), 2′,5′- oligoadenylate synthetase (OAS), and the Mx proteins [29]. This study validates that Ebola virus VP35 is therefore likely to inhibit induction of type I IFN in Ebola virus-infected cells and may be an important determinant of Ebola virus virulence in vivo [29]. Figure 9.This image shows the crystal structure of the Ebola virus protein VP35 bound to a molecule of double- stranded helical RNA (green). Two VP35 molecules come together to mask the end of the double-stranded RNA helix from detection by the immune system. The interface between the two VP35s (white) may provide new targets for drugs to counteract the virus. [30]
  • 12. 12 Medical Hypothesis for developing an anti-EBOV Therapeutic The Ebola VP35 protein is multifunctional, acting as a component of the viral RNA polymerase complex, a viral assembly factor, and an inhibitor of host interferon (IFN) production [31]. All the studies above have established the fact the VP35 is absolutely essential for Ebola virus pathogenesis as well virulence. Therefore I conclude that it makes an excellent target for development of a therapeutic. From whatever scientific publications I have read so far, I think that there are two ways you can tackle this problem. 1. Using antisense Phosphorodiamidate morpholino oligomers They are a class of uncharged single-stranded DNA analogs modified such that each subunit includes a phosphorodiamidate linkage and morpholine ring [13]. Data suggests that antisense PMO and P-PMO (PMO conjugated to arginine-rich cell-penetrating peptide) have the potential to control EBOV infection by binding to the translation start site region of EBOV VP35 positive-sense RNA and generating sequence-specific and time- and dose-dependent inhibition of EBOV amplification in cell culture [13]. AVI BioPharma’s proprietary NEUGENE drug uses a combination of EBOV-specific PMOs targeting sequences of viral mRNAs for the viral proteins (VPs) VP24, VP35, and RNA polymerase L and found it to have reasonably good success in EBOV-infected rodents and primates. It is currently under clinical trials [14]. 2. Targeting C-terminal of VP35 IID Mutation of select basic residues within the C-terminal half of VP35 inhibits its dsRNA-binding activity, impairs VP35-mediated IFN antagonism, and attenuates EBOV growth in vitro and in vivo [31]. A functional VP35 is required for efficient viral replication and pathogenesis; knockdown of VP35 leads to reduced viral amplification and reduced lethality in infected mice [31]. VP35 contains an N-terminal coiled-coil domain required for its oligomerization (required for viral replication) and a C-terminal dsRNA-binding region (blocks signaling that triggers IFN process) [31]. Data suggest that the dsRNA binding activity mediated by the C terminus of VP35 is critical for viral suppression of innate immunity and for virulence. There are 2 basic patches that are highly conserved among members of the Filoviridae family (identical among EBOV isolates). Biochemical and NMR-based structural analyses show that one patch contains residues that are required for dsRNA binding and IFN inhibition, whereas the functional significance of the other basic patch remains unknown [31]. Therefore a compound that would target the conserved region in the C-terminal of VP35 IID which are critical for dsRNA binding may result in impaired IFN suppression and yield attenuated viruses in vivo because of reduced dsRNA binding ([31]; see Figures 10-12 below).
  • 13. 13 Figure 10. Crystal structure of VP35 C-terminal IFN-inhibitory domain (IID) reveals a fold that binds dsRNA. (A) Domain organization of VP35. (B) Ribbon representation of VP35 IID. Secondary structural elements that form the α-helical subdomain (orange) and the β-sheet subdomain (yellow). (C) Topology and delimiting sequence markers of VP35 IID [31]. Figure 11. Polymerase cofactor VP35, Key word 3FKE [32]. Figure 12. Structures of the ] Ebola VP35 IID dsRNA binding domain [31].
  • 14. 14 CONCLUSION A molecule or compound that inhibits VP35 of the Ebola virus will inhibit binding of the virus-specific dsRNA produced by the host cell as part of RNA interference (RNAi) mechanism thus activating both double-stranded RNA- and virus-mediated induction of an IFN-stimulated response as well as double-stranded RNA- and virus- mediated induction of the IFN-β promoter [28][29]. This activated latent transcription factors, including IRF-3 and NF-κB, resulting in the transcriptional up-regulation of type I IFN, IFN-α, and IFN-β, genes. Secreted type I IFNs signal through a common receptor, activating the JAK/STAT signaling pathway. This signaling stimulates transcription of IFN-sensitive genes, including a number that encode antiviral proteins, and leads to the induction of an antiviral state [29]. This leads to up regulation of the innate immune system. Virus-specific dsRNAs on the other hand are processed into small interfering RNAs (siRNAs; a 21- nucleotide dsRNA duplex) by the RNAse III–like endonuclease-denoted Dicer. Subsequently, one strand of the siRNA duplex, the guide-strand, is incorporated into the RNA-induced silencing complex (RISC) to target viral mRNAs bearing complementary sequences for destruction. [28] Thus I conclude that a compound that inhibits VP35 leads to activation and up regulation of the innate and adaptive immune system. In addition inhibition of VP35 exposes viral mRNAs to host cell antiviral response. This leads to reduction in the viral load, and thus will stop progression of the infection. Why is my target better? I think that this target is better than the others • As scientific evidence doesn’t prove that inhibition of other viral targets will elicit the same degree of recovery as VP35 inhibition. • VP35 is very integral to EBOV replication and pathogenesis. Figure 13: Model for VP35-mediated • C-terminal of VP35 IID is conserved among members IFN inhibition and immune of the Filoviridae family (identical among EBOV isolates). suppression [31].
  • 15. 15 ASSAY FOR HTS SCREENING OF POTENTIAL ANTI-EBOLA THERAPEUTICS There are a few assays that are in use for Ebola research. It is listed as follows along with their principle and a brief description of their methodology. LUCIFERASE BUDDING ASSAY It is a functional budding assay that has been developed based on the ability of VP40 matrix protein of EBOV in bringing about the budding of virus-like particles (VLP) from mammalian cells. This well-defined assay has been modified for potential use in a high-throughput format in which the detection and quantification of firefly luciferase protein in VLPs represents a direct measure of VP40 budding efficiency. Luciferase was found to be incorporated into budding VP40 VLPs. In contrast, when luciferase is co-expressed with a budding deficient mutant of VP40, luciferase levels in the VLP fraction decrease significantly. Although VP40 does not require the presence of additional viral proteins to achieve VLP egress, the contribution of glycoprotein (GP), nucleoprotein (NP), VP35 and VP24 to budding has been studied. BRIEF DESCRIPTION OF THE METHODOLOGY Human 293 T cells are grown in DMEM supplemented with 10% FCS and 1% penicillin-streptomycin at 5% CO2at 37OC. pCAGGS-luciferase vectors are constructed by insertion of luciferase into the SacI and NsiI sites of the pCAGGS vector. Subsequently, VP40 is joined to the C-terminal end of luciferase by insertion into the NsiI and XhoI sites of pCAGGS- luciferase. One hundred millimeter plates of 293T cells are transfected with 10µg of each plasmid DNA using Lipofectamine in OptiMem. Cells are metabolically labeled 24h post-transfection with Met-Cys. Six hours later, media was harvested, clarified, and layered over a 20% sucrose cushion in STE buffer, then centrifuged. The pellet was resuspended in Figure 14. Luciferase activity in VLPs correlates STE buffer and lysed with radioimmunoprecipitation assay with expression and budding efficiency of VP40. Human 293T cells were transfected with pCAGGS + (RIPA) buffer. Cells were washed twice with PBS and then luciferase, VP40 + luciferase, VP40 + GP + lysed in RIPA buffer. Both cells and VLP lysates were luciferase, or VP40d-PT/PY + luciferase. VLPs were isolated and then combined with luciferase reagent. immunoprecipitated with the appropriate antibodies and Relative light units (RLU) were measured using a analyzed by SDS polyacrylamide gel electrophoresis (PAGE) luminometer. The RLU value of luciferase + pCAGGS was normalized to one [33]. [33]. Subsequently Protease Protection assay and VLP Luciferase Assay was performed [33],
  • 16. 16 IMMUNOCAPTURE ASSAY It is basically a rapid ELISA assay that detects both Ebola virus and EBOV-like particles (VLPs) directly from cell- culture supernatants with the VP40 matrix protein serving as antigen. This assay can be used in surrogate models in non-biocontainment environment, facilitating both basic research on the mechanism of EBOV assembly and budding as well as drug-discovery research. Such an assay could also replace or be complementary to the conventional viral plaque assays for virus detection, which require more than a week to accomplish [34]. BRIEF DESCRIPTION OF METHODOLOGY cDNA for EBOV Zaire NP, VP30, VP35, and VP24 was cloned into pWRG7077 vector. Polyclonal anti-VP40 antibody was generated by immunizing rabbits with a peptide corresponding to the N-terminal 15 amino acids of VP40 protein. Sera from convalescent EBOV-infected guinea pigs were used as the polyclonal anti-EBOV antibody. Bacterial expression of EBOV VP40 was done by using a pET16b-VP40 clone to introduce a His6-fusion at the N-terminus in E.coli. Subsequently purification and extraction of EBOV VP40 was done from the cell lysate. Fractions were analyzed by SDS-PAGE and appropriate fractions were pooled [34]. HB 96-well plates were coated with AE11 monoclonal mouse anti-VP40 antibodies. The plates were washed with PBST. PBST containing 5% milk was added to block non-specific binding sites. After binding, plates were washed with PBST. Cellular extracts or culture supernatants were loaded with 1% Figure 15. Establishing an EBOV VP40 ELISA. (A) Coomassie staining of purified VP40 protein (lane 1). milk PBST. The samples were removed and the wells washed Immunoblotting of VP40 protein gel using anti-VP40 three times for 5 min. Rabbit polyclonal anti-VP40 antibodies antibodies showing the monomeric and oligomeric forms of VP40 (lane 2). (B) Standard curve for VP40 were added in PBST/5% milk and incubated for 1 h. The plate ELISA generated using purified VP40 protein. (C) was washed again and anti-rabbit HRP-conjugated antibodies Standard curve for VP40 ELISA using inactivated were added in PBST for 30 min. The plate was washed with EBOV lysed in 1.5% Triton X100. Capture antibodies were monoclonal mouse anti-VP40 and detection PBST and PBS. HRP substrate was added to each well and antibodies rabbit anti-VP40 polyclonal antisera [2]. absorbance at 650 nm was read [34]. CELL-BASED ASSAY USING RECOMBINANT GFP-ZEBOV In this assay the GFP acts as a receptor for virus replication. As such this assay can be used to screen compounds that inhibit replication of the virus [35]
  • 17. 17 BRIEF DESCRIPTION OF METHODOLOGY Confluent Vero E6 cells (96-well plate format) were pretreated with compound (drug) diluted in cell-culture medium for 18h. Cells were then infected with GFP-ZEBOV. Compound was reapplied to cells after infection by adding media containing the same pretreatment concentration. Cultures were incubated for 48h before fixation. After nuclear staining (Hoechst dye), viral infection was determined using a high content imaging system, Discovery 1, which compiles GFP fluorescence data from approximately 25,000 cells per well. This procedure identifies the total number of adherent cells, and fraction of infected cells, thus providing an immediate assessment of efficacy and Figure. 16: FGI-106 blocks Ebola virus infection. (A) Vero E6 cells were infected with Zaire-Ebola virus toxicity. Validation of the results was done by means of Virus (ZEBOV) for 72 h in the presence of 0–4 µM FGI-106. Yield Reduction Assay. [35] Antiviral efficacy was evaluated using plaque assays of infectious Ebola particles to assess the number of Another paper which utilizes the same principle engineered an plaque forming units per unit volume (pfu/mL). (B) The number of viable Vero E6 cells was assessed EboZ-eGFP by inserting eGFP open reading frame preceded following 72 h incubation in the presence of the by a 90-nucleotide fragment containing an authentic indicated concentrations of FGI-106. (Drug) [35] transcription stop sequence, to direct the termination of the NP mRNA, and a transcription start sequence to direct the initiation of the eGFP message into the cloned BsiW1 restriction site present in the NP 3′ UTR. The “wild-type like” virus was termed EboZ-BsiW1. As a proof of principle for using the EboZ-eGFP virus to rapidly screen antiviral compounds, consensus interferon alpha (conIFN-α), a compound with known EboV antiviral properties, was used at multiple concentrations to treat Vero E6 cells infected with either EboZ- eGFP virus or a control Zaire ebolavirus. The infected cells were monitored for evidence of virus replication by either fluorescence microscopy (for EboZ-eGFP virus) or a previously published CPE-based assay [37]. Figure 17: (A) Time course of Vero E6 cells infected with EboZ- eGFP, EboZ-BsiW1, or EboZ-wt. Vero E6 cells were infected at a MOI of two and virus (B) Fluorescent microscopy of human PBMCs either mock-infected or infected with EboZ-eGFP and analyzed at the indicated times post-infection. (C) Electron micrograph of a human macrophage infected with EboZ-eGFP virus [37].
  • 18. 18 PLAQUE ASSAY FOR EBOLA VIR PLAQUE ASSAY Confluent Vero cells are infected with an EBOV strain. After absorption of the viral innoculum for 1h, 30 ml of EBM containing 5% FBS, antibiotics and HEPES buffer are incubated in flasks for 10 days. Harvested medium is centrifuged and the supernatant fluid is made into dilutions using L-15 medium and inoculated into monolayers of cell lines being tested. After various incubation periods, cell monolayers are stained using an agarose overlay containing neutral red. Plates are then examined for plaques 24 and 48 hours after adding the stain overlay. The viral origin of the plaques is verified by inoculating one of the plaques mixed with L-15 medium into suckling mice. After death of the mice, the livers are removed, processed for virus isolation, and examined for viral antigens with the IFA procedure. Liver tissue collected from mice which had died post- inoculation will show EV-specific fluorescence by IFA and from this the PFUs can be counted [36]. Figure 18: From [36]. THE ASSAY THAT IS SELECTED FOR HTS SCREENING-A LUCIFERASE REPORTER BASED GENE ASSAY PRINCIPLE: The Ebola VP35 protein is multifunctional, acting as a component of the viral RNA polymerase complex, a viral assembly factor, and an inhibitor of host interferon (IFN) production [31]. The EBOV virus VP35 protein, which plays an essential role in viral RNA synthesis, acts as a type 1 IFN antagonist by blocking phosphorylation of the IFN-regulatory factor 3 (IRF3), which acts as a transcription factor for IFN production and turns on a number of anti-viral genes [25, 27]. Figure19: From [31]
  • 19. 19 RF-3 is activated early during virus infection by the presence of dsRNA molecules within the cell, and it acts in conjunction with other transcription factors to turn on the expression of immediate early antiviral genes such as IFN- α/β, IL-15, ISG15, and ISG56. It was found that a C-terminal basic amino acid motif in VP35 is required for inhibition of ISG56 reporter gene expression as well as IFN-β production [39]. Previous studies showed that infection of 293 T cells with Sendai virus (SeV), a potent inducer of the type I interferon system, activates the IFN-responsive promotor ISG56 in an IRF-3-dependent manner, and this activation is significantly inhibited by the presence of VP35 from Ebola virus. In this transfection-based reporter gene assay, 293T cells were transfected with 1. ISG56-firefly luciferase plasmid (pISG56/pGL3b) 2. a VP35 expression plasmid (pCEZ-VP35), and 3. a constitutively expressed Renilla luciferase plasmid (phRG-TK) [39]. After induction of the interferon pathway by SeV infection, firefly luciferase activity was measured and normalized relative to Renilla expression. SeV infection induced approximately a 10-fold increase in firefly luciferase activity in control wells, and this expression was significantly inhibited by the presence of VP35 but not the ebolavirus NP protein [39]. So if VP35 is inhibited by a particular compound in EBOV-infected cells then fluorescence emitted will be the same as that of SeV infected cells. Figure 20: The effect of C-terminal deletion mutations on IFN-antagonism mediated by VP35. (A) SeV induction of ISG56- luciferase expression in the presence of an empty vector control plasmid, ebolavirus VP35, or ebolavirus NP. (B) Schematic illustration of the 5 C-terminal deletion constructs. Full-length VP35 is illustrated on top. Stop codons were inserted at the indicated residues. (C) Evaluation of each deletion construct in the ISG56-luciferase reporter gene assay. Error bars indicate the average of each construct tested in duplicate; results shown here are representative of three independent experiments [39]. This study found that the 40 amino acids of the C Terminal is essential for the inhibitory activity of VP35 as demonstrated by an experiment in which five C-terminal deletion constructs were generated by inserting a stop
  • 20. 20 codon at the indicated position using the overlap-extension PCR method. When each of these constructs was tested in the reporter gene assay, all five truncated proteins lost the ability to block ISG56 activation. The largest of the proteins, R300T, has a stop codon inserted at arginine 300, resulting in a deletion of the C-terminal 40 amino acids [39]. METHODOLOGY: [39] 293T cells were transfected in suspension by the calcium phosphate method with the following: 2 µg of pCEZ-VP35 150 ng of pISG56/pGL3b 150 ng of phRG-TK 4.4 µg of salmon testes carrier DNA Combined plasmid DNA is diluted to 110µl in sterile water. 125 µl of 2 × HEPES-buffered saline was added, followed by 15.5 µl of 2 M CaCl2 added dropwise with gentle shaking. The DNA mixture was allowed to sit at room temperature for 30 min before using it to resuspend a cell pellet containing approximately 0.5–1.0 × 106 cells. The mixture of cells and DNA complexes was incubated at room temperature for 15 min, at which time 2.3 ml of DMEM/10% FBS/Pen-Strep was added, and the cells were transferred to 6-well plates for incubation at 37 ° C. Twenty-four hours following transfection, the cells were infected with 1024 HAU of Sendai virus per well. For each well, 50 µl of virus was diluted in PBS/BA (Dulbecco's PBS w/Ca2+/Mg2++, 0.2% Bovine Albumin, Penicillin/Streptomycin) to a total volume of 600 µl. The media from each well was removed, and the cells were washed once with PBS before adding 600 µl of virus mixture. The plates were incubated at room temperature for 1 h, at which time the inoculum was removed and replaced with 2.3 ml of DMEM containing 0.2% BA and Penicillin-streptomycin. After 18–20 h at 37 ° the cells were harvested i n 500 µl of passive lysis buffer, and 20 µl of each sample was C, tested in the Dual-luciferase assay according to the manufacturer's instruction. The firefly luciferase values were normalized to the Renilla values for each sample. The relative fold-induction of reporter gene expression was assessed by comparing SeV-infected wells versus uninfected control wells. This methodology can be adapted for HTS screening of a library for compounds that effectively inhibits the activity of VP35, as this assay specifically measures the inhibition of vp35 with regards to the particular pathway that it inhibits.
  • 21. 21 Illustration of the principle for the luciferase-based assay (Figure 21) Infected with Sendai virus. Image Taken from [40] The firefly luciferase values are normalized to the Renilla values for each sample. The relative fold-induction of reporter gene expression is assessed by comparing SeV-infected wells versus uninfected control wells. SeV induction of ISG56-luciferase expression [39]
  • 22. 22 LEAD COMPOUND Several studies have found that the Influenza virus protein NS1 has striking similarity in terms of function and structure of the effecter domains. Like VP35, NS1 induces antiviral RNAi response in the infected host cells. VP35 has a dsRNA-binding motif with high similarity to the dsRNA-binding domain of the NS1 protein [41]. Additionally, the NS1 protein is able to prevent viral activation of IRF-3 and NF-κB, central components in promoting type I IFN synthesis [38]. The NS1 protein of influenza A virus blocks both the production of type I IFN and the activation of the IFN-induced antiviral proteins PKR and OAS [41]. A bioinformatics study identified an 8-residue motif in VP35 that has 75% sequence identity to the influenza NS1A protein, including basic residues essential for binding of dsRNA by NS1 [38]. Mutation of these residues in both NS1 and VP35 inhibited their IFN antagonist function [38]. Therefore like NS1, VP35 inhibits phosphorylation, activation, and nuclear localization of IRF-3 and inhibits viral- and dsRNA-induced expression of the IFNβ gene. VP35 also inhibits activation of the cellular antiviral kinase RNA-dependent protein kinase (PKR) and activation of the RNAi pathway [38]. Bioinformatic comparison revealed that a 9-amino acid region (a.a. 304–312) in the C-Terminal region shows remarkable similarity to part of the N-terminal RNA-binding domain of the influenza A virus NS1 protein (a.a. 36–46). No other known viral interferon antagonists showed any similarity with VP35 [39]. Figure 22: A short C-terminal motif within VP35 displays high identity with part of the N-terminal Alanine substitution studies found that out of the three basic RNA-binding domain of influenza virus NS1 protein. amino acids changed to alanine, the R312A mutation resulted Part of the N-terminal RNA binding domain of NS1 from influenza A/WSN/33 (M12597) compared to the in the greatest disruption to the inhibitory capacity of VP35. C-terminal domain from Zaire ebolavirus Mayinga Thus, of the three amino acids studied here, R312 appeared VP35 (AF272001). Asterisks below NS1 sequence to be the most important amino acid for inhibiting ISG56 indicate residues that have been shown to be responsible for RNA-binding. Asterisks above VP35 promotor activation (IFN pathway) [39]. sequence are the basic amino acids changed to alanine residues in this study [39]. Double mutations in K309A and R312A completely abolished the inhibitory activity [39]. Within the 73-amino acid N- terminal RNA-binding domain, substitution of the highly conserved residues R38 and K41 with alanines prevented RNA binding, destroyed some of the ability to antagonize IFN [39]. From all of these findings we can safely conclude that NS1 and VP35 are very similar and therefore a drug that can inhibit NS1 could also theoretically have some degree of inhibitory activity against VP35. There is currently no approved NS1 protein inhibitor in the market, but I was able to find one that had been applied for patent ship. So, technically this inhibitor should also be able to have some activity against VP35.
  • 23. 23 In this invention the inventors have identified small “organopharmaceuticals” that inhibit the activity of NS1 protein, but do not affect NS1 protein gene expression. NS1 protein inhibitors typically have a molecular weight of about 500g/mol or less [42]. The inventors screened a number of compounds of which Compound 8.3 was found to be the most potent with Compound 8.6 being the second most effective in the infection assay (Figure 23 and 24). The library that they used for high-throughput screening constituted about 200,000 small molecules which is owned by the University of Texas, Southwestern [42]. Figure 23: Data taken from [42]. The table depicts the compounds that were screened. Compound 8 was the most effective.
  • 24. 24 Figure 24: Data taken from [42]. Compound 8.3 was found to be the most potent followed by Compound 8.6 SELECTED LEAD COMPOUND CANDIDATE Figure 25: My lead compound This is the compound that I have selected as the lead compound. Since it shows efficacy against NS1 and since NS1 and VP35 domains are strikingly similar, I hypothesize that this organopharmaceutical would also be effective in inhibiting VP35 and thus allowing the initiation of the IFN pathway and subsequent activation of various other immune responses thus stopping the progression of the infection.
  • 25. 25 LEAD OPTIMIZATION The 3D structure of the target VP35 Interferon Inhibitory Domain is given below. The yellow portion indicates the 40 amino acids essential for VP35 activity-residues from 300-340 [39]. The red portion indicates the three basic residues – ARG305, LYS109 and ARG312, which are critical for VP35 to effectively block activation of an IRF-3 responsive promoter [39]. The following image (Figure 26) is taken from the RCSB Protein Database; Key word 3FKE [32]. The following is a 3D structure of VP35 inhibitory domain (shown in pink, red, blue and green showing the three essential residues) complexed with dsRNA (shown in cyan). The PDB file is taken from RCSB Protein database; Keyword- 3L25. [43] Figure 27:
  • 26. 26 LEAD OPTIMIZATION & CREATION OF NCEs BY SAR STUDIES FOR AN ANTI- LEAD OPTIMIZATION RESULTS The lead compound that I had selected is given below Table 1: Structural modifications of the lead:
  • 27. 27 Since lysine and arginine are basic amino acids I have included a lot of Hydrogen bond forming groups in the structures. Table 2: The R1 and R2 groups of the different structures are given below: COMPOUND R1 R2 LEAD -piperdine -(Ch3)5 - COOH NCE (1) -piperdine - CH3-C4H9NO NCE (2) -NO-OH -CH3-C4H9NO NCE (3) -NO-OH -(Ch3)5 - COOH NCE (4) - -CH3-COOH NCE (5) -COOH -CH3-COOH NCE (6) -NO-OH -CH3-NO-OH NCE (7) -NO-OH -CH3-CH3 NCE (8) - -CH3-NO-OH NCE (9) -(CH3)3-C4H9NO -CH3-NO-OH NCE (10) -NO-OH -methylmaleic anhydride Figure 28 Figure 29 In the figure to the left I have shown the side chains of the three residue protruding outside into the active site [32]. In the figure to the right I have shown the amino acids in the active site and how they are configured/oriented. I got this image by using the RCSB PDB Ligand Explorer 3.7 for VP35 (Key word-3L25).
  • 28. 28 IN VITRO SAFETY PHARMACOLOGY PROFILING Anti-viral drugs are difficult to develop and the two broad-spectrums anti-viral compounds in the market- Ribavirin and Interferon alpha have several side effects such as anemia, headache, irritability, anxiety, alopecia, itchiness, insomnia, arthralgia, myalgia, anorexia, neutropenia, nausea, vomiting, fever, chills and fatigue [44]. My drug is going to be administered by intravenous injection; therefore I can hypothesize that those problems of nausea and vomiting can be avoided. The adverse effects of my lead compound could therefore be associated with hematology, musculoskeletal, dermatology and to certain extent maybe neurology as well. Therefore the in vitro SPP assays that are used to screen the NCEs are as follows: • Luciferase Reporter based gene assay [39] – Measure the binding affinity This assay measures the inhibition of VP35 by NCE compounds in Sendai virus (SeV) infected 293 T cells, a potent inducer of the type I interferon system, which activates the IFN-responsive promotor ISG56 in an IRF-3-dependent manner, and this activation is significantly inhibited by the presence of VP35 from Ebola virus. In this transfection-based reporter gene assay, 293T cells were transfected with ISG56-firefly luciferase plasmid (pISG56/pGL3b) Figure 30: SeV induction of ISG56- • A VP35 expression plasmid (pCEZ-VP35), and luciferase expression in the presence of an empty vector control plasmid, • a constitutively expressed Renilla luciferase plasmid (phRG-TK) ebolaviruss VP35, OR ebolavirus NP.[39] After induction of the interferon pathway by SeV infection, firefly luciferase activity was measured and normalized relative to Renilla expression. SeV infection induced approximately a 10-fold increase in firefly luciferase activity in control wells, and this expression was significantly inhibited by the presence of VP35 but not the ebolavirus NP protein. So if VP35 is inhibited by a particular compound in EBOV-infected cells then fluorescence emitted will be the same as that of SeV infected cells.
  • 29. 29 • Plaque assay for Ebola virus [36] – Confirmatory assay for VP35 inhibtion by NCE This assay is also referred to as the Neutral Red Uptake Assay. Confluent Vero cells are infected with an EBOV strain. After absorption of the viral innoculum for 1h, 30 ml of EBM containing 5% FBS, antibiotics and HEPES buffer are incubated in flasks for 10 days. Harvested medium is centrifuged and the supernatant fluid is made into dilutions using L-15 medium and inoculated into monolayers of cell lines being tested. They are then treated with the different NCEs possibly in different concentrations as well. After various incubation periods, cell monolayers are stained using an agarose overlay containing neutral red. Plates are then examined for plaques 24 and 48 hours after adding the stain overlay. The viral origin of the plaques is verified by inoculating one of the plaques mixed with L- 15 medium into suckling mice. After death of the mice, the livers are removed, processed for virus isolation, and examined for viral antigens with the IFA procedure. Liver tissue collected from mice which had died postinoculation will show EV-specific fluorescence by IFA and from this the PFUs can be counted. Figure 31: From [36] • In vitro absorption, distribution, metabolism and excretion (ADME) assays: ADME-Tox assays define or investigate a lead candidate’s pharmacokinetic and metabolic stability/toxicity. They help in flagging adverse clinical effects at low cost and can prevent high attrition rates during clinical trials.[50] The ADME-Tox assays that I have selected for screening my NCEs for are as follows: Cytochrome P450 (CYP450) inhibition [45][46] Anti-viral drugs have been shown to have an effect on this family of enzymes [50]. It is a luminescence based method to measure cytochrome P450 (CYP) activity. The assay is designed to measure the activities of CYP enzymes and test the effects of the NCEs on CYP activitiesThe CYP enzyme substrate which are usually derivatives of luciferin is converted by CYP enzymes to a luciferin product that is detected in a second reaction with the Luciferin Detection Reagent. The amount of light produced in the second reaction is proportional to CYP activity.
  • 30. 30 A. P450-Glo™ Substrate (proluciferin) CYP Enzyme O B. R N N O Figure 32: taken from [46] HO S Luciferin Detection Reagent Light Human Liver Microsome Assay [47] HLM assays are carried for investigating whether the lead compounds have any effect on as well as for measuring UDP glucuronosyltransferase (UGT) activity. The UGT family of enzymes are involved in the metabolism of various compounds in the body by transferring a hydrophilic glucuronic acid moiety to their substrates, rendering them more water soluble and suitable for excretion. To measure UGT activity, two glucuronidation reactions are set up in parallel. Both reactions contain a source of UGT and the proluciferin substrate, but only one of them contains the uridine 5´-diphosphoglucuronic acid (UDPGA) cofactor. During the incubation period with the UGT enzyme or enzymes, a portion of the proluciferin substrate is glucuronidated in the reaction containing UDPGA. None of the proluciferin is glucuronidated in the reaction lacking UDPGA. In the second step of the assay, incubation of the proluciferin substrates with D- Cysteine in the Luciferin Detection Reagent results in conversion of the proluciferins into luciferin molecules. Luciferin produced from the unmodified proluciferin will give light in the Luciferin Detection Reagent, but the luciferin produced from the glucuronidated proluciferin will not give light. Therefore, the decrease in light output when comparing the plus-UDPGA reaction to the minus- UDPGA reaction is proportional to the glucuronidation activity in the first step [47].
  • 31. 31 OH O N UGT HO O N C N C N HO S HO O S UDPGA UDP OH Plus D-Cysteine OH O O O HO N N O N OH N OH HO O S S S S HO OH Figure 33 taken from [47] Plus Luciferin Detection Reagent Light No Light 7984MA Figure 33:. Conversion of UGT Multienzyme Substrate by UGT enzymes. UGT enzymes attach a glucuronic acid moiety to the proluciferin substrate. During the detection step, the proluciferin is simultaneously converted to a luciferin by cyclization with D-Cysteine. Luciferase uses the luciferin analog of the initial substrate to produce light but does not produce light with the glucuronidated luciferin. Light output is inversely proportional to UGT enzymatic activity.[47] hERG Potassium Channel Screening The protein product of the human ether-a-go-go gene (hERG) is a potassium channel that when inhibited may lead to cardiac arrhythmia [48]. Prediction of the hERG affinity value, QT prolongation, and action potential parameters are recommended as test criteria by the FDA [49]. RLB assays are commonly used as primary screens for hERG inhibition, whereas patch clamp analysis remains the gold standard [50]. Planar patch clamp is the one that I have selected for Figure 34:The cell-attached patch clamp uses measuring hERG liability because it allows for a multiwall a micropipette attached to the cell membrane to format and automation, which is much faster and less labor allow recording from a single ion channel [51]. intensive [49]. Patch clamp technique which is a refinement of the voltage clamp allows the study of ion channels in cells. Patch clamp recording uses, as an electrode, a glass micropipette , a size enclosing a membrane surface area or "patch" that often contains just one or a few ion channel molecules. The interior of the
  • 32. 32 pipette is filled with a solution matching the ionic composition of the bath solution, as in the case of cell- attached recording, or the cytoplasm for whole-cell recording. A chlorided silver wire is placed in contact with this solution and conducts electrical current to the amplifier. The investigator can change the composition of this solution or add drugs to study the ion channels under different conditions. The micropipette is pressed against a cell membrane and suction is applied to assist in the formation of a high resistance seal between the glass and the cell membrane [51]. In case Planar patch clamp assay, cell suspension (containing a single cell randomly chosen by application of suction) is pipetted on a chip containing a microstructured aperture. A single cell Figure 35. The patch pipette is moved to the cell using a is then positioned on the hole by suction and a tight micromanipulator under optical control. The cell is then connection (Gigaseal) is formed. The planar positioned by suction. [51] geometry offers a variety of advantages compared to the classical experiment: • it allows for integration of microfluidics, which enables automatic compound application for ion channel screening. • the system is accessible for optical or scanning probe techniques • perfusion of the intracellular side can be performed [51, 52]. Figure 36: (A)Procedure of cell contacting. (B)Measured current response to a voltage pulse and after a cell is sealed onto the aperture by suction (C) Close-up view of the mechanically and electrically tight contact of the cell membrane and the chip in cell attached mode [51]
  • 33. 33 IN VITRO SPP PROFILE RESULTS Table 3: VP35 Inhibition Assays ADME-Tox Assays NCE ST 1: VP35 ST2: PLAQUE ST3: hERG ST4: CYP 450 ST5: UGT INHIBITION, ASSAY(confirmatory INHIBITION INHIBITION ACTIVITY LUCIFERASE assay for VP35 (Planar patch ASSAY ASSAY BASED GENE inhibition) clamp assay) (Human Liver ASSAY IC50 (µM) Microsome PFU IC50 (µM) assay) IC50 (µM) IC50 (µM) NCE (1) 5.3 3.8x103 6.8 7.6 2.2 NCE (2) 3.4 103 8.9 8.2 7.0 NCE (3) 2.3 100 >10 >10 9.1 NCE (4) 9.1 5.4x103 3.0 2.9 2.2 NCE (5) 4.1 1.2x103 7.2 7.6 8.1 NCE (6) 4.2 2x103 5.6 5.2 5.0 NCE (7) 7.0 4.5x103 4.1 4.7 4.9 NCE (8) 7.5 5x103 4.6 5.0 5.1 NCE (9) 2.1 75 >10 >10 >10 NCE (10) 3.9 3.1x103 6.2 5.6 6.7
  • 34. 34 CONCLUSION I hypothesize that NCE (9) is the best as: • It is functionalized with hydrogen bond forming groups that could engage the basic amino acids ARG305, LYS109 and ARG312, which are critical for VP35 to effectively block activation of an IRF-3 responsive promoter. • It has got good inhibitory activity that is evident from the IC50 values of the luciferase reporter based gene assay and plaque assay. • It has got a safe ADME-pharmacokinetic profile that is evident from the ADME-tox (In vitro SPP) assays. The second best is NCE (3). NCE (9) is going to be referred as NCT 1087 in future studies.
  • 35. 35 PATENT APPLICATION A standard patent application for NCT1087, a VP35 protein inhibitor with the potential to act as an anti-ebola small molecule drug will be filed with the United States Patent and Trademark Office (USPTO). ABSTRACT Compounds in the present invention inhibit the activity of VP35 protein, thereby mitigating Ebola Viral infection as VP35 is critically involved in its pathogenesis. The compounds, the method of use and formulation is included in this patent. Compound being patented: • NCE-9 also known as NCT1087 • NCE-3 • NCE-2 • NCE-5 NCT1087 shows the maximum potency as a VP35 inhibitor. The other compounds show comparable potency. Therefore they will also be patented to slow down potential competition. Background of the invention: Ebola hemorrhagic fever is a severe, often-fatal viral hemorrhagic disease in humans and nonhuman primates that has appeared sporadically since its initial recognition in the Democratic Republic of Congo in 1976 [1]. It is caused by the Ebola Virus which interferes with the interior endothelial cell lining of blood vessels and coagulation which leads to hypovolemic shock [2]. Destruction of endothelial surfaces is associated with disseminated intravascular coagulation, and this may contribute to the hemorrhagic manifestations.[5] Fatality rate differs from 50-83% [2]. To date, beyond supportive care, no effective treatments, therapies, or vaccines are approved to treat or prevent Ebola virus infections because natural immunity to this infection is difficult to find, plus there are no immune correlates in humans [10]. Summary of the invention: The present invention has identified small organopharmaceuticals that can inhibit VP35, a major virulence factor in EBOV infection. It can be hypothesized that EBOV infection can be treated by administering an effective amount of compound NCT1087.
  • 36. 36 SELECTION OF APPROPRIATE ANIMAL MODELS FOR EVALUATING THE EFFICACY OF THE ANTI-EBOLA DRUG In order to effectively evaluate the efficiency of the lead compounds in halting the progression of the disease through inhibition of VP35, we need an animal model that can accurately represent the pathogenic mechanisms that underlie EHF. The three widely used animal models in Ebola research are mice, guinea pigs and NHPs with EHF being best reproduced in NHPs. There are several disadvantages associated with the use of rodent models and these are: [27] • Wild type EBOV is not lethal in them. • EBOV strains have to be adapted to these models through specific mutations in NP and VP24. • Some aspects of the human form of the disease cannot be authentically replicated such as the haemorrhagic manifestations. • There is considerable difference in rodent and human immunology. • Bystander apoptosis of lymphocytes, which is observed in humans, is not prominent in rodents. • a number of antiviral therapies and vaccines that were effective in mice failed to protect nonhuman primates. [53] Table 4: Comparison of animal models with the human disease [27][54] Mouse Guinea pig NHP Human Adaptation Yes Yes No No required Macular rash No No Yes Yes Haemorrhagic Not profound No Yes Yes manifestations Coagulation Not profound Conflicting data Yes Yes abnormalities
  • 37. 37 Mouse Guinea pig NHP Human Liver enzymes Elevated Elevated Elevated Elevated Thrombocytopen Yes Yes Yes Yes ia Bystander No No Yes Yes apoptosis Cytokine Yes Yes Yes Yes response Time to death 4–6 days 6–9 days 6–9 days 6–16 days Required infrastructure Moderate Moderate Very high NA and/or costs Ethical concerns Moderate Moderate High NA Monocytes, Monocytes, Monocytes, macrophages, macrophages, Monocytes, macrophages, dendritic dendritic cells, dendritic cells, macrophages, dendritic Permissive host cells, fibroblasts, fibroblasts, fibroblasts, cells, fibroblasts, cells hepatocytes, adrenal hepatocytes, hepatocytes, hepatocytes, endothelial cortical cells, endothelial adrenal cortical adrenal cortical cells, epithelial cells cells, epithelial cells, endothelial cells, endothelial cells, epithelial cells, epithelial Cytokines/chem okines IFN-a, IL-6, IL-18, IFN-a, IL-2, IL-6, IL-10, (increased MCP-1, TNF-a NE MIP-1a, MIP-1b, TNF-a circulating MCP-1, TNF-a levels) The only drawback with using NHPs is that the typical time of death is 6-9 days after exposure to ZEBOV infection, whereas in humans the time of death is prolonged which might be due to an adaptive immune response which is not seen in NHPs. However this doesn’t matter much in the drug that I will be testing as its primary function is to activate the innate immune system through initiation of the type I IFN pathway. In addition, experimentally infected NHPs are normally exposed to higher doses of EBOV than humans would be during a natural exposure [27].
  • 38. 38 Because of the ethical issues associated with the use of NHPs for research, I would first like to evaluate the efficacy of the NCEs on a mouse model and then proceed testing the promising candidates using a primate model. MOUSE MODEL USED FOR PRECLINICAL STUDIES (Efficacy evaluation) The most commonly used mouse strains for EHF research are: • BALB/c mice [55] • SCID mice [56] • ICR (CD-1) outbred mice [57] The Ebola VP35 protein is multifunctional, acting as a component of the viral RNA polymerase complex, a viral assembly factor, and an inhibitor of host interferon (IFN) production [31]. Therefore in my initial study with mouse models, SCID mice will be used in initial drug evaluation studies to gauge the lead compound’s ability to inhibit viral replication independent of immunological factors. Then in the second phase of the Mouse preclinical studies, adult, immunocompetent BALB/c mice will be used to evaluate the efficacy of the lead compound in inhibiting the VP35 antagonism of Type 1 IFN pathway.and thus see if this leads to increased expression of antiviral genes and subsequent immune cell clearance of the virus. Figure 37- Image taken from [58] **Image taken from [59]
  • 39. 39 The adaptation of EBO-Z to mice passaging intracerebrally in suckling mice, then in Vero cells and finally by subcutaneous or i.p. inoculation in progressively older suckling BALB/c mice. Virus recovered from the liver of a moribund ninth-passage mouse was plaque-purified twice and amplified. This ‘mouse-adapted virus’ is lethal for adult BALB/c and other mouse strains when inoculated i.p. The 50% lethal dose (LD50) is 0.03 plaque-forming units (pfu), or approximately one virion [60]. Adult SCID and BALB/c mice will be divided into four groups: • Control • EBOV infected (Negative control group) • EBOV infected treated with NCE at one concentration • EBOV infected treated with NCE at a different concentration. Infection will be done by inoculatiion (i.p.) with 1000 pfu (30 000 LD50) of mouse-adapted EBO-Z. Negative control groups can be treated with a placebo (PBS) or left as such [61]. Parameters checked for in the animal models: In case of the SCID mice group:[60] • serum viral titer levels will be examined every day postinfection. In case of BALB/c mice group: [60][61] • Serum cytokine concentrations of IFN-α, macrophage chemotactic protein-1 (MCP-1), interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) will be measured using commercial kits. • Liver and spleen of infected and uninfected mice will be examined by Immunohistochemistry and Electron microscopy so that we can study the effect of the NCE on the progression of the infection in major target organs. An experiment we can do to support my hypothesis that the NCE elicits its effect by way of the type I IFN pathway is by co-administering antibodies to interferon alpha/beta. This will eliminate the efficacy of the drug that it mediates via the IFN pathway. This will prove beyond doubt that the NCE acts by inhibiting VP35 and subsequent activation of the IFN pathway. We can again confirm this by comparing the production of IFN-α in Ebola virus-infected mice treated with the NCE or with placebo. [61]
  • 40. 40 NON-HUMAN PRIMATE MODEL USED FOR PRECLINICAL STUDIES EBOV has been shown to cause fatal hemorrhagic manifestations in: [57][53] • Rhesus macaques ( Macaca rhesus ) • Cynomolgus macaques ( Macaca fascicularis ) • African green monkeys ( Cercopithecus aethiops) • Baboons ( Papio hamadryas ) • Vervet monkeys ( Cercopithecus aethiops ) These are the NHP models most commonly used for Ebola research as they accurately replicate the clinical manifestations of EHF in humans. I will be choosing a combination of Rhesus and Cynomolgus macaques for the preclinical testing of my NCEs as these two models have been the most widely used NHP model for EBOV research. Figure 37 -Representative cutaneous rashes from cynomolgus monkeys experimentally infected with EBOV- Zaire. A: Characteristic petechial rash of the right arm at day 4.B: Petechial rash of the inguinal region at day 5. Image taken from [62]. Figure 38. Rhesus monkey infected with Ebola virus showing an extensive rash on the face and anterior aspects of the arms [63].
  • 41. 41 As in the case of mouse models the NHPs will be divided into three groups: • Control • EBOV infected (Negative control group) • EBOV infected treated with NCE at a concentration defined in the mouse model study. Infection will be done by inoculation (i.p.) with 1000 pfu (30 000 LD50) of EBO-Z. Adaptation of the EBOV strain is not required. EBOV from a fatally infected patient can be directly used. [62] Negative control groups can be treated with a placebo (PBS) or left as such. Parameters checked for in the animal models: • Cytokine/chemokine levels of interleukin (IL)-2, IL-4, IL-10, IL-12, interferon (IFN), tumor necrosis factor (TNF), IL-6, IFN-α, IFN-ß, MIP-1, MIP-1ß, IL-1ß, IL-8, IL-18, and MCP-1 in monkey sera/plasma can be assayed using commercially available ELISA kits. (generally their levels should have increased in treated models) [62][64]. • Nitrate levels were determined using a colorimetric assay. (reduction should be seen in treated models) [62][64]. • Viral titer levels will be determined every day post infection. (reduction should be seen in treated models). • Necropsy will be performed on the models. Tissue samples of the liver, spleen and the lung were collected from each monkey for histopathological, immunohistochemical, in situ hybridization examination as well as electron microscopy examination [62][64]. • Total white blood cell counts, white blood cell differentials, red blood cell counts, platelet counts, total hemoglobin concentration will be determined to account for bystander apoptosis which is a common manifestation of EBOV infection [64]. • Concentrations of albumin (ALB), amylase (AMY), alanine aminotransferase (ALT), aspartate aminotransferase (AST) will be analyzed as AST and ALT levels are found to be elevated during EBOV infection. So there should be a reduction in their levels in treated models [64][65]. • Plasma levels of fibrin degradation products (D-dimers) will be analyzed using ELISA as they are found to decrease during the infection [64],
  • 42. 42 BIOMARKERS Biomarkers in the context of a viral infection are biological molecules associated with a disease caused by an infectious viral agent. They are essential in diagnostic and therapeutic processes as they: [66][76] • Enable early diagnosis • Reduce risk in drug development • Improve patient outcomes • Patient stratification • Assessment of drug toxicity and efficacy • Disease staging • Disease prognosis • Guide molecularly targeted therapy • Monitor the activity and therapeutic responses across a variety of diseases. Figure 39: Left: Disease pathway and potential impact of biomarkers. Image taken from [67] Right: .Biomarker science is a multifaceted discipline which impacts all aspects of drug development and modern medicine. Image taken from [81]
  • 43. 43 Figure 40: Biomarker discovery and application as an integral component of drug development. (a) Disease program approaches should yield biomarker candidates. (b) Parallelism of biomarker development in the drug development pipeline. Biomarkers can be applied to many steps in the drug-discovery pipeline. They can also be evaluated for use in both a pre- and post-market follow-up study. Image taken from [82]. DISEASE BIOMARKERS There are essentially three kinds of disease biomarkers: 1. Disease Trait- is essentially a biomarker of exposure that is used to calculate the risk of exposure, that is the likelihood that an individual is susceptible to a particular disease. [67] The likelihood of getting Ebola hemorrhagic fever is not governed by any genetic factors. Therefore this disease doesn’t have a disease trait biomarker as every segment of the population is equally susceptible to it.
  • 44. 44 2. Disease State- is a biormarker used for diagnostic purposes. The potential uses of this biomarker is the [67] • identification of individuals who will be or are in the preclinical stages of the disease. • reduction in disease heterogeneity in clinical trials. • reflection of a disease pathogenesis that is the phase of induction, latency and detection. • target for a clinical trial. Table 5: Biomarker for diagnosis and typing of Ebola Hemorrhagic Fever [68] Validated Potential Detection/Diagnosis Glycoprotein (GP) VP24 Nucleoprotein VP30(minor GP gene nucleoprotein) NP gene VP35 (P-like protein) VP40 (Matrix Protein) VP24 gene VP30 gene VP35 gene VP40 gene Pathogen Typing GP gene Virulence Factor Glycoprotein (GP) 3. Disease Rate - This biomarker is used to indicate or evaluate the stage of the disease. It can also be referred to as a prognostic biomarker [84]. In case of ebola hemorrhagic fever it will be the viral titer that is copies of EBOV/ml of blood or serum.
  • 45. 45 SURROGATE BIOMARKER “A surrogate biomarker is defined as a biomarker intended to substitute for a clinical endpoint, the latter being ‘a characteristic or variable that reflects how a patient feels, functions, or survives” [69]. In case of Ebola Hemorrhagic Fever, hypotension, generalized fluid distribution problems, lymphopenia, coagulative disorders, and hemorrhages are its characteristic features [70]. In Ebola-infected patients, there is extensive apoptosis of leukocytes which is associated with the fatal outcome of the disease. This observation appears consistent with considerable lymphopenia and severe damage to lymphoid tissues, such as spleen, lymph node and bone marrow, seen in patients and experimentally infected monkeys [71]. Therefore the surrogate marker that I have selected is the level of lymphocytes in the blood which will progressively decrease with disease pathogenesis and will increase with a therapeutic intervention. TOXICITY BIOMARKERS These biomarkers are used to monitor the adverse effects of a specific drug on any unintended cellular processes, cells, tissues or organs [84]. RENAL TOXICITY Due to the possible renal side effects of my NCE, the following nephrotoxic biomarkers which has been approved by the FDA will be used to monitor the patients during the treatment period [72][73]. • Serum creatinine • Blood urea nitrogen • Glomerular filtration rate HEPATIC TOXICITY Pro-inflammatory cytokines such as: • tumor necrosis factor alpha (TNFalpha), • interleukin 1beta(1L-1beta) • and interleukin 6 (IL6) are involved in acute and chronic liver damage. They are released both from the liver and from distal sites during hepatic toxic injury [74]. These cytokines are also involved in disseminated intravascular
  • 46. 46 coagulation (DIC), which is a prominent manifestation of Ebola virus (EBOV) infection. DIC is a syndrome characterized by coagulation abnormalities including systemic intravascular activation of coagulation leading to widespread deposition of fibrin in the circulation, which contributes to the multiple organ failure and high mortality rates characteristic of EBOV infections [70]. Alternatively alanine transaminase and bilirubun could be used as a measure of liver toxicity [75]. TARGET BIOMARKER A target biomarker is used to report the interaction of the NCE with the target. The Ebola VP35 protein is multifunctional, acting as a component of the viral RNA polymerase complex, a viral assembly factor, and an inhibitor of host interferon (IFN) production [77]. The EBOV virus VP35 protein, which plays an essential role in viral RNA synthesis, acts as a type 1 IFN antagonist by blocking phosphorylation of the IFN-regulatory factor 3 (IRF3), which acts as a transcription factor for IFN production and turns on a number of anti-viral genes such as IFN-α/β, IL-15, ISG15, and ISG56 [78][79]. Inhibition of VP35 by the NCE will result in the phosphorylation of IRF3 that forms a complex with CREBBP that translocates to the nucleus and activates the transcription of the different anti-viral genes; therefore IRF3 or phosphorylation of IRF3 will be the target biomarker [86]. MECHANISM BIOMARKER Mechanism biomarker is a biomolecule associated with a specific pathway that gets affected by the drug and brings about the desired clinical effect. In case of Ebola hemorrhagic fever, the NCE will inhibit VP35 bringing about the phosphorylation of IRF3. This leads to the transcriptional up-regulation of type I IFN, IFN-α, IFN- γ and IFN-β, genes. This signaling stimulates transcription of IFN-sensitive genes, including a number that encode antiviral proteins, and leads to the induction of an antiviral state. Among the antiviral proteins induced in response to type I IFN are dsRNA-dependent protein kinase R (PKR), 2′,5′- oligoadenylate synthetase (OAS), and the Mx proteins [80]. So the mechanism biomarker for the EHF will be IFN- α, IFN- γ and IFN-β. IFN- γ is indicative of the adaptive immune system getting activated [85].
  • 47. 47 EFFICACY BIOMARKER Efficacy biomarkers are used to predict a patient’s response to a drug [81]. They essentially monitor the beneficial effects of a specific drug on the intended drug target or medical condition [84]. In case of EHF, the efficacy biomarker will be the viral load in the patient’s blood. It should decrease if the drug is effective which is indicative of its efficacy. TRANSLATIONAL BIOMARKER Translational biomarker is a bridging biomarker that can be used in multiple species and can be used to translate the results from preclinical animal studies to clinical human studies i.e. directly link responses between species and follow such injury in both preclinical and clinical settings [83]. In case of EHF, translational biomarkers can be several things such as: • viral titer • interferon (IFN-α and IFN-ß) levels • interleukin (IL-2, IL-4, IL-10, IL-12) level • tumor necrosis factor levels STRATIFICATION BIOMARKER Stratification biomarker is used to identify patients likely to respond to a specific drug or suffer from its side-effects prior to administration of the drug [84]. Since the NCE targets VP35, therefore one of the stratification biomarkers as follows: • presence of EBOV infection so that the drug can target VP35 • the patient must be immunocompetent as the drugs works by removing the block imposed by the virus on the initiation of the innate and adaptive immune system. Therefore the second stratification biomarker would be the presence of functional type I IFN genes as well as IRF3.
  • 48. 48 EFFICACY STUDIES FOR MINIMUM EFFECTIVE DOSE Table 6: ANIMAL NO. OF NCT108 EFFICAC SURROG TARGET MECHANISM EFFICACY MODELS ANIMA 7 Y BM- ATE BM- BM- IRF3 BM- IFN- γ ENDPOINT ( LS CONC. VIRAL LYMPHO CONC. CONC. No. of (mg/kg) TITER CYTES (% animals (pfu/ml) CONC. (% activation (p recovered total ) g/ from EBOV cells) ml infection) ) 0/2 SCID 2 Placebo 1.3 x 109 5% 0.1 <100 1/4 SCID 4 1mg/kg 4.8 x 105 10% 11 800 3/4 4 2 mg/kg 5.2 x 104 18% 45 1200 4/4 4 4 mg/kg 2.2 x 102 35% 65 2000 0/2 BALB/c 2 Placebo 4.1 x 1010 3.5% 1 <100 2/4 BALB/c 4 1 mg/kg 2.3 x 105 12% 12 900 4/4 4 2 mg/kg 1.4 x 103 22% 56 1300 4/4 4 4mg/kg 102 40% 75 2500 Rhesus 0/2 2 Placebo 2 x 109 6% 1.2 <100 Monkeys Rhesus 2/4 4 1 mg/kg 3.4 x 105 13% 14 780 Monkeys 4 2 mg/kg 1.3 x 104 20% 57 1500 3/4