Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTI-RABIES MONOCLONAL ANTIBODIES AND COCKTAIL THEREOF
Document Type and Number:
WIPO Patent Application WO/2020/089742
Kind Code:
A1
Abstract:
The disclosure provides a murine monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses. It also provides a cocktail of at least two monoclonal antibodies with said properties. The cocktail can neutralize the virus that is derived from species such as bats, dogs, cows, mongooses, skunks, and wolves, and thus can be useful in treating a patient that has potentially been infected. Further, the disclosure provides a combination of muring monoclonal antibody or cocktail of at least two monoclonal antibodies and anti-rabies vaccine for use in post-exposure prophylaxis (PEP) with rabies or rabies-related viruses.

Inventors:
MENDIRATTA SANJEEV KUMAR (IN)
BANDYOPADHYAY SANJAY (IN)
KALITA PANKAJ (IN)
KANSAGRA KEVINKUMAR (IN)
Application Number:
PCT/IB2019/059118
Publication Date:
May 07, 2020
Filing Date:
October 24, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
CADILA HEALTHCARE LTD (IN)
International Classes:
A61K39/42; A61P31/14; C07K16/10; A61K39/00
Domestic Patent References:
WO2016078761A12016-05-26
WO2017027805A12017-02-16
WO2013048130A22013-04-04
Other References:
THE JOURNAL OF INFECTIOUS DISEASES, vol. 210, 2014, pages 200 - 8
BULL. WID HLTH ORG., vol. 45, 1971, pages 303 - 315
J, GEN. VIROL., vol. 64, 1983, pages 843 - 851
J. GEN. VIROL., vol. 64, 1983, pages 1649 - 1656
PROC. NATL. ACAD. SCI. USA, vol. 81, November 1984 (1984-11-01), pages 7194 - 7198
Attorney, Agent or Firm:
ELANGBAM, Nanda et al. (IN)
Download PDF:
Claims:
We claim:

1. A monoclonal antibody comprising:

(a) CDRH1 comprising a polypeptide set forth in SEQ ID NO: 1;

(b) CDRH2 comprising a polypeptide set forth in SEQ ID NO: 2;

(c) CDRH3 comprising a polypeptide set forth in SEQ ID NO: 3;

(d) CDRL1 comprising a polypeptide set forth in SEQ ID NO: 4;

(e) CDRL2 comprising a polypeptide set forth in SEQ ID NO: 5; and

(f) CDRL3 comprising a polypeptide set forth in SEQ ID NO: 6.

2. The monoclonal antibody as claimed in claim 1 comprising:

(a) a heavy chain variable region comprising a polypeptide set forth in SEQ ID NO: 7 and

(b) a light chain variable region comprising a polypeptide set forth in SEQ ID NO: 8.

3. The monoclonal antibody as claimed in claim 2 comprising:

(a) heavy chain comprising a polypeptide set forth in SEQ ID NO: 9 and

(b) light chain comprising a polypeptide set forth in SEQ ID NO: 10.

4. A monoclonal antibody comprising:

(a) heavy chain comprising a polypeptide set forth in SEQ ID NO: 11 and

(b) light chain comprising a polypeptide set forth in SEQ ID NO: 12.

5. The cocktail of at least two monoclonal antibodies wherein one of the antibodies is the monoclonal antibody as claimed in claim 1.

6. A cocktail of at least two monoclonal antibodies wherein one of the antibodies is the monoclonal antibody as claimed in claim 4.

7. The cocktail of at least two monoclonal antibodies wherein two antibodies are monoclonal antibodies as claimed in claim 1 and claim 4.

8. The antibody as claimed in any preceding claims which is an IgG isotype, preferably IgGl or IgG2.

9. The monoclonal antibody as claimed in any preceding claim is murine monoclonal antibody.

10. A murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies for the PEP of exposure with rabies and rabies -related viruses wherein the antibody allows the active immunization to generate and maintain an effective immune response.

11. The murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies as claimed in claim 10 is selected from claims 1-9.

12. A combination of a murine monoclonal antibody as claimed in any preceding claim and an anti-rabies vaccine for the PEP of exposure with rabies and rabies -related viruses.

13. ETse of murine monoclonal antibody as claimed in any preceding claim for the PEP of exposure with rabies and rabies-related viruses.

14. A combination of cocktail of at least two murine monoclonal antibody as claimed in any preceding claim and anti-rabies vaccine for the PEP of exposure with rabies and rabies-related viruses.

15. ETse of cocktail of at least two monoclonal antibodies as claimed in any preceding claim for the PEP of exposure with rabies and rabies-related viruses.

16. A composition comprising monoclonal antibody or a cocktail of at least two monoclonal antibodies of any one of claims 1-14, and a pharmaceutically acceptable carrier.

17. The composition as claimed in claim 15 comprising excipients selected from buffer, sugar, polyol, amino acid, surfactant, and polymer.

18. The composition as claimed in claim 16 comprising excipients selected from citrate buffer, polysorbate 80, sodium chloride.

19. The composition as claimed in claims 15-17 which is a lyophilized formulation.

20. The composition as claimed in claim 15, wherein monoclonal antibody or one of the antibodies of a cocktail is present in the range of 1 IU/mL to 5000 IU/mL, preferably 100 IU/mL to3000 IU/mL, more preferably 150 IU/mL or 300 IU/mL or 600 IU/mL.

21. The composition as claimed in claim 15, wherein cocktail is present in the range of 100 IU/mL to 150 IU/mL, preferably 400 IU/mL to 1500 IU/mL, more preferably 300 IU/mL or 600 IU/mL or 1200 IU/mL.

22. The monoclonal antibody or cocktail of at least two monoclonal antibodies as claimed in any preceding claim which is administered via infiltration in to and around the site of exposure and/or intramuscularly at site(s) distant from the wound site.

23. The monoclonal antibody or cocktail of at least two monoclonal antibodies as claimed in any preceding claim which is administered at a dose of 1 IU/Kg to 100 IU/Kg, preferably 10 IU/Kg to 80 IU/Kg, more preferably 40 IU/Kg.

24. The monoclonal antibody or a cocktail of at least two monoclonal antibodies as claimed in any preceding claim which is administered up to 7 days after the exposure beginning immediately after the exposure.

25. A kit comprising monoclonal antibody or cocktail of at least two monoclonal antibodies as claimed in any preceding claims.

26. A method for diagnosing rabies using the monoclonal antibody or cocktail of at least two monoclonal antibodies as claimed in any preceding claims.

27. A method of detecting rabies virus using the monoclonal antibody or cocktail of at least two monoclonal antibodies as claimed in any preceding claims.

28. A process of making antibody as claimed in any preceding claim by cell culture method comprising:

i) culturing the host cell of expression system, preferably Sp2/0 cell expressing monoclonal antibody capable of binding and neutralizing rabies virus; and

ii) isolating and recovering monoclonal antibody capable of binding and neutralizing rabies virus expressed in the said host cell.

29. A process of purifying the antibodies as claimed in any preceding claim by suitable chromatography or purification techniques.

Description:
ANTI-RABIES MONOCLONAL ANTIBODIES AND COCKTAIL THEREOF

FIELD OF THE INVENTION

The present invention relates to a monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses. More specifically, it relates to a cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses.

BACKGROUND OF THE INVENTION

Rabies is a zoonotic disease caused by rabies virus (RABV), the type member of the Lyssavirus genus, and is responsible for >55 000 deaths per annum largely in the developing world, where transmission usually occurs following the bite of an infected dog. If left untreated, the virus progressively infects surrounding neurons and propagates in the central nervous system leading, almost invariably, to death. The disease can be prevented by post exposure prophylaxis (PEP), which consists of administration of inactivated RABV vaccine together with passive antibody therapy. In a standard passive antibody therapy, polyclonal rabies immunoglobulins (RIG), derived either from immunized human (HRIG) or equine (ERIG) sources, is infused into the wound site. (The Journal of Infectious Diseases 2014; 210:200-8)

Given that the rabies or rabies related viruses cannot be neutralized after it has entered the peripheral nerves at the site of the bite, all post-bite prophylaxis activities that work by neutralizing the virus have to be done at the bite site only. RIGs have been used to neutralize the virus in the wound before the virus enters the nerve. It is obvious that having more amount of drug available at the bite site will provide a better chance of protection of the patient.

As described above, a standard post-bite prophylaxis regimen after a rabid animal bite involves giving of both RIGs and vaccine. RIGs provide early immunity against the virus in a passive manner while vaccine provides later immunity against the virus in an active manner. Any successful regimen has to maintain the right balance of i) RIGs which provide high anti-rabies virus protection in the early phase i.e., immediately (72 hours) after the animal bite whilst ii) it should not neutralize the anti-rabies vaccine (which is also given immediately after the animal bite) and thus developing anti-rabies virus protection via active immunity in the patient in the late phase i.e., up to day 42 or longer.

Clinical trial data of ERIGs, which are horse derived polyclonal anti-rabies sera obtained from repeatedly immunized animals, show that these drugs can be given up to 40 IU/kg in humans. On the other hand, HRIGs which are human donor derived polyclonal anti-rabies sera obtained from immunized human donors, cannot be given beyond 20 IU/kg as a higher dose ends up neutralizing the vaccine thereby failing the post-bite prophylaxis regimen itself. (Bull. Wid Hlth Org., 1971 45, 303-315)

An ideal regimen should provide higher amounts of RIG at the bite site to provide early phase protection while still allowing the vaccine to provide late phase protection. The difference in doses between the human and horse RIGs is that the human patient can easily recognize the horse derived RIGs as a foreign entity and thereby raising an immune response against the RIG itself. Thus, high amount of ERIG drug becomes available at the bite site and does not remain in the body long enough and allowing the vaccine to provide late phase protection. On the other hand, the human derived RIGs are not recognized as foreign entity and therefore the patient’s immune system does not react to it, allowing the HRIG to circulate in patient for longer period of time and neutralizing the vaccine if it is given beyond 20 IU/kg. A higher dose of 40 IU/kg of an HRIG failed the regimen itself.

Additionally, most likely due to similar reason, a recently approved human monoclonal antibody for the post-bite prophylaxis regimen of rabies has been approved to be used at 3.33 IU/kg. (Product insert of Rabishield ® ).

Therefore, there is a need of an antibody with a low circulating half-life such as a non-human or non-humanized antibody capable of binding and neutralizing the rabies or rabies related viruses as a part of a post -bite prophylaxis regimen for rabies.

Further, these serum derived antibodies often suffer from various drawbacks, including limited availability, batch-to-batch variation, high cost, contamination with blood-borne adventitious agents, and/or risk of adverse reactions. For these reasons, the World Health Organization (WHO) encourages the development and evaluation of alternative biologies for RIG replacement. One such alternative strategy is offered by monoclonal antibodies (mAbs) that are capable of neutralizing a wide range of RABV isolates. Both RIGs and monoclonals are directed against the viral G protein, as several studies have demonstrated that anti-G protein antibodies provide protection against rabies or rabies related viruses. G protein consists of three distinct antigenic sites. Antigenic site I, II and III exist on the rabies virus glycoprotein which have been determined based on the antigenic analysis of glycoprotein variants of CVS-l 1 virus using 12 neutralizing monoclonal antibodies. (J, gen. Virol. (1983), 64, 843-851). The rabies virus glycoprotein (G) forms surface projections through the viral lipid envelope and is the only protein capable of inducing and reacting with virus - neutralizing antibody (VNA). Studies have established that the isolated G is capable of protecting animals against rabies (J. gen. Virol. (1983), 64, 1649-1656; Proc. Natl. Acad. Sci. USA. Vol. 81, 7194-7198, November 1984). The antigenic sites on the glycoprotein has been studied by using mutants resistant to neutralization by monoclonal antibodies. Three major sites - Site I, II and III has been described in this literature.

RIGs by design comprise of anti-G antibodies directed against multiple epitopes on the G- protein. A monoclonal antibody can bind to only one single epitope on the G-protein. A passive immunity providing product that comprises of only one monoclonal antibody suffers from the risk of a mutant rabies virus escaping the drug if the epitope in question has been lost due to the mutation. Therefore a cocktail of two monoclonal antibodies directed to two distinct non-overlapping epitopes on the G-protein would provide better protection against mutants.

W02013048130 discloses an anti-rabies antibody which binds rabies virus. The said patent application discloses and teaches chimeric, humanized or human monoclonal antibodies. Such antibodies may not be given in high dose to provide high anti-rabies virus protection in the early phase due to reasons discussed herein above related to human origin RIGs.

Accordingly, there is an urgent need for the development of a monoclonal antibody preparation for treatment of rabies which can be produced and supplied in large amounts by cultural synthesis, and can have uniform quality. Such a product would be produced from well characterized cell banks and therefore free from the risk of viral infections and infections from other adventitious agents. Being a monoclonal antibody based product the absolute amount of protein given to the patient as a drug would be much lower than a polyclonal RIG based drug and therefore carry lesser risk on anaphylactic reactions.

Therefore, the current invention provides as described herein, a preparation of monoclonal antibodies capable of binding at two separate sites and neutralizing rabies or rabies related viruses and further a cocktail of such preparation comprising the said two monoclonal antibodies mixed with equipotent amounts for neutralizing rabies or rabies related viruses.

SUMMARY OF THE INVENTION

The present invention provides a murine monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses. It also provides a cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses.

According to the present invention, the said cocktail can neutralize the rabies and rabies related virus that is derived from species such as bats, dogs, cows, mongooses, skunks, and wolves, and thus can be useful in treating a patient that has potentially been infected by the rabies or rabies related viruses derived from a wide variety of species. Further, current invention provides combination of murine monoclonal antibody or cocktail of at least two mono clonal antibodies and anti-rabies vaccine for use in Post exposure prophylaxis (PEP) of exposure with rabies or rabies-related viruses.

BRIEF DESCRIPTION OF FIGURE

Figure 1 shows linear plot of mean RVNA concentration versus time curves after administration of cocktail of mAh A and mAh B + vaccine and Imogam + vaccine to patients with suspected rabid animal bite. Cocktail was delivered at a dose of 40 IU/kg while Imogam (HRIG) was given at its approved dose of 20 IU/kg. After day 14, the cocktail of the present invention maintains higher mean RVNA titers in comparison to HRIG.

List of abbreviations used herein the specification:

Abbreviations of amino acids as used in the current application are provided in below table:

Other abbreviations used in the present application:

AB V : Australian bat virus

BHK: Baby Hamster Kidney fibroblasts

CDRH1 : Complementarity determining region 1 of heavy chain CDRH2: Complementarity determining region 2 of heavy chain CDRH3 : Complementarity determining region 3 of heavy chain CDRL1 : Complementarity determining region 1 of light chain CDRL2: Complementarity determining region 2 of light chain CDRL3: Complementarity determining region 3 of light chain CHO: Chinese hamster ovary

CSO: Cynoglossus Semilaevis Ovary

CVS: Challenge Virus Strain

DP: Drug product

DS: Drug substance

EBLV : European bat lyssavirus

ERIGs: Equine rabies immunoglobulins

FAVN: Fluorescent antibody virus neutralisation

FFU: Fluorescence-Forming Unit

FITC: Fluorescein isothiocyanate

HC: Heavy chain

HCVR: Heavy chain variable region

HEK: Human embryonic kidney

HeFa: Henrietta Facks

HRIGs: Human rabies immunoglobulins

EC: Fight chain

FCVR: Fight chain variable region

mAb A: Monoclonal antibody A

mAb B: Monoclonal antibody B

MCB: Master cell bank

NA cells: Neuroblastoma cells

PBS: Phosphate buffer saline

PEP: Post exposure prophylaxis

RAB V : Rabies virus

RFFIT: Rapid fluorescent focus inhibition test

RIGs: Rabies immunoglobulins

RVNA: Rabies virus neutralizing antibody

SRIG: Standard rabies immunoglobulin

SV: Street Virus

WCB: Working cell bank LIST OF SEQUENCE LISTINGS USED HEREIN THE SPECIFICATION:

SEQ ID NO. 1: SSWMH (CDRH1 of mAb A)

SEQ ID NO. 2: QTHPN S G YTN YNEKFKG (CDRH2 of mAb A)

SEQ ID NO. 3: ES GDGPHW YFD V (CDRH3 of mAb A)

SEQ ID NO. 4: KASQDVSTAVA (CDRL1 of mAb A)

SEQ ID NO. 5: SASYRYT (CDRL2 of mAb A)

SEQ ID NO. 6: QQHYSSPHT (CDRL3 of mAb A)

SEQ ID NO. 7:

QVQLQQPGSVLVRPGASVKLSCKTSGYAFTSSWMHWAKQRPGQGLEWIGQTHPNS GYTNYNEKFKGKATLTVDTSS STA YVDLS SLTSEDS A VYYCARES GDGPHWYFD V WGAGTAVTVSS (HCVR of mAb A)

SEQ ID NO. 8:

DIVMTQSHKFMSTSVGDRVSITCKASQDVSTAVAWYQQKPGQSPKLLIYSASYRYT GVPDRFTGSGSGTDFTFTISSVQAEDLAVYYCQQHYSSPHTFGGGTKLETK (LCVR of mAh A)

SEQ ID NO. 9:

QVQLQQPGSVLVRPGASVKLSCKTSGYAFTSSWMHWAKQRPGQGLEWIGQTHPNS GYTNYNEKFKGKATLTVDTSS STA YVDLS SLTSEDS A VYY CARES GDGPHWYFD V WGAGTAVTVSSAKTTPPSVYPLAPGSAAQTNSMVTLGCLVKGYFPEPVTVTWNSGS LSSGVHTFPAVLQSDLYTLSSSVTVPSSTWPSETVTCNVAHPASSTKVDKKIVPRDCG CKPCICTVPEVSSVFIFPPKPKDVLTITLTPKVTCVVVDISKDDPEVQFSWFVDDVEVH TAQTQPREEQFNSTFRSVSELPIMHQDWLNGKEFKCRVNSAAFPAPIEKTISKTKGRP KAPQVYTIPPPKEQMAKDKVSLTCMITDFFPEDITVEWQWNGQPAENYKNTQPIMD TDGSYFVYSKLNVQKSNWEAGNTFTCSVLHEGLHNHHTEKSLSHSPGK (HC of mAb A)

SEQ ID NO. 10:

DIVMTQSHKFMSTSVGDRVSITCKASQDVSTAVAWYQQKPGQSPKLLIYSASYRYT GVPDRFTGSGSGTDFTFTISSVQAEDLAVYYCQQHYSSPHTFGGGTKLETKRADAAP TVSIFPPSSEQLTSGGASVVCFLNNFYPKDINVKWKIDGSERQNGVLNSWTDQDSKD STYSMSSTLTLTKDEYERHNSYTCEATHKTSTSPIVKSFNRNEC (LC of mAb A)

SEQ ID NO. 11: QVQLKESGPGLLAPSQSLSITCTVSGFSLTGHGVNWVRQPPGKGLEWLGIIWADGTT NYNSALKSRLSISKDNSKSQVFLKMNSLQTDDTASYYCAREGDISGYYFDYWGQGT TFTVSSAKTTPPSVYPFAPGCGDTTGSSVTFGCFVKGYFPESVTVTWNSGSFSSSVHT FPAFFQSGFYTMSSSVTVPSSTWPSQTVTCSVAHPASSTTVDKKFEPSGPISTINPCPP CKECHKCPAPNFEGGPS VFIFPPNIKDVFMISFTPKVTCVVVDVSEDDPDV QIS WFVN NVEVHTAQTQTHREDYNSTIRVVSTFPIQHQDWMSGKEFKCKVNNKDFPSPIERTIS KIKGFVRAPQVYIFPPPAEQFSRKDVSFTCFVVGFNPGDISVEWTSNGHTEENYKDT APVFDSDGSYFIYSKFNMKTSKWEKTDSFSCNVRHEGFKNYYFKKTISRSPGK (HC of mAh B)

SEQ ID NO. 12:

DVQMTQTTSSFSASFGDRVTITCRPSQDINNYFSWYQQKPDGTVKFFIYYTSRFHSG VPSRFSGSGSGTDYSFTISNFEQEDFATYFCQQGNTFPPTFGGGTKFEIKRADAAPTV SIFPPSSEQFTSGGASVVCFFNNFYPKDINVKWKIDGSERQNGVFNSWTDQDSKDST YSMSSTFTFTKDEYERHNSYTCEATHKTSTSPIVKSFNRNEC (EC of mAh B)

DEFINITIONS

The term“cocktail” as used herein is a mixture of at least two monoclonal antibodies as two independent active drug substances. The cocktail according to the present invention comprises preferably mixture of mAh A and mAh B. Preferably, the said cocktail is prepared in the formulation buffer. Cocktail is prepared by mixing therapeutic amounts, preferably equipotent amounts of the two active drug substance materials of monoclonal antibodies in the formulation buffer. Finally, it is formulated with other suitable pharmaceutical excipient(s) to prepare a stable pharmaceutical composition of anti-rabies monoclonal antibodies which can be used for therapeutic or treatment purpose.

The term“PEP” or“post exposure prophylaxis” as used herein includes following steps:

1. All bite wounds, scratches and RABV-exposure sites should be attended to as soon as possible after the exposure; thorough washing and flushing of the wound for approximately 15 minutes, with soap or detergent and copious amounts of water, is required. Where available, an iodine-containing, or similarly viricidal, topical preparation should be applied to the wound. 2. A series of rabies vaccine injections should be administered promptly after an exposure as a part of the active immunization component of the PEP.

3. RIG should be administered for severe category III exposures as a part of the passive immunization component of the PEP.

Rabies vaccine according to the present invention may include any anti-rabies vaccine known in the art. RIG according to the present invention may include murine monoclonal antibody or cocktail of at least two monoclonal antibodies.

The term“mAb A” as used herein is a monoclonal antibody which can bind rabies or rabies related viruses and neutralize it. A monoclonal antibody mAh A according to the current invention comprising a CDRH1 comprising a polypeptide set forth in SEQ ID NO: 1, a CDRH2 region comprising a polypeptide set forth in SEQ ID NO: 2, a CDRH3 region comprising a polypeptide set forth in SEQ ID NO: 3, a CDRL1 comprising a polypeptide set forth in SEQ ID NO: 4, a CDRL2 region comprising a polypeptide set forth in SEQ ID NO: 5 and a CDRL3 region comprising a polypeptide set forth in SEQ ID NO: 6. HCVR and LCVR region of mAh A according to the current invention is a polypeptide set forth in SEQ ID NO: 7 and a polypeptide set forth in SEQ ID NO: 8, respectively. Further, heavy chain and light chain of mAh A according to the current invention is a polypeptide set forth in SEQ ID NO: 9 and a polypeptide set forth in SEQ ID NO: 10, respectively. Preferably, mAh A according to the present invention is a murine monoclonal antibody. In one of aspects, the current invention provides mAh A comprising IgGl constant region, preferably murine IgGl. It binds to site II on G protein of rabies virus envelope.

The term“mAb B” as used herein is a monoclonal antibody which can bind rabies or rabies related viruses and neutralize it. A monoclonal antibody mAh B according to the current invention comprising heavy chain and light chain of mAh B according to the current invention is a polypeptide set forth in SEQ ID NO: 11 and a polypeptide set forth in SEQ ID NO: 12, respectively. Preferably, mAh B according to the present invention is a murine monoclonal antibody. In one of aspects, the current invention provides mAh B comprising IgG2b constant region, preferably murine IgG2b. It binds to site III on G protein of rabies virus envelope. The term“antibody” as referred to herein includes whole antibodies and any antigen-binding fragment (i.e.,“antigen-binding portion”) or single chains thereof. An“antibody” refers to a glycoprotein comprising at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds, or an antigen-binding portion thereof. Each heavy chain is comprised of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region is comprised of three domains, CH1, CH2 and CH3. Each light chain is comprised of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region is comprised of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR), interspersed with regions that are more conserved, termed framework regions (FR). Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clq) of the classical complement system. In a preferred aspects, the antibody according to the present invention is a murine antibody.

The terms“monoclonal antibody” or“monoclonal antibody composition” as used herein refer to a preparation of antibody molecules of single molecular composition. A monoclonal antibody composition displays a single binding specificity and affinity for a particular epitope.

The term“pharmaceutical composition” refers to preparations which are in such form as to permit the biological activity of the active ingredients to be unequivocally effective. The term“pharmaceutical formulation” or“pharmaceutical composition” or“composition” can be used here interchangeably.“Pharmaceutically acceptable” carrier or excipients are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed. The term“excipient” refers to an agent that may be added to a formulation to stabilize the active drug substance in the formulated form to adjust and maintain osmolality and pH of the pharmaceutical preparations. Examples of commonly used excipients include, but are not limited to, sugars, polyols, amino acids, surfactants, and polymers. “Pharmaceutically acceptable” excipients are those which can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient employed.

In a pharmacological sense, in the context of the present invention, a“therapeutic amount” or “effective amount” of an antibody refers to an amount effective in the prevention or treatment of a disorder for the treatment of which the antibody is effective.

The term“equipotent amount” according to the present invention refers to an amount of each antibody which provides same potency with respect to another antibody or antibodies present in cocktail preparation. Potency of an antibody according to the current invention can be measured in international unit. The potency of the rabies or rabies related viruses neutralizing antibodies is determined by RFFIT method in comparison with an‘Anti-rabies Immunoglobulin, Human WHO International Standard’ (RAI) or a validated internal reference standard against the said international standard.

The terms “patient” and “subject” are used interchangeably and are used in their conventional sense to refer to a living organism suffering from or prone to a condition that can be prevented or treated by administration of a composition of the present invention, and includes animals. The term“Animal” refers to a human or non-human animal, including, but not limited to, farm animals such as cattle, sheep, pigs, goats and horses; domestic mammals such as dogs and cats; laboratory animals including rodents such as mice, rats and guinea pigs; birds, including domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese and non-human primates, including, but not limited to, monkeys, chimpanzees and other apes and monkey species. The term does not denote a particular age. Thus, adult, juvenile and newborn individuals are of interest. The term“rabies virus” as used herein include rabies and rabies related viruses. According to the present invention, murine antibodies which can bind to site II or site III of G protein of any virus can be used for the treatment in which activity of said virus is detrimental.

The term“exposure” as used herein includes bites, scratches and licks by a rabid animal.

The term“Imogam ® ” as used herein is an approved HRIG available in the market. It is known to the person skilled in the art.

EMBODIMENTS OF THE INVENTION

In one embodiment, the present invention provides murine monoclonal antibody for the PEP of exposure with rabies and rabies-related viruses.

In a preferred embodiment, the present invention provides cocktail of at least two murine monoclonal antibodies for the PEP of exposure with rabies and rabies -related viruses.

In one of the embodiments, the present invention provides murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies which can be used at higher dose than the approved dose of HRIG or human monoclonal antibodies or humanized monoclonal antibodies. In a preferred embodiment, the present invention provides murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies which can be used at a dose more than 20 IU/kg. In a more preferred embodiment, the present invention provides murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies which can be used at a dose between 20 IU/kg to 100 IU/kg.

In another embodiment, the present invention provides murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies for the PEP of exposure with rabies and rabies-related viruses wherein the antibody or cocktail allows the active immunization to generate and maintain an effective immune response.

In one of the embodiments, the present invention provides combination of a murine monoclonal antibody and an anti-rabies vaccine for the PEP of exposure with rabies and rabies-related viruses. In a preferred embodiment, the present invention provides combination of cocktail of at least two murine monoclonal antibody and anti-rabies vaccine for the PEP of exposure with rabies and rabies-related viruses.

In one of the embodiments, the present invention provides use of murine monoclonal antibody for the PEP of exposure with rabies and rabies-related viruses.

In preferred embodiment, the present invention provides use of cocktail of at least two monoclonal antibodies for the PEP of exposure with rabies and rabies -related viruses.

In one embodiment, the current invention provides a monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses.

In one of the embodiments, the current invention provides a polynucleotide encoding the monoclonal antibody sequence.

In one of the embodiments, the current invention provides a monoclonal antibody comprising a CDRH1 comprising a polypeptide set forth in SEQ ID NO: l, a CDRH2 region comprising a polypeptide set forth in SEQ ID NO: 2, and a CDRH3 region comprising a polypeptide set forth in SEQ ID NO: 3.

In one of the embodiments, the current invention provides a monoclonal antibody comprising a CDRL1 comprising a polypeptide set forth in SEQ ID NO:4, a CDRL2 region comprising a polypeptide set forth in SEQ ID NO: 5, and a CDRL3 region comprising a polypeptide set forth in SEQ ID NO: 6.

In a preferred embodiment, the current invention provides a monoclonal antibody comprising:

(a) CDRH1 comprising a polypeptide set forth in SEQ ID NO: 1 ;

(b) CDRH2 comprising a polypeptide set forth in SEQ ID NO: 2;

(c) CDRH3 comprising a polypeptide set forth in SEQ ID NO: 3;

(d) CDRL1 comprising a polypeptide set forth in SEQ ID NO: 4; (e) CDRL2 comprising a polypeptide set forth in SEQ ID NO: 5; and

(f) CDRL3 comprising a polypeptide set forth in SEQ ID NO: 6.

In one embodiment, the current invention provides a monoclonal antibody comprising HCVR comprising a polypeptide set forth in SEQ ID NO: 7.

In one embodiment, the current invention provides a monoclonal antibody comprising LCVR comprising a polypeptide set forth in SEQ ID NO: 8.

In a preferred embodiment, the current invention provides a monoclonal antibody comprising: (a) a heavy chain variable region comprising a polypeptide set forth in SEQ ID NO: 7 and (b) a light chain variable region comprising a polypeptide set forth in SEQ ID NO: 8.

In one embodiment, the current invention provides a monoclonal antibody comprising heavy chain comprising a polypeptide set forth in SEQ ID NO: 9.

In one embodiment, the current invention provides a monoclonal antibody comprising light chain comprising a polypeptide set forth in SEQ ID NO: 10.

In a preferred embodiment, the current invention provides a monoclonal antibody comprising: (a) heavy chain comprising a polypeptide set forth in SEQ ID NO: 9 and (b) a light chain comprising a polypeptide set forth in SEQ ID NO: 10.

In one embodiment, the current invention provides a monoclonal antibody comprising heavy chain comprising a polypeptide set forth in SEQ ID NO: 11.

In one embodiment, the current invention provides a monoclonal antibody comprising light chain comprising a polypeptide set forth in SEQ ID NO: 12.

In a preferred embodiment, the current invention provides a monoclonal antibody comprising: (a) heavy chain comprising a polypeptide set forth in SEQ ID NO: 11 and (b) a light chain comprising a polypeptide set forth in SEQ ID NO: 12. In a preferred embodiment, the current invention provides a cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses.

In more preferred embodiment, the current invention provides a cocktail of two monoclonal antibodies mAh A and mAh B.

In another embodiment, the current invention provides an expression vector comprising the polynucleotide encoding the monoclonal antibody sequence.

In one embodiment, the current invention provides host cell transformed with the polynucleotide encoding the monoclonal antibody sequence.

In another embodiment, the current invention provides a method of producing the monoclonal antibody of the present invention by culturing the cell line.

In further embodiment, the current invention provides a composition comprising monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses.

In a preferred embodiment, the current invention provides a composition comprising cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses.

In a more preferred embodiment, the current invention provides a composition comprising cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses wherein one of the monoclonal antibodies comprising a CDRH1 comprising a polypeptide set forth in SEQ ID NO: l, a CDRH2 region comprising a polypeptide set forth in SEQ ID NO: 2, and a CDRH3 region comprising a polypeptide set forth in SEQ ID NO: 3 and/or comprising a CDRL1 comprising a polypeptide set forth in SEQ ID NO:4, a CDRL2 region comprising a polypeptide set forth in SEQ ID NO: 5, and a CDRL3 region comprising a polypeptide set forth in SEQ ID NO: 6.

In a further embodiment, the current invention provides a composition comprising cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses wherein one of the monoclonal antibodies comprising HCVR comprising a polypeptide set forth in SEQ ID NO: 7 and/or comprising LCVR comprising a polypeptide set forth in SEQ ID NO: 8.

In a furthermore embodiment, the current invention provides a composition comprising cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses wherein one of the monoclonal antibodies comprising heavy chain comprising a polypeptide set forth in SEQ ID NO: 9 and/or comprising light chain comprising a polypeptide set forth in SEQ ID NO: 10.

In one of the preferred embodiments, the current invention provides a composition comprising cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses wherein one of the monoclonal antibodies comprising heavy chain comprising a polypeptide set forth in SEQ ID NO: 11 and/or comprising light chain comprising a polypeptide set forth in SEQ ID NO: 12.

In a preferred embodiment, the current invention provides a composition comprising cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses wherein one antibody comprising heavy chain comprising a polypeptide set forth in SEQ ID NO: 9 and second antibody comprising heavy chain comprising a polypeptide set forth in SEQ ID NO: 11 and/or one antibody comprising light chain comprising a polypeptide set forth in SEQ ID NO: 10 and second antibody comprising light chain comprising a polypeptide set forth in SEQ ID NO: 12.

In another embodiment, the composition according to the present invention comprises cocktail of at least two monoclonal antibodies and pharmaceutical excipients. Cocktail according to the current invention is as described in above embodiments.

In one of the embodiments, the current invention provides a process of making antibody as claimed in any preceding claim by cell culture method comprising:

i) culturing the host cell expressing monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses; and ii) isolating and recovering monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses expressed in the said host cell.

In another embodiment, the current invention provides a process of purifying antibody as claimed in any preceding claim by suitable chromatography or purification technique.

In another embodiment, the present invention provides a kit comprising monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses.

In a preferred embodiment, the present invention provides a kit comprising cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses.

In one of the embodiments, present invention provides a method for diagnosing rabies using the monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses, preferably using cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses.

In another embodiment, the present invention provides a method of treating and preventing rabies using the monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses, preferably using cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses.

In one of the embodiment, the present invention is to provide a method of detecting rabies or rabies related viruses using the monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses, preferably using cocktail of at least two monoclonal antibodies capable of binding and neutralizing rabies or rabies related viruses.

DETAILED DESCRIPTION OF THE INVENTION

As described herein above, rabies is one of the most dangerous disease for human which causes death if not treated on time. A Post-exposure prophylaxis (PEP) regimen preferably involves two components - one providing active immunity and the other providing passive immunity. The active immunity is provided by a vaccine which works by activating the immune system of the patient/subject and generates immunity against the virus in question in a delayed manner i.e., only on or after seven days of the vaccination. The passive immunity is provided by readymade antibodies, such as polyclonal antibodies (HRIG, ERIG etc.), monoclonal antibodies or their cocktail, which provides anti-virus protection immediately after injection into the patient/subject. Since the passive immunization carries significant risk of neutralizing the active immunization of debilitating the PEP and putting the patient’s life at risk, a right balance of dose of passive immunization and type of passive immunization in terms of its circulating half-life in the patient is important in providing sufficient amount and type of neutralizing antibodies through passive immunization that provide protection/immunity in the early phase while allowing the vaccine (active immunization) to carry out its function uninterruptedly together providing an unbroken window of immune protection to the patient. So far passive immunization has largely been provided by conventional products namely, HRIG and ERIG which are used at 20 IU/kg and 40 IU/kg, respectively. HRIG and ERIG as described previously are unsafe because of the potential risk of carrying pathogens. Attempts at developing safer replacement products in the form of monoclonal humanized/human antibodies have led to products that are capable of providing only very low potency of antibodies as early passive immunization because at higher doses they end up neutralizing the active immunization. Thus there is a need of a passive immunization product which is safer than the conventional products and also provides comparable potency.

Present invention provides a murine monoclonal antibody or a cocktail of at least two murine monoclonal antibodies as the passive immunization component of the PEP of exposure with rabies or rabies related viruses along with a vaccine as active immunization component. A murine monoclonal antibody or a cocktail of at least two murine monoclonal antibodies according to the present invention can be given at a higher dose than approved dose of HRIG or human monoclonal antibodies or humanized monoclonal antibodies without significantly neutralizing the active immunization provided by an anti-rabies vaccine given as a part of PEP. The murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies according to the current invention can be given at a dose more than 20 IU/kg, preferably at a dose between 20 IU/kg to 100 IU/kg. Antibodies or cocktail of the present invention does not significantly neutralize the active immunization provided by an anti- rabies vaccine given as a part of PEP at a dose more than 20 IU/kg, preferably at a dose between 20 IU/kg to 100 IU/kg. Thus, a murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies according to the current invention, for PEP allows the active immunization to generate and maintain an effective immune response against rabies and rabies-related viruses. Vaccine according to the present invention is anti-rabies vaccine known in the art. The present invention provides a murine monoclonal antibody or cocktail of at least two murine monoclonal antibodies capable of binding and neutralizing rabies and rabies related viruses for the PEP of exposure with rabies and rabies -related viruses.

Monoclonal antibodies mAb A and mAb B :

A monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses according to the current invention comprises HCVR comprising a CDRH1 comprising a polypeptide set forth in SEQ ID NO: l, a CDRH2 region comprising a polypeptide set forth in SEQ ID NO: 2, and a CDRH3 region comprising a polypeptide set forth in SEQ ID NO: 3. A monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses according to the current invention comprises LCVR comprising a CDRL1 comprising a polypeptide set forth in SEQ ID NO: 4, a CDRL2 region comprising a polypeptide set forth in SEQ ID NO: 5, and a CDRL3 region comprising a polypeptide set forth in SEQ ID NO: 6.

A monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses according to the current invention comprises HCVR comprising a polypeptide set forth in SEQ ID NO: 7.

A monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses according to the current invention comprises LCVR comprising a polypeptide set forth in SEQ ID NO: 8.

A monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses according to the current invention comprises heavy chain comprising a polypeptide set forth in SEQ ID NO: 9.A monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses according to the current invention comprises light chain comprising a polypeptide set forth in SEQ ID NO: 10. A monoclonal antibody comprising polypeptides set forth in SEQ ID NO: 9 and 10 of heavy chain and light chain, respectively is referred herein as mAh A. mAh A comprising amino acid sequences of HCVR and LCVR as set forth in SEQ ID NO.s 7 and 8, respectively. Further, mAh A comprising amino acid sequences of CDRH1, CDRH2 and CDRH3 as set forth in SEQ ID NO.s 1, 2 and 3, respectively. Furthermore, mAh A comprising amino acid sequences of CDRL1, CDRL2 and CDRL3 as set forth in SEQ ID NO.s 4, 5 and 6, respectively.

A monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses according to the current invention comprises heavy chain comprising a polypeptide set forth in SEQ ID NO: 11.

A monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses according to the current invention comprises light chain comprising a polypeptide set forth in SEQ ID NO: 12.

A monoclonal antibody comprising polypeptides set forth in SEQ ID NO: 11 and 12 of heavy chain and light chain, respectively is referred herein as mAh B.

Preferably, mAh A and mAh B are murine antibodies according to the present invention. Further, the present invention provides a cocktail of two or more monoclonal antibodies. Preferably, the present invention provides a cocktail of at least two monoclonal antibodies i.e. mAh A and mAh B capable of binding and neutralizing rabies or rabies related viruses. Preferably, cocktail comprises equipotent amounts of antibodies capable of binding and neutralizing rabies or rabies related viruses. One of the two antibodies according to the current invention is a monoclonal antibody comprising amino acid sequences of HCVR and LCVR as set forth in SEQ ID NO.s 7 and 8, respectively. Second monoclonal antibody according to the current invention is a monoclonal antibody comprising an amino acid sequences of heavy chain and light chain as set forth in SEQ ID NO.s 11 and 12, respectively. Preferably, a cocktail according to the present invention comprising one monoclonal antibody having heavy chain and light chain comprising polypeptides set forth in SEQ ID NO.s 9 and 10, respectively and second monoclonal antibody having heavy chain and light chain comprising a polypeptides set forth in SEQ ID NO.s 11 and 12, respectively.

Monoclonal antibodies for a cocktail preparation are selected based on criteria, such as binding specificity, spectrum of neutralization of lyssaviruses, biological activity, neutralizing potency etc and industrial viability. Various lyssaviruses strains are isolated from specific species across the various geographical areas. For example, RABV (genotype 1) of dogs from Asia (India, Philippines, and Thailand etc.), Africa (North Africa, sub- Saharan Africa, etc.), and the New World and from mongoose(s) from South Africa were included.

Primarily, RABV (genotype 1) of dogs from Asia (India, Philippines, and Thailand etc), Africa (North Africa, sub-Saharan Africa, etc.), and the New World and, from mongoose(s) from South Africa were included. Furthermore, the neutralization capability of the antibodies was also tested against rabies related viruses, namely, EBLV-l, -2, (genotype 5, 6) ABLV (genotype 7), Duvenhage virus (genotype 4), Irkut virus, Khujand and Aravan.

Virus neutralization was undertaken with rabies and rabies related viruses CVS 11, SAD B19, PV, Kelev, EBLV1, EBLV2, ABV, Duvenhage, Irkut, Aravan, and Khujand. Virus neutralization was also undertaken with rabies and rabies related isolates from European Fox, Dog Turkey, Dog Ethiopia, Dog India, Dog Mexico, Wolf Sarajevo, Bobcat-USA, East European fox, Polar fox, Dog Azerbaijan, Dog Nepal, Raccoon, SE US, Gray fox TX, Arctic fox Alaska, Skunk SC US, Skunk CA.

Rapid fluorescent focus inhibition test (RFFIT) or fluorescent antibody virus neutralisation test (FAVN) assays were performed to study neutralization of broad spectrum of lyssaviruses strains as mentioned in examples provided herein below.

The antibodies are produced in an Expression System which may comprise of an Expression Vector and a host cell line. Any expression vector known to the skilled person can be used in the present invention, and the choice of the expression vector is dependent on the nature of the host cell of choice. Introduction of the vector in host cells can be effected by, but not limited to, calcium phosphate transfection, virus infection, DEAE dextran mediated transfection, lipofectamin transfection or electroporation, and skilled person can select and use an introduction method suitable for the expression vector and host cell used. The vector contains all essential elements required to express antibody(s). Preferably, the vector contains one or more selectable markers, but is not limited thereto, and a vector containing no selectable marker may also be used.

The present invention provides a host cell that comprises the expression vector transformed into a host cell to produce the monoclonal antibody of the present invention. In the present invention, the host cell may comprise cells of mammalian, plant, insect, fungal or bacterial origin, but is not limited thereto. A mammalian cell can be selected from, but is not limited thereto, CHO cells, F2N cells, CSO cells, BHK cells, Bowes melanoma cells, HeLa cells, 911 cells, AT1080 cells, A549 cells, HEK 293 cells and HEK293T cells. Suitable mammalian host cell known to skilled person can be used for the development of antibodies and their cocktail of the present invention. Preferably, host cell used to produce antibodies of the present invention is host cell derived from SP2/0.

A skilled person can also use another Expression System which comprises of fusion cells where one host cell containing nucleotide sequences expressing amino acid sequences of monoclonal antibodies of the present invention is fused with another host cell to develop the expression system. Method for producing an antibodies capable of binding and neutralizing rabies or rabies related viruses of the present invention comprising mainly i) culturing the host cell expressing monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses; and ii) isolating and recovering monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses expressed in the said host cell.

Culturing the host cell expressing monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses includes preferably production of monoclonal antibody by Sp2/0 cell culture process in suitable media which is known in the art. Expression of the monoclonal antibody occurs in a constitutive manner and secreted by the cells in soluble form.

Isolating and recovering monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses expressed in the said host cell include mainly cell clarification, affinity chromatography in combination with other suitable chromatography techniques and/or filtration techniques to prepare a purified bulk drug substance of monoclonal antibody of interest. The purified monoclonal antibody or its cocktail preparation is tested for neutralization of various lyssaviruses strains.

The present invention also provides a stable pharmaceutical composition comprising monoclonal antibody capable of binding and neutralizing rabies and rabies related virus, preferably a cocktail of at least two monoclonal antibodies and a pharmaceutically acceptable carrier or excipient. Preferred excipients are buffer, salt and surfactant. Suitable excipients to formulate and to prepare final bulk drug product can be selected by the skilled person. The pharmaceutical composition according to the current invention includes therapeutic potency of monoclonal antibody or a cocktail of at least two monoclonal antibodies. The said therapeutic potency is in the range from 1 IU/mL to 5000 IU/mL, preferably 100 IU/mL to 3000 IU/mL. The therapeutic potency according to present disclosure include each integer and non-integer number between the mentioned ranges i.e. 1 IU/mL to 5000 IU/mL. For Example, the said range includes 10 IU/mL, 25 IU/mL, 50 IU/mL, 100 IU/mL, 150 IU/mL, 200 IU/mL, 300 IU/mL, 400 IU/mL, 500 IU/mL, 600 IU/mL, 700 IU/mL, 800 IU/mL, 900 IU/mL, 1000 IU/mL, 1200 IU/mL, 2000 IU/mL, 3000 IU/mL, 4000 IU/mL and 5000 IU/mL. Cocktail of antibodies according to the current invention may include therapeutic potency of each monoclonal antibody in the described therapeutic range. Preferably, cocktail of antibodies includes therapeutic amount, preferably equipotent amount of each monoclonal antibody in the described therapeutic range. The preferred therapeutic potency of antibody according to the present invention is between 100 IU/mL - 3000 IU/mL. In one of the embodiments, preferred therapeutic potency of cocktail according to the present invention is between 400 IU/mL - 1500 IU/mL. More preferably, therapeutic potency of antibody according to the present invention is 150 IU/mL or 300 IU/mL or 600 IU/mL. More preferably, therapeutic potency of cocktail according to the present invention is 300 IU/mL or 600 IU/mL or 1200 IU/mL. The preventive and therapeutic composition of the present invention is sterile and stable under the conditions of manufacture and storage. It can be in solution and an appropriate pharmaceutically acceptable excipient can be added and/or mixed before or at the time of delivery to provide a unit dosage injectable form. Alternatively, the composition of the present invention can be in lyophilized form which can be reconstituted in an appropriate pharmaceutically acceptable excipient before or at the time of delivery. In the case of sterile powders for the preparation of sterile injectable solutions, preferred preparation method is freeze-drying. The “lyophilized or freeze-drying composition” is a dosage form which is prepared by lyophilization or freeze drying process. The lyophilization was performed with conventional lyophilization technique known in the literatures involving steps such as freezing, annealing, primary drying and secondary drying.

A composition of monoclonal antibody capable of binding and neutralizing rabies or rabies related viruses, preferably cocktail of at least two monoclonal antibodies according to the present invention can be used for treatment and prevention of rabies, preferably for post-bite prophylaxis. The monoclonal antibodies or their cocktail according to the present invention is studied to check rabies virus neutralization activity. The potency of the rabies or rabies related viruses neutralizing antibodies is determined by RFFIT in comparison with an‘Anti- rabies Immunoglobulin, Human WHO International Standard’ (RAI) or a validated internal reference standard against the international standard. RFFIT assay is useful for the determination of antibodies against the rabies or rabies related viruses in both early phase (passive immunization) and the late phase (active immunization).

The routes of administration of the preventive and therapeutic composition of the present invention can be divided into oral and parenteral routes. The preferred route of administration is infiltration in to and around the site of exposure. Optionally, remaining volume is injected intramuscularly at site(s) distant from the wound site, but is not limited thereto.

The monoclonal antibody and cocktail of at least two monoclonal antibodies was found to have the ability to neutralize various rabies or rabies related viruses and thus is useful for the treatment and prevention of rabies in the patients and animals infected with rabies or rabies related viruses. Preferably, monoclonal antibody and cocktail of two monoclonal antibodies can be administered directly in the surrounding area of the exposure upon bite by a rabid animal. Optionally, remaining volume was administered in the different parts of the body intramuscularly. Preferably, cocktail of antibodies comprising mAh A and mAh B of the present invention.

The dose of the preventive and therapeutic antibodies of the present invention is selected from 1 IU/Kg to 100 IU/Kg. Preferred range of the dose of the preventive and therapeutic antibodies of the present invention is 10 IU/Kg to 80 IU/Kg. More preferably, dose of the preventive and therapeutic antibody of the present invention is 40 IU/Kg. In one of the embodiments, dose of the preventive and therapeutic cocktail according to the present invention is selected from 1 IU/Kg to 100 IU/Kg. Preferably, dose of the preventive and therapeutic cocktail according to the present invention is selected from 10 IU/Kg to 80 IU/Kg. More preferably, dose of the preventive and therapeutic cocktail according to the present invention is 40 IU/Kg.

The dose regime of the preventive and therapeutic antibodies of the present invention include an administration of antibody of the current invention in conjunction with anti-rabies vaccine. Anti-rabies vaccine according to the present invention can be anti-rabies vaccines available in market or under clinical development. For example, anti-rabies vaccine according to the present invention can be selected from Imovax, Rabavert, Rabipur, Rabivax, Vaxirab N, Abhayrab, Indirab, Verorab, SPEEDA, etc. These are the brand names of the anti-rabies vaccines available in the market. In a preferred embodiment, antibody of the present invention is administered without delay as an emergency procedure. It can also be injected up to 7 days after administration of first vaccine dose. In a preferred embodiment, the antibody or cocktail of the present invention can be used for the PEP of exposure with rabies and rabies-related viruses. More preferably, the antibody or cocktail of the present invention is used in conjunction with anti-rabies vaccine for the PEP of exposure with rabies and rabies-related viruses. Analytical methods used in the present invention:

Rapid fluorescent focus inhibition test (RFFIT) for the potency and RVNA titer determination. The RFFIT assay was carried out in 96-well tissue culture plates. A fixed dose of CVS-l l was incubated with each of the 1:2 serial dilutions of the test and the reference materials. After 90 minutes of such incubation, the test and reference materials along with the CVS-l l virus were added over BHK-21 cells and incubated for a further 22 ± 2 hours in a 5% CO 2 incubator at 37°C. On completion of the incubation period, the cell monolayer was washed and fixed with 80% acetone. The presence of non-neutralized virus at each serial dilution is detected by identifying cells infected by residual virus using a FITC conjugated antibody against the rabies virus nucleoprotein and counting the fluorescing infected cells under a fluorescent microscope. The RVNA titre was calculated as shown below: Calculation of RVNA Titer

Here, the present invention is illustrated with the following non-limiting examples which should not be interpreted as limiting the scope of the invention in any way.

Examples

Example 1: Manufacturing of monoclonal antibody mAb A active drug substance

Drug substance manufacturing process of mAh A monoclonal antibody involves -

- Upstream Sp2/0 cell culture process and

- Downstream purification process. Upstream culture process to produce mAb A monoclonal antibody

mAh A monoclonal antibody was produced by host cell of the expression system in Sp2/0 cell culture process in the presence of animal origin free media and feed components. Serum components containing media can also be used to produce the said antibody. Each batch production in upstream was initiated with the revival of a master cell bank (MCB) or Working Cell Bank (WCB) vial content of Sp2/0 cell harboring the mAh A polynucleotide sequence. Following revival of the cell bank vial, a series of seed development steps was performed to generate the final inoculum with adequate number of cells and inoculated into the production bioreactor. The cell culture process in bioreactor was conducted in a well- controlled manner with various on-line and off-line control parameters to express the mAh A monoclonal antibody, in a constitutive manner. Protein was secreted by the cells in soluble form.

Multiple batches of a composition comprising the monoclonal antibody were produced. An inoculum of the mAh A was cultured in 200L bioreactors containing a liquid cell culture medium. The mature antibody encoded by these nucleic acids is mAh A and comprises the heavy chain amino acid sequence set forth in SEQ ID NO:9 and the light chain amino acid sequence shown in SEQ ID NO: 10. Cell culture was performed in a controlled environment by maintaining pH 7.0 ± 0.3 using C02 gas and /or sodium bicarbonate, as and when required. The dissolved oxygen concentration was maintained at 15 ± 5% saturation with sparging of air and/or oxygen gas and by controlling agitation speed in the bioreactor. Bioreactors were operated at a temperature of about 36.0° C to about 37.0° C.

Growth media contains following components: Components Concentration per liter

BD CELLTM MAB ACF 12.860 gm

Cells were grown under the above-mentioned conditions for two days from day 2, feeding was initiated and continued until the end of batch. Following media components were fed to the cell culture medium as common feed - Components Concentration per liter

L-Glutamine 2 irM daily from Day 2 onwards until the day before harvest

Concentrated growth media 2% Feed (v / v) with respect to the initial working volume in bioreactor from Day 3 onwards until the day before harvest

The culture duration was about 7 to 9 days. The in vitro cell age (culture days from initial thaw of master cell bank to harvest) was 26 days or less. After this, the clarified cell culture supernatant was harvested by centrifugation and depth filtration of the cell culture medium. This clarified supernatant was then reconditioned to match substantially to the next purification column equilibration conditions. Supernatant was then subjected to protein a chromatography and impurities were allowed to flow off this chromatography column.

Downstream purification process to obtain the mAh A monoclonal antibody drug substance Purification process of mAh A monoclonal antibody was carried out with several unit operations comprising cell clarification, reconditioning, chromatography, viral inactivation, virus clearance and membrane ultrafiltration-diafiltration. Purification by chromatography involved various interaction chromatography techniques, like affinity chromatography and other suitable chromatography and filtration techniques. Viral inactivation was conducted at low pH condition for a certain period of time and virus clearance was performed by nano filtration. Reconditioning and/or buffer exchange was carried out by conventional membrane ultrafiltration-diafiltration.

At the end of purification process, the purified mAh A monoclonal antibody was filtered through a 0.2 pm filter, aseptically, and stored under frozen condition in suitable container- closure system. The entire purification process was monitored and controlled with appropriate physical and physicochemical parameters related to mAh A monoclonal antibody and its purification process. When assessed by analytical high-performance size exclusion chromatography, the mAh A drug substance was found to show greater than 99 % purity. Example 2: Manufacturing of mAb B monoclonal antibody active drug substance

Drug substance manufacturing process of mAh B monoclonal antibody involves -

- Upstream Sp2/0 cell culture process and

- Downstream purification process.

Upstream culture process to produce mAb B monoclonal antibody

mAh B monoclonal antibody was produced by host cell of the expression system Sp2/0 in cell culture process in the presence of animal origin free media and feed components. Serum components containing media can also be used to produce the said antibody. Each batch production in upstream was initiated with the revival of a master cell bank (MCB) or Working Cell Bank (WCB) vial content of Sp2/0 cell harboring the polynucleotide sequence of mAh B. Following revival of the cell bank vial, a series of seed development steps was performed to generate the final inoculum with adequate number of cells and inoculated into the production bioreactor. The cell culture process in bioreactor was conducted in a well- controlled manner with various on-line and off-line control parameters to express the mAh B monoclonal antibody, in a constitutive manner. Protein was secreted by the cells in soluble form.

Multiple batches of a composition comprising the monoclonal antibody were produced. An inoculum of the mAh B was cultured in 200L bioreactors containing a liquid cell culture medium. The mature antibody encoded by these nucleic acids is mAh B and comprises the heavy chain amino acid sequence set forth in SEQ ID NO: 11 and the light chain amino acid sequence shown in SEQ ID NO: 12. Cell culture was performed in a controlled environment by maintaining pH 7.0 ± 0.3 using C02 gas and /or sodium bicarbonate, as and when required. The dissolved oxygen concentration was maintained at 30 ± 10% saturation with sparging of air and/or oxygen gas and by controlling agitation speed in the bioreactor. Bioreactors were operated at a temperature of about 36.0° C to about 37.0° C.

Growth media contains following components:

Components Concentration per liter

BD CellTM MAb ACF 12.860 gm Cells were grown under the above-mentioned conditions for two days. From day 2, feeding was initiated and continued until the end of batch. Following media components were fed to the cell culture medium as common feed -

Growth media contained following components:

Components Concentration per liter

L- Glutamine 2 mM daily from Day 2 onwards until the day before harvest

Feed media Cell Boost 7a 1% Feed (v / v) with respect to the initial working volume in bioreactor from Day 3 onwards until the day before harvest

Feed media Cell Boost 7b 0.3% Feed (v / v) with respect to the initial working volume in bioreactor from Day 3 onwards until the day before harvest The culture duration was about 5 to 7 days. The in vitro cell age (culture days from initial thaw of master cell bank to harvest) was 33 days or less. After this, the clarified cell culture supernatant was harvested by centrifugation and depth filtration of the cell culture medium. This clarified supernatant was then reconditioned to match substantially to the next purification column equilibration conditions. Supernatant was then subjected to protein A chromatography and impurities were allowed to flow off this chromatography column.

Downstream purification process to obtain the mAh B monoclonal antibody drug substance Purification process of mAh B monoclonal antibody was carried out with several unit operations comprising cell clarification, reconditioning, chromatography, viral inactivation, virus clearance and membrane ultrafiltration-diafiltration. Purification by chromatography involved various interaction chromatography techniques, like affinity chromatography and other suitable chromatography and filtration techniques. Viral inactivation was conducted at low pH condition for a certain period of time and virus clearance was performed by nano filtration. Reconditioning and/or buffer exchange was carried out by conventional membrane ultrafiltration-diafiltration. At the end of purification process, the purified mAh B monoclonal antibody was filtered through a 0.2 pm filter, aseptically, and stored under frozen condition in suitable container- closure system. The entire purification process was monitored and controlled with appropriate physical and physicochemical parameters related to mAh B monoclonal antibody and its purification process. When assessed by analytical high-performance size exclusion chromatography, the mAh B drug substance was found to show greater than 99 % purity.

Example 3: Preparation of antibody cocktail comprising equipotent antibodies

To prepare an antibody cocktail comprising equipotent amount of two antibodies, a potency value in IU/mg was assigned to the purified drug substance of mAbs A and mAbs B by undertaking a RFFIT test. Individual drug substances mAbs A and mAbs B were diluted based on the concentration (mg/mL) to arrive at a similar potency value in IU/mL by citrate buffer of pH 6.0 containing sodium chloride and polysorbate. To prepare an antibody cocktail comprising equipotent antibodies, both the diluted drug substance of mAbs A and mAbs B were suitably mixed.

Example 4 Manufacturing of monoclonal antibody cocktail

Antibody cocktail was prepared by mixing equipotent amounts of the mAh A and mAh B monoclonal antibody drug substances in the presence of citrate buffer of pH 6.0 containing sodium chloride and polysorbate. Compounding was performed in a step-wise manner in a suitable vessel. pH of the solution at different stages of compounding was controlled and mixing of solution was conducted thoroughly. Upon completion of compounding, an aliquot of the formulated bulk was withdrawn and analyzed for pH and osmolality. For bioburden reduction and sterile filtration of the final formulated bulk, 0.2 pm sterilizing grade filters were used, prior to filling in to the glass vials or syringes. Filter integrity was tested before (pre) and after (post) filtration of the bulk. Filling was performed by using a pump and fill volume was checked by weight throughout the entire filling process. On-line stoppering of the filled-in primary containers was conducted, simultaneously, under sterile conditions. After stoppering, flip-off sealing was carried out. The final product thus prepared provides a stable pharmaceutical composition which was found to remain stable for at least 36 months under Real-Time temperature storage conditions in the specified container-closure system. Example 5: In vitro studies of mAb A and mAb B

In-vitro cross-reactivity of the selected mAbs candidates was measured on a selection of RABV and rabies-related viruses isolated from specific host species and geographical areas in various sets of experiments. Primarily, RABV (genotype 1) of dogs from Asia (India, Philippines, and Thailand etc.), Africa (North Africa, sub-Saharan Africa, etc.) and the New World were included. Also, RABV (genotype 1) from mongoose(s) from South Africa were included. Furthermore, the neutralization capability of the mAbs (mAb A and mAh B) against rabies related viruses, such as EBLV-l, EBLV -2, (genotype 5, 6) ABLV (genotype 7), Duvenhage virus (genotype 4) isolated species including Irkut virus, Khujand and Aravan were tested.

Monolayers of NA cells were infected with selected RABV and rabies-related virus strains at a multiplicity of infectivity of 0.1 for 1 h at 37°C in 5% C0 2 . The virus inoculum was then removed, fresh medium was added to the cells, and cells were subsequently incubated for 72 h at 37°C in 0.5% C0 2 . Subsequently, the culture supernatants were collected and titrated on BHK 21 cells. Up to three passages were conducted to obtain sufficient virus titres. Viruses were stored at -80 °C until further use.

Rapid fluorescent focus inhibition test (RFFIT) or fluorescent antibody virus neutralisation test (FAVN) assays were performed on BHK 21 cells using a constant virus and varying mAbs method approach. Each mAbs (purified or supernatant) was serially diluted (10.0 - 0.03 IU/ml) and incubated with 10 4 FFU/ml of the RABV and rabies-related viruses to be tested. To determine the neutralizing potency of each mAbs, 100% reduction of the test virus by mAh A and mAh B was compared to the 100% reduction of the same test virus by an international standard rabies immunoglobulin (SRIG, 2 nd human rabies immunoglobulin preparation, National Institute for Biological Standards and Control, Potters Bar, UK). SRIG, used herein is a human derived rabies immunoglobulin which can also be mentioned as HRIG. The test was read after 24 hours of incubation.

5.1 In vitro study of virus neutralization by the selected mAbs

Table: 1 Neutralisation pattern of Mab A and SRIG using 17 RABV and rabies related viruses. Closed (grey) boxes represent presence of viable virus

# closed boxes represent presence of viable virus

Table:2 Neutralisation pattern of Mab B and SRIG using 17 RABV and rabies related

viruses. Closed (grey) boxes represent presence of viable virus

# closed boxes represent presence of viable virus

Both the mAbs were tested as a re-suspended purified antibody starting from a potency of 10 IU/mL up to a dilution of 0.03 IU/mL. Additionally, the antibodies were found to be complementary in their neutralization potential. The test demonstrated that the antibodies selected was comparable to SRIG with respect to the breadth of virus neutralization.

5.2 In vitro study of virus neutralization by the selected mAbs

In the in vitro testing assays, seven different RABV (2 laboratory strains and 5 RABV field strains) were used to determine the neutralisation potency of each mAh. Furthermore, 4 different rabies-related viruses representing genotypes 4, 5 and 6 as well as 4 isolates of bat lyssaviruses from Central Asia and East Siberia [Australian bat virus (ABV), Aravan virus (ARAV), Khujand virus (KHUV) and Irkut virus (IRKV)] were included in the virus panel.

Table: 3 Neutralisation pattern of SRIG, mAh A and B using RABV and rabies related viruses

# closed boxes represent presence of viable virus

There is a broad spectrum in-vitro cross-reactivity of the candidate mAbs against rabies and some other lyssaviruses. All the rabies and rabies related viruses were neutralized by either of the monoclonal antibodies. Additionally, the antibodies were found to be complementary in their neutralization potential. The test demonstrated that the antibodies selected were comparable to SRIG with respect to the breadth of virus neutralization. The data demonstrated similarity between the antibodies and SRIG in their ability to neutralize the different rabies and RABV like lyssaviruses. 5.3 In vitro study of virus neutralization by cocktail of mAb A and mAb B, HRIG and

ERIG

Experiment was conducted against street rabies virus isolates and the CVS strain.

Table 4: Neutralisation pattern of mAh cocktail, HRIG and ERIG

The cocktail of mAbs A and B was found to neutralize all the isolates similar to the neutralization by HRIG (Human rabies immunoglobulin preparation) and ERIG (Equine rabies immunoglobulin preparation). Example 6: In vivo potency studies of mAb cocktail

6.1 In vivo potency study of mAb cocktail, ERIG and HRIG

In this study, mice was used as animal model. Virus challenged animals were divided in 5 study groups: 1) Control group injected with normal saline; 2) injected with 0.2 IU/mice mAh cocktail; 3) injected with 0.4 IU/mice mAh cocktail; 4) injected with 0.2 IU/mice HRIG; 5) injected with 0.4 IU/mice ERIG.

In this study, CVS virus and 10 strains of street virus were used to challenge the animals. The mAh cocktail was administered at two doses 0.2 IU/mice and 0.4 IU/mice in order to compare against a similar dose of HRIG and ERIG. The dilution of the mAh to be tested was calculated on the basis of recommended doses of HRIG (20 IU/Kg) and ERIG (40 IU/Kg). Each mice was injected with HRIG at 0.2 IU/mice in group 4 and ERIG at 0.4 IU/mice in group 5. mAh cocktail was tested on both the doses 0.2 IU/mice and 0.4 IU/mice in group 2 and in group 3, respectively . Each group having 8 mice were challenged with 100 LD 50 of each mentioned virus. After 1 hour of virus challenge, control group received 100 m L of normal saline inoculated on the same site as the virus inoculation. Rest of the four groups received one of the dilution of mAbs infiltered in the same area where virus was inoculated. The mice were kept under observation for 1 month.

Table: 7 Animal study of mAh cocktail, ERIG and HRIG in virus challenged mice

Results provided in table 7 shows that survival rate of 100% was observed in study group treated with anti-rabies monoclonal antibody (HRIG, ERIG and mAh cocktail). While in the control group challenged with CVS, 100% mortality was observed.

It also proves that mAh cocktail of the present invention is as potent as currently approved antibodies (HRIG and ERIG).

6.2 In vivo potency study of mAb cocktail and HRIG

In order to establish PK/PD correlation for appropriate design of clinical study in human, the in-vivo efficacy study in rabies virus (CVS-l 1) challenged hamsters was performed.

The efficacy determination was based on the number of hamsters surviving a lethal dose of rabies virus, upon treatment with the equal dose of the test sample mAh cocktail and the standard HRIG (Kamrab, Kamada Ltd., Beit-Kama, Israel).

Study animals were randomized based on their body weight and divided in different treatment groups. Virus challenged animals were divided (10 animals in each group) into the following three groups: 1) a group injected with CVS- 11 virus alone; 2) a group injected with CVS- 11 virus and Anti rabies mAbs cocktail individually; 3) ) a group injected with CVS-l l virus and standard HRIG (Kamrab, Kamada Ltd., Beit-Kama, Israel). Rabies virus preparation with a known LD50 titer was suitably diluted and 200 m L administered into the right gastrocnemius muscle of each hamster. The formulated solution of the Anti-rabies mAbs cocktail and the standard Kamrab were administered at 6 hours into defined groups at the same site of virus injection in fed conditions. The dose level selected was 15 IU/hamster to minimize the dose volume at injection site (-100 pL). Hamsters were observed daily for any clinical signs of rabies such as paralysis or death. Upon observation of any signs of rabies virus infection, the animals were euthanized by carbon dioxide asphyxiation. % survival was checked after 30 days from virus challenge. The results of the study are shown in table 8. Table: 8 Animal study of mAh cocktail and HRIG dosing without vaccine after 6 hours of CVS- 11 virus challenge

The in vivo efficacy study of mAh cocktail in comparison with HRIG (Kamrab, Kamada Ltd., Beit-Kama, Israel) was undertaken to demonstrate the virus neutralization potential of mAh cocktail in rabies virus challenged hamsters. In this study, where animals were treated 6 hours after virus challenge showed 90% survival by mAh cocktail of the present invention. It is better than 50% survival by HRIG (Kamrab, Kamada Ltd., Beit-Kama) Example 7: Phase III clinical study of mAb cocktail

The efficacy of a cocktail comprising Ah A and mAh B was studied in a randomized, multi-centric, open-label, comparator-controlled, phase III human clinical study to evaluate and compare the efficacy and safety of the cocktail with an HRIG in a PEP setting, where, both were administered in conjunction with an anti-rabies vaccine.

The study comprised of 308 subjects with WHO Category III exposure(s) from suspected rabid animals that were divided equally into two arms. In one arm, subjects were given mAh cocktail (40 IU/kg) in conjunction with an anti -rabies vaccine (VaxiRab N; Cadila Healthcare Ltd. Ahmedabad, India) and in the other arm, an HRIG (Imogam ® (20 IU/kg, Sanofi Pasteur SA France) with anti-rabies vaccine.

In both arms the drugs being compared were administered directly in the surrounding area of the wounds from the bite of a rabid animal (single site/multiple sites) and remaining volume was administered in the different parts of the body intramuscularly. Both drugs were administered on day 0 in conjunction with an anti-rabies vaccine as per Essen regimen for rabies vaccination (five intramuscular doses on days 0, 3, 7, 14 and, 28). The study was followed for 84 days for each subject.

Results:

Both the drugs, Mab cocktail and HRIG, were found to be at least comparable in terms of RVNA titers as assessed at both the early time -points (days 3 and 7), and late time -points (days 14, 28, 42, and, 84). The study demonstrated that the mAh cocktail like HRIG, provided an unbroken window of protection i.e., without significantly neutralizing the anti rabies vaccine throughout the study duration. Further, the late time-point titers as observed on days 28, 42 and 84 were higher in the mAh cocktail arm as compared to HRIG, confirming that the murine mAh cocktail neutralized the vaccine (active immunization) lesser than the human-derived polyclonal (HRIG), demonstrating the advantage of murine monoclonal antibodies over human antibodies in the PEP setting. Incorporation by reference

The entire disclosure of each of the patent documents and scientific articles referred to herein is incorporated by reference for all purposes.

Equivalents The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The foregoing embodiments are therefore to be considered in all respects illustrative rather than limiting the invention described herein. Scope of the invention is thus indicated by the appended claims rather than by the foregoing description, and all changes that come within the meaning and range of equivalency of the claims are intended to be embraced therein.