Title of Invention

ANTI-RHD MONOCLONAL ANTIBODY

Abstract Anti-RhD monoclonal antibodies and methods for the production thereof
Full Text FORM 2
THE PATENTS ACT, 1970
(39 OF 1970)
AND
THE PATENT RULES, 2003
PROVISIONAL SPECIFICATION
(See section 10; rule 13)
Title of the Invention:


93
"Anti-RhD Monoclonal Antibodies
Applicant(s):
(a) Name BHARAT SERUMS AND VACCINES LTD.
(b) Nationality An Indian company Incorporated under the
Companies Act 195S,
(c) Address- 16th and 17th Floor, Hoechst House,
Nariman Point Mumbai - 400 021. Maharashtra, India.


The present invention relates to the production and use of anti-Rhesus D mononclonal antibodies and antigen binding fragments thereof.
Background and prior art
5
Rhesus D antigen (also referred to in the art as RhD antigen, Rhesus
factor, and/or Rh factor) is an antigen which may be present on the surface of
human red blood cells. Those individuals whose red blood cells have this
antigen are usually referred to as "RhD-positive", while those individuals whose
10 red blood cells do not have this antigen are referred to as "RhD-negative".
A person who is RhD-negative and has never been exposed to the RhD antigen will not produce anti-RhD antibodies (antibdoies against the RhD antigen). However, transfer of RhD-positive blood to a RhD-negative individual
15 will lead to sensitisation (immunization) of the RhD-negative individual against the RhD antigen. This can lead to a number of complications. In particular, where a RhD-negative woman gives birth to a RhD-positive infant there is a risk of small amounts of the infant's blood entering the maternal circulation, causing the the mother to produce anti-RhD antibodies. Whilst this will not normally harm
20 the first baby, should the now immunized mother fall pregnant with another RhD positive child then maternal anti-RhD antibodies may cross the placenta and attack the infant's blood cells, leading to a condition known as haemolytic disease of the newborn (HDN).
25 Anti-RhD antibodies are therefore routinely administered to RhD-
negative patients where there is a risk of exposure to RhD-positive blood, in order to prevent the patient from becoming immunized against the RhD-positive blood. For example, a RhD-negative patient may be given anti-RhD antibodies: prior to and/or shortly after giving birth to or having an abortion of an RhD-positve
30 baby; after any incident during pregnancy which may have lead to bleeding across the placenta; as a routine preventative measure during pregnancy; or prior to or soon after any transfusion of blood components containing RhD-positive red blood cells.

Traditionally, the anti-RhD antibodies used have been polyclonal antibodies obtained from the blood plasma of RhD negative volunteers who have been repeatedly immunized against RhD-positive red blood cells. However, the use of polyclonal antibodies has a number of recognised drawbacks, not least of 5 which are the continuing need for a number of volunteer donors sufficient to meet the demand for antibody, and the risk of contamination of the antibody preparation with any viruses or other pathogens that may be present in the donor's blood.
10 Whereas polyclonal antibodies constitute antibodies secreted by a
number of different plasma cells, and thus constitute a mixture of immunoglobulin molecules secreted against a specific antigen and potentially recognising a variety of epitopes, monoclonal antibodies are produced from cells that are all clones of a single parent cell, and thus constitute a homogeneous population of
15 antibodies, as is well known in the art. The cell lines from which monoclonal antibodies are produced are developed and cultured in-vitro, and this means monoclonal antibodies have the potential to be produced as and when required both in large amounts and at high levels of purity. Accordingly, monoclonal anti-RhD antibodies have a number of potential advantages over the polyclonal anti-
20 RhD antibody preparations that have traditionally been used.
A number of techniques for producing human monoclonal antibodies in general, and human monoclonal anti-RhD antibodies in particular, have been described. For example, EP-A2-0251440 discloses an anti-RhD monoclonal 25 antibody producing heterohybridoma formed by fusion of non-lg secreting mouse myfenoma cells with an anti-RhD Ig producing population of Epstein Barr virus (EBV) transformed human lymphocytes.
US 5,665,356 describes the production of human monoclonal anti-RhD 30 antibodies having certain defined characteristics, produced by culturing selected EBV-transformed human B-lymphocytes.
US 6,312,690 describes the production anti-RhD monoclonal antibodies by recombinant techniques. An EBV immortalized human cell line producing an

anti-Rhesus D monoclonal antibody called D7C2 was selected. The sequences encoding the variable regions of the heavy (H) and light (L) chains of D7C2 were cloned, sequenced, and inserted into a recombinant baculovirus expression vector under the control of a strong baculovirus promoter. Insect ceils 5 transfected with the recombinant baculovirus were cultured, and the recombinant D7C2 monoclonal antibody recovered from the cell supernatant.
US-A1-2003/0175969 describes a method for preparing a anti-RhD monoclonal antibodies capable of activating effector cells expressing FcyRIII,
10 comprising: a) purifying monoclonal antibodies obtained from cell lines selected from human B lymphocyte heterohybridomas, or recombinant animal or human cell lines (such as CHO-K, CHO-LedO, CHO Lec-1, CHO Pro-5, CHO dhfr-, Wii-2, Jurkat, Vero, Molt-4, COS-7, HEK293, YB2/0, BHK, K6H6, NSO, SP2/0-Ag 14 and P3X63Ag8.653 cells); b) adding each antibody obtained in step a) to a
15 different reaction mixture comprising RhD-positive red blood cells, effector cells comprising cells expressing FcyRIII, polyvalent IgGs; and c) determining the percentage lysis of the target cells and selecting the monoclonal antibodies which activate the effector cells causing significant lysis of the RhD-positive red blood cells.
20
US 6,475,787 discloses a method for preparing monoclonal antibodies, in which a suitable eukaryotic host cell is transformed with a DNA sequence encoding an antibody heavy chain and a DNA sequence encoding an antibody light chain, the two sequences being (inked to different amplifiable marker genes
25 so as to allow differential amplification of the heavy and light chain DNAs in order to optimize the relative gene copy numbers of the heavy and light chain DNAs. In a preferred embodiment the host cell is a Chinese Hamster Ovary (CHO) cell which is DHFR deficient (i.e. incapable of producing dihydrofolate reductase), one of the amplifiable marker genes is an adenosine deaminase (ADA) gene,
30 and the other is a DHFR gene. Amplification of the DNA encoding one antibody chain and linked in the ADA gene can then be achieved by treating the recombinant cells with increasing concentrations of 2'-deoxycoformycin, whilst amplification of the DNA encoding the other antibody chain and linked in the

DHFR gene is achieved by treating the ceff with increasing concentrations of methotrexate (MTX).
Nevertheless, there remains a need for further anti-RhD monoclonal 5 antibodies and methods for the production thereof.
Description of the Invention
According to a first aspect of the present invention there is provided an 10 isolated anti-RhD monoclonal antibody comprising:
a) a heavy chain variable region having first, second and third CDRs
(complementarity determining regions) which are identical or substantially
identical to the respective first, second, and third CDRs of SEQ ID NO: 2, and a
light chain variable region having first, second and third CDRs which are identical
15 or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4; or
b) a heavy chain variable region having first, second and third CDRs
which are identical or substantially identical to the respective first, second, and
third CDRs of SEQ ID NO: 6, and a light chain variable region having first,
20 second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 8; or
c) a heavy chain variable region having first, second and third CDRs
which are identical or substantially identical to the respective first, second, and
third CDRs of SEQ ID NO: 10, and a light chain variable region having first,
25 second and third CDRs which are identical or substantially identical to the respective first, second, and third CORs of SEQ ID NO; 12.
As used herein, the term "anti-RhD antibody" refers to both whole 30 antibodies and to fragments thereof that have binding specificity for RhD antigen. The binding affinity/specificity of an antibody can be measured by a various assays, as will be known to and can be routinely implemented by one of ordinary skill in the art. For example, antibodies recognising and specifically binding to RhD antigen can be determined using one or more standard techniques as

known to one of ordinary skill in the art, such as but not limited to: EIA / ELISA techniques, such as competitive EIA (enzyme linked-immunoassay); flow cytometry; and/or ADCC (antibody-dependant cellular toxicity) assays. Exemplary competitive EIA, flow cytometry, and ADCC techniques are described 5 in further detail in the Examples that follow.
As is well known in the art, whole antibodies are typically formed of one or two heavy and one or two light chains. The heavy and light chains each comprise a variable region and a constant region. The variable regions (also
10 referred to as the variable domains) dictate the antibody's antigen binding specificity. Each variable domain is composed of complementarity determining regions (CDRs, of which there are typically three, designated CDR1, CDR2 and CDR3) interspersed with more conserved regions known as framework regions. On folding of the antibody to adopt the correct quaternary structure, the CDRs of
15 a heavy and light chain together form the antigen binding site. The constant region of the heavy chain is composed of three or more constant domains and is dependent on the class (eg. IgA, IgD, IgE, IgG, or IgM) and isotype (eg. lgA1, lgA2, lgG1, lgG2, lgG3, lgG4) of the antibody. It is identical in all antibodies of the same class and isotype, but differs in antibodies of different isotypes. The
20 light chain constant region is composed of a single constant domain of which is of one of two isotypes, kappa or lambda, and is likewise identical in all antibodies of the same isotype. The constant regions of the antibodies typically mediate binding of the antibody to host tissues or factors.
25 Antibody fragments according to the present invention typically include at
least the CDRs and sufficient of the framework regions to specifically bind the antigen. Exemplary types of fragment include, but are not limited to, a Fab' fragment (consisting of the variable domain and a constant domain of both the light and heavy chains), a F(ab')2 fragment (two Fab' fragments linked by a
30 disulfide bridge at the hinge region), a Fv fragment (consisting of the variable domains only of the light and heavy chains), and other types of fragment as known to one skilled in the art.
SEQ ID NOs: 2 and 4 are the amino acid sequences of the heavy and light chains of the anti-RhD monoclonal antibody referred to herein as RhD1 and

described befow in further detail SEQ fD NOs: 6 and 8 are the amino acid sequences of the heavy and fight chains of the anti-RhD monoclonal antibody referred to herein as RhD2 and described below in further detail. SEQ ID NOs: 10 and 12 are the amino acid sequences of the heavy and light chains of the 5 anti-RhD monoclonal antibody referred to herein as RhD3 and described below in further detail.
The antibodies according to the first aspect of the present invention therefore comprise heavy chain and light chain variable regions having first 10 second and third complementarity determining regions (i.e. CDR1, CDR2 and CDR3) which are identical or substantially identical to the first second and third complementarity determining regions (CDR1, CDF*2 and CDR3) of antibody RhD1,RhD2orRhD3.
15
20
As used herein, two CDRs are "substantially identical" if they have amino acid sequences that preferably are at least 80% identical and/or differ in no more than one amino acid. More preferably the sequences are at least 90% identical and/or differ in no more than one amino acid. Preferably, where amino acid substitutions occur such substitutions are conservative substitutions. Where the CDRs of two antibodies are at least substantially identical, it is reasonable to predict that the resulting antigen binding site of the two antibodies will have similar antigen binding properties. For example, antibodies RhD1 and RhD2 have highly similar CDRs, as can be seen from Figures 1 and 2 (described below in further detail), and both have high binding affinity for the RhD antigen.
25
Most preferabfy, the CDRs of the antibody ar§ identical to those of RftDl, RhD2 or RhD3.
As used herein the term "an isolated monoclonal antibody" refers to an 30 antibody which has been produced by monoclonal techniques and which has been isolated from antibodies of other types. In other words, the only other antibodies present will be antibodies produced by ceils of the same cell line (i.e. cells all originating from the same single parent cell) as the cell which produced the monoclonal antibody. This is of course in contrast to, for example, polyclonal

antibodies where the antibodies constitute a mixture of different antibodies originating from different plasma cells.
fn a preferred embodiment, the isolated anti-RhD monoclonal antibody 5 comprises heavy and light chain variable regions which are at least 80%, more preferably at least 90%, more preferably at least 95%, more preferably at least 98%, most preferably 100% identical to the respective variable regions of the heavy and light chains of the RhD1, RhD2 or RhD3 antibody to which its CDRs are at least substantially identical. Thus, in this embodiment the antibody 10 comprises either:
a) a heavy chain variable region which is at least 80%, 90%, 95%,
98%r or 100% jdenticaJ to the variabJe repjon of Sj=Q \D NO: 2 and has first,
second and third CDRs which are identical or substantially identical to the
15 respective first, second, and third CDRs of SEQ ID NO: 2, and a light chain variable region which is at least 80%, 90%, 95%, 98%, or 100% identical to the variable region of SEQ ID NO: 4 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4; or
20
b) a heavy chain variable region which is at least 80%, 90%, 95%,
98%, or 100% identical to the variable region of SEQ ID NO: 6 and has first,
second and third CDRs which are identical or substantially identical to the
respective first, second, and third CDRs of SEQ ID NO: 6, and a light chain
25 variable region which is at least 80%, 90%, 95%, 98%, or 100% identical to the variable region of SEQ ID NO: 8 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 8; or
30 c) a heavy chain variable region which is at least 80%, 90%, 95%,
98%, or 100% identical to the variable region of S£Q ID NO: 10 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 10, and a light chain variable region which is at least 80%, 90%, 95%, 98%r or 100% identical to the

variable region of SEQ ID NO: 12 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 12.
5 Techniques for identifying antibody variable regions and CDRs,
comparing and aligning amino acid sequences, and determining the % identity between two amino acid sequences are well known in the art. For example, the CDRs, variable regions, and constant regions of an antibody can be determined using software such as IMGT/V-QUESTtooi
10 (http://imgt.cines.fr/IMGT vquest/share/textes/) using default settings, and/or via comparison with databases of known immunoglobulin sequences such as IMGT/GENE-DB (http://imgt.cines.fr/IMGT GENE-DB/GENEIect?livret=Q) or V-BASE (http://vbase.mrc-cpe.cam.ac.uk/). Amino acid or nucleic acid sequence sequences, whether for whole antibodies or specific parts thereof, can be aligned
15 and their % identity determined using ClustalW (http://www.ebi.ac.uk/Tools/clustaiw/). ClustalW2 (http://www.ebi.ac.uk/Tools/clustalw2/) or GAP
(http://genome.cs.mtu.edu/align/alian.html) using default parameters, or using proprietary software such as Vector NTI v. 10.3.
20
In a preferred embodiment, the antibody further comprises a light chain constant domain and at least one heavy chain constant domain. The light chain constant domain may be of either the kappa or lambda type. The heavy chain constant domain is preferably an IgG class constant domain. Thus, in this
25 embodiment the antibody may for example be a Fab' or F(ab')2 fragment, as discussed above, or it may be a whole antibody. If the latter, preferably all the heavy chain constant domains are IgG domains (i.e. the antibody comprises an IgG heavy chain constant region). In a particularly preferred embodiment the constant domain or region is an IgG 1 or IgG 3 constant domain or region.
30 Preferably all constant domains (both light and heavy) are human constant domains.

According to a second aspect of the present invention, there is provided an isolated polynucleotide encoding the light and/or heavy chain of an antibody according to the first aspect.
5 As used herein, the term an "isolated polynucleotide" refers to a
polynucleotide that has been isolated from a cellular environment (i.e. it is not present in a cell or organism), and it can be in purified form (i.e. substantially free of other polynucleotides, proteins, and cellular components) of form part of composition containing other polynucleotides and/or compounds. The term
10 "encoding a light chain" refers not only to sequences encoding whole light chains, but also to sequences encoding fragments thereof (such as the variable domain only) where the antibody to be expressed is an antibody fragment as described above. Similarly, the term "encoding a heavy chain" refers not only to sequences encoding whole heavy chains, but also to sequences encoding
15 fragments thereof (such as the variable domain only or the variable domain plus one or more but not all of constant domains) where the antibody to be expressed is an antibody fragment as described above.
Exemplary nucleic acid sequences include the relevant coding sequences of 20 SEQ ID NOs: 1, 3, 5, 7, 9, and 11, which sequences are the coding sequences for, respectively, amino acid SEQ ID NOs: 2, 4, 6, 8, 10, and 12. Thus, for example, if the antibody comprises identical variable regions to the variable regions of SEQ ID NOs: 2 and 4 (the heavy and light chains of the anti-RhD antibody designated RhD 1), then an exemplary nucleic acid sequence could 25 comprise the sections of SEQ ID NOs: 1 and 3 that encode said variable regions. Alternatively, such nucleic acid sequences could be modified for optimised expression (i.e. transcription and/or translation) in the desired host cell, for example via techniques known to one of skill in the art. For example, optimization of the native nucleic acid sequence may comprise one or more of: 30 optimizing the GC distribution, and AT/GC stretches (to enhance the stability of mRNA); removing inhibitory motifs (such as premature polyA signals); removing cryptic splice sites (to prevent alternative, incorrect splicing of mRNA); optimizing mRNA secondary structure (to avoid tight hairpins possibly stalling translation); optimizing open reading frames (to avoid secondary or alternative reading

frames); and optimizing codon usage (to avoid rare codons that can slow down translation).
According to a third aspect of the present invention, there is provided an 5 expression system comprising one or more expression vectors and including:
a coding sequence encoding the light chain of an antibody according to the first aspect; and
a coding sequence encoding the heavy chain of an antibody according to the first aspect. 10
The expression vector(s) may be of any type used in the art, such as for
example plasmids and viral vectors. The expression vectors of the present
invention are preferably piasmids. In addition to the antibody chain coding
sequences, the vector(s) will include the necessary regulatory sequences for
15 proper transcription and translation of the coding sequences in the intended host
cell, such as for example a suitable promoter and polyadenylation (polyA)
sequence. The vector(s) may further comprise a Kozak sequence for increased
efficiency of expression, and/or a sequence encoding for a signal peptide for post
translational transport of the antibody chains (for example for secretion of the
20 antibodies). A further preferred feature is the presence of one or more antibiotic
resistance genes and/or other forms of selection marker, allowing for selection of
cells that have been stably transfected with the vector, and/or that display
stronger expression of the antibody coding sequences, as discussed below in
more detail.
25
The promters and poly(A) sequences used to drive expression of the light and heavy chain coding sequences may be of any type used in the art. A variety of different promoters and poly(A) seqences are known, the selection of appropriate promoters and poly(A) sequences for use in the chosen host cell 30 being well within the abilities of one of ordinary skill in the art. For example, suitable promoters for use in a mammalian host cell include the SV40 early and late, elgongation factor 1 (EF-1), and cytomegalovirus (CMV) promoters. Suitable poly(A) sequences include those from SV40 poly(A), bovine growth hormone (BGH), thymidin kinase (TK), and human growth hormone (hGH). In a

preferred embodiment, the light and heavy chain coqjng sequences are driven by the human elongation factor 1 alpha (hEF-1ct) foromoter and BGH poly(A) sequence.
5 In one embodiment, the expression systery, comprises an expression
vector that includes both the coding sequence for the light chain and the coding sequence for the heavy chain.
In an alternative embodiment, the light andj heavy chain coding 10 sequences are carried by separate vectors, the expression system comprising:
a first expression vector including a coding sequence encoding the light chain of an antibody according to the first aspect; an^
3 second expression vector .includinp a cooling sequence encoding the heavy chain of an antibody according to the first asp%ct. 15
In this embodiment, one or both of said first and second expression
vectors may include a dihydrofolate reductase (ofy,) selection marker. This
marker comprises a coding sequence for DHFR, wich is coupled to suitable
promoter and polyadenylation sequences, preferably the SV40 early (SV40E)
20 promoter and poly(A) sequences. DHFR allows de novo synthesis of the DNA
precursor thymidine. Therefore, by transfecting a host cell-line which is DHFR
deficient (i.e. which is itself incapable of producing t)HFR), one can then select
for cells which have stably integrated the vector into their genome by growing the
cells in a medium deficient in deoxyribonucfeosides and ribonudeosides.
25 Moreover, once the successfully transfected cell^ have been isolated, the
expression of the desired coding sequence(s) (i.e. the light and/or heavy chain)
can be amplified by using the DHFR inhibitor methotrxate (MTX) which causes
some cells to react by amplifying large regions of DNA surrounding the dhfr
gene.
30
In a preferred embodiment, one of said first and second expression vectors includes an antibiotic resistance gene (a nucleic acid sequence that imparts resistance to the antibiotic in question) but does not include the DHFR coding sequence, and the other of said expression vectors includes the DHFR

coding sequence but does not include a gene providing resistance to the same antibiotic as said antibiotic resistance gene. The antibiotic resistance gene may be of any type used in the art. For example, suitable antibiotic resistance genes for imparting resistance to a mammalian host cell include: aminoglycoside (e.g. 5 neomycin, hygromycin B) resistance genes, such as neomycin phosphotransferase {npt) and hygromycin B phosphotransferase {hpt, hph); aminonucleoside (eg. puromycin) resistance genes such as puromycin N-acetyltransferase (pac); glycopeptide (eg. bleomycin, phleomycin) resistance genes such as the ble gene; and peptidyl nucleoside (eg. blasticidin) resistance 10 genes such as the bis, bsr or bsd genes. As with the dhfr selection marker, the antibiotic resistance gene may as needed be coupled to any suitable promoter and polyadenylation sequences. Preferred are the SV40 early (SV40E) promoter and polyfA) sequences.
15 In a particularly preferred embodiment, the antibiotic resistance gene
comprises a neomycin phosphotransferase (NPT) coding sequence. The cells stably transfected with the vector including the NPT coding sequence can then be selected for by growing the cells in a medium containing neomycin, or a neomycin analog such as G418, the toxic effects of which are neutralized by
20 NPT.
Thus, the above described embodiment, in which one vector has the dhfr selection marker and the other has the antibiotic selection gene, allows for selection of only those cells which have stably integrated both vectors into their
25 genome by growing the cells in a medium deficient in deoxyribonucleosides and ribonucleosides and containing the relevant antibiotic (such as neomycin or a suitable analogue where the antibiotic resistance gene is the npt gene). Cells that were not transfected or were transfected with only one plasmid will not survive the selection process. Moreover, because the co-transfected plasmids
30 often integrate into one spot of the genome, Subsequent growth of the successfully transfected cells in increasing concentrations of MTX can still be used to effectively amplify expression of the antibody chains encoded by both vectors (i.e. to amplify expression of both the heavy and light chain sequences).

ft should be noted that while, in this embodiment, the vector carrying the dhfr selection marker does not include a gene providing resistance to the same antibiotic as the antibiotic resistance gene carried by the other vector, it and indeed both vectors may further comprise a different antibiotic resistance gene 5 providing resistance against a further antibiotic. Again, the additional antibiotic gene may be of any type used in the art. For example, where one but not both vectors carries an NPT coding sequence (providing resistance against neomycin and analogues thereof) both vectors may usefully additionally comprise an ampicillin resistance (AmpR) gene, for the purpose of providing ampicillin
10 resistance when incorporated into a bacterial host cell. Other antibiotic resistance genes that are commonly used to impart resistance in bacterial hosts include: plactamase genes (providing resistance to piactam antibiotics such as ampicillin and other penicillins), such as TEM-1 ^lactamase; penes providing resistance to aminoglycosides such as streptomycin, kanamycin, tobramycin,
15 and amikacin; and tetracycline (e.g. tetracycline, doxycycline, minocycline, oxtetracycline) resistance genes, such as the tetA genes.
According to a fourth aspect, the present invention provides a cell transformed with an expression system according to the third aspect or fourth 20 aspects.
The host cells for use in the present invention may be of any suitable type. However, in a preferred embodiment the host cell (cell to be transfected) is a eukaryotic cell, more preferably a vertebrate ceil, most preferably a mammalian
25 cell. A variety of suitable mammalian host cells are available, such as are for example listed in US-A1-2003/0175969 referred to above. Preferred mammalian host cells include: all variants of CHO cells, such as CHO K1 and dhfr-deficient CHO (DG44, DXB11); HEK293; BHK; COS-1 and COS-7; NSO; and PER.C6. The preferred host cells are Chinese Hamster Ovary (CHO) cells, in particular
30 dftfr-deficient CHO cells {dfhr- CHO cells). The host cells may be transfected with the expression vectors using standard techniques and transfection conditions, such as are known in the art. Exemplary transfection conditions are provided in the Examples that follow.

According to a fifth aspect, the present invention provides a method of manufacturing monoclonal antibodies, comprising cultivating recombinant cells according to the fifth aspect, and recovering the monoclonal antibody from the culture medium. Exemplary growth media and conditions are provided in the 5 Examples that follow, but any suitable growth conditions and commercial or custom growth media can be used, as are routinefy employed in the art. Likewise, any standard technique for purifying secreted antibodies from growth media can be employed, exemplary techniques being again outlined below.
10 According to a sixth aspect, the present invention provides a
pharmaceutical composition comprising: a monoclonal antibody according to the first aspect; and a pharmaceutically acceptable carrier.
The monoclonal antibodies can be formulated as desired dependent on 15 the intended route of administration. For example, the monoclonal antibodies may be formulated for injection (for example intra-muscularly) analogous to conventional polyclonal anti-D formulations. Exemplary dosages range from 150 to 300 micrograms (as measured by agglutination titer, as described below in further detail). Exemplary carriers include: phosphate-buffered saline; and 20 glycine saline buffer.
The composition may comprise monoclonal antibodies of a single type only (i.e. the only antibodies present in the composition are antibodies produced by cells of the same cell line). Alternatively, the composition may comprise a
25 combination of more than one type of monoclonal antibody. For example, the composition could comprise two or more distinct types of monoclonal antibodies that are in accordance with the first aspect of the invention, such as a combination of two or all three of monoclonal antibodies RhD1, RhD2 and/or RhD3. Alternatively or additionally, the composition could comprise, in addition
30 to monoclonal antibodies according to the first aspect of the present invention, other anti-RhD monoclonal antibodies as for example are known from the art. In a preferred embodiment, the composition comprises at least one monoclonal antibody that has an IgG 1 constant domain or region, and at least one monoclonal antibody that has an IgG 3 constant domain or region.

Where the composition comprises a combination of more than one type of monoclonal antibody, it is preferred that the composition comprises no more than 50 different types of monoclonal antibody. More preferably, the composition 5 comprises at most 25, 20, 15, 10 or 5 different types.
According to a seventh aspect, the present invention provides a method
of inhibiting or preventing immunization of a RhD-negative human patient against
RhD-positive blood, comprising administering a prophylactically effective amount
10 of a monoclonal antibody according to the first aspect or pharmaceutical
composition according to the sixth aspect.
Specific indications and/or circumstances in which the monoclonal antibodies may be administered correspond to those for which the existing anti-15 RhD polyclonal antibodies are administered.
According to an eighth aspect, the present invention provides a
monoclonal antibody according to the first aspect, or a pharmaceutical
composition according to the sixth aspect, for use in a method of inhibiting or
20 preventing immunization of a RhD-negative human patient against RhD-positive
blood.
According to a ninth aspect, the present invention provides the use of a monoclonal antibody according to the first aspect in the manufacture of a 25 medicament for inhibiting or preventing immunization of a RhD-negative human patient against RhD-positive blood.
The invention is further illustrated in the following non-limiting Examples, with reference also to the accompanying drawings in which: 30
Figure 1 is an alignment of amino acid sequences of the heavy chains of monoclonal antibodies RhD1, RhD2 and RhD3, in which the variable regions have been underlined and the complementarity determining regions highlighted in bold and shaded;

Figure 2 is an alignment of amino acid sequences of the light chains of monoclonal antibodies RhD1, RhD2 and RhD3, in which the variable regions have been underlined and the complementarity determining regions highlighted 5 in bold;
Figure 3 is a map of plasmid vector pCB3;
Figure 4 is a map of plasmid vector pCB11; 10
Figure 5 is a map of pCB3 containing an anti-RhD antibody heavy chain (RhD HC) coding sequence; and
Figure 6 is a map of pCB11 containing an $nti-RhD antibody light chain 15 (RhD LC) coding sequence;
Figure 7 is an example of a dose-response Figure 8 is an example of linear regression performed on the relevant data points taken from Fig. 7.
Sequence listings which are 48 in number are provided after the 25 Drawings.
The Sequence listings are also provided separately in accompanying CD in electronic form.


Examples
Isolation of peripheral blood mononuclear cells (PBMCs) and B cells from peripheral blood of healthy volunteers hyperimmumzed with Rhesus D 5 (RhD)-positive red blood cells
Blood from healthy RhD-negative volunteers repeatedly immunized with red blood cells isolated from healthy RhD-positive individuals of the same ABO blood group was sourced from Cliniqa. Within four weeks after the last
10 immunization the anti-RhD titer in serum was checked, the volunteers were bled, their peripheral blood mononuclear cells (PBMCs) were separated from other blood cell populations by Ficoll-Hypaque (Pharmacia) gradient centrifugation, and the cells were either used fresh or cryopreserved for later use. T cells were routinely depleted by resetting with 2% S-(2-Aminoethyl)isothiouronium bromide
15 hydrobromide (AET)-treated sheep red blood cells and the resulting enriched B cells were transformed by Epstein-Barr virus (EBV).
EBV transformation
20 Since EBV activation has been shown to be advantageous for
subsequent fusion of human B cells with the respective fusion partner, enriched B cells were transformed by EBV using spent supernatant from the B95-8 marmoset cell line as a source of the virus. The B cells resuspended in a complete IMDM medium (Gibco) with 30% fetal calf serum (PCS) were seeded in
25 96-well plates at a concentration between 5x103 and 2.5x104 cells/well. The B95-8 supernatant was added to the wells in an amount ranging from 5% to 40% of the total volume. The plates were incubated in a humidified 5% C02 incubator at 37°C for two to four weeks before screening.
30 Screening of plates for transformants secreting anti-RhD antibodies
Supernatants of transformed B cells were screened for the presence of anti-RhD antibodies by competitive enzyme-linked immunoassay (EIA). The principle of the test is as follows: a labeled monoclonal anti-RhD reference

antibody of known binding affinity and specificity (Brad-5; NIBSC) competes with an unlabeled antibody (in this case, the secreted antibodies in the supernatants) for binding to RhD-positive erythrocytes. An inhibition of the reference monoc/ona/ antibody (mAb) binding indicates the presence of RhD-specific 5 antibodies that bind to the same immunodominant epitope as the reference mAb. The degree of inhibition of the reference mAb binding correlates to the concentration and affinity of the interfering antibodies.
RhD-positive erythrocytes (R2R2 haplotype; ImmucorGamma) treated
10 with papain were fixed with glutaraldehyde and immobilized on the bottom of 96-
well flat-bottom test plates. After extensive washing and blocking of the plates,
the supernatants from transformed B cells, the standards, and negative controls
were added to the wells and the plates were incubated for 30-60 min at room
temperature (RT). The plates were washed three times. The biotinylated
15 reference mAb was added and the plates were incubated for 30 more minutes at
RT. The plates were washed again and incubated with a secondary reagent,
ExtrAvidin-Alkaline Phosphatase conjugate (Sigma) for 30 min at RT. After
another washing step, Sigma Fast PNPP (p-Nitrophenyi Phosphate) substrate
(Sigma) was added. When the color developed sufficiently, the reaction was
20 stopped with 3N NaOH and the binding of the reference mAb was detected by
reading the optical densities (at 405nm) on a plate reader (Bio-Rad). The data
was analyzed with a software package supplied with the plate reader.
Cell fusion
25
Because human B cells transformed with EBV are unstable and can rapidly cease to produce antibodies, fusion with a suitable fusion partner is usually necessary to prolong their lifetime and enable their subcloning. Therefore, any cultures of transformed B cells that produced antibodies inhibiting
30 binding of the biotinylated reference antibody to Rh[)+ erythrocytes as assessed by EIA (see above) were fused to a human heterohybridoma KelVBS either by the standard polyethylene glycol (PEG) method or by electrofusion. The electrofusion was performed with the electrofusion apparatus (Eppendorf

Multiporator) and an electrofusion buffer (Eppendorf) according to manufacturer's protocols.
Subcloning of hybridomas
5
Subclones were grown on feeder layers established from newborn foreskin fibroblast line CCD-1114Sk (ATCC). Feeders were maintained in IMDM media containing 2-20% fetal bovine serum (FBS), depending on cell growth. Feeder trays were treated with UV light on the day of subcloning. The cell lines
10 to be subcloned were counted, the appropriate dilutions to plate approximately 0.3 cells/well were prepared, and the cell suspensions were pipetted into the 96-well plates containing the feeder layer. Each cell line was seeded in at least two plates. The cultures were fed every 3-4 days. The supernatants from wells exhibiting growth of hybridomas were tested by EIA usually in 3-4 weeks.
15
Hybridoma clones selected for development of recombinant cell lines
Hybridoma clones selected for development of recombinant antibodies are listed in Table 1 (below). Each clone was assigned a simplified designation 20 for the purpose of recombinant cell line development.
Table 1, Designation ofAnti-RhD Antibodies
Hybridoma clone: Antibody isotype: Clone designation:
25 SD30.06.F5.1G2 human lgG1, lambda RhD1
SD30.02.C3.3D11 human lgG1, lambda RhD2
SD412.04.G11.2D10 human lgG3, kappa RhD3
30 RNA isolation
Total RNA from the hybridoma cells was purified using Trizol reagent (Invitrogen) according to the protocol suggested by the manufacturer with the , additional step of RNA extraction with chloroform to remove traces .of phenol.

Spectrophotometrical RNA quantification was carried out at 260 nm assuming 1 OD to be equivalent to 40 ug/ml RNA.
First strand synthesis
5
The first strand of cDNA was synthesized using the Super Script III First-Strand System for RT-PCR (Invitrogen) according to the protocol suggested by the supplier. Oligo d(T) primer from the kit was used in all cases to prime the reactions. 10
RNA hydrolysis
The removal of RNA molecules from reverse transcription reaction was 15 carried out by RNaseH digestion (Super Script III First-Strand System for RT-PCR) according to manufacturer's instructions. First-strand cDNA was cleaned using QIAquick PCR Purification Kit (Qiagen).
Tailing of first-strand cDNA
20
25
To facilitate amplification of first-strand cDNA with unknown 3' sequence, poly(A) tail was appended to the 3' end of each cDNA to create a defined priming site. For this purpose, recombinant Terminal Deoxynucleotidyl Transferase (Invitrogen) was used. The reaction was carried out according to manufacturer's recommendations. Reaction product was cleaned using QIAquick PCR Purification Kit (Qiagen).
PCR amplification of Ig heavy- (HCs) and light chains (LCs)
30 The primers (SEQ ID NOs: 13 to 19) used for PCR amplification of the
heavy and light chain coding sequences from the first-strand cDNA are listed below (EcoRI restriction sequence in each primer is underlined).

Forward primer (compatible with the poly(A) extension of the first strand of cDNA):
For all chains:
5 5-GACTGAATTCI I I I I I I I I I I I I I I I I I I IV-3'
Reverse primers (gene specific):
For gamma chains:
10 5'-ACTGGAAIICGGTGCTTTATTTCCATGCTGG-3'
5'-ACTGGAAnCGTACGTGCCAAGCATCCTCG-3'
For kappa chains:
5'-ACTGGMnCAGAGGCCAAAGGATGGGAGG-3'
15 5'-GACTGAATTCCTGGAACTGAGGAGCAGGTGG-3'

For lambda chains:

5'-GACTGAATTCCCTGGGATCCTGCAGCTC-3' 5'-ACTGGAATTCGGGGTGAGGGTTGAGAACC-3J

20
PCR was carried out using PfuUltra High-Fidelity thermostable DNA-polymerase (Stratagene). Typically the first five cycles were primed only with the forward primer; annealing temperature was 45°C. After that, the reverse, gene-specific primer was added and the PCR was extended for another 30-35 cycles 25 at annealing temperature of 50-65°C. Resulting fragments were gel purified using QIAquick Gel Extraction Kit (Qiagen), subcloned into pBluescript cloning vector and sequenced.

30

Subcloning of PCR Products into pBluescript cloning vector
The purified PCR products were ligated using the Quick Ligation Kit (NEB) into pBluescript cloning vector (Stratagene) cut with EcoRV. DH5ct bacterial cells were transformed with the resulting DNA and spread onto LB plates supplemented with 40ug/ml ampicillin and pre-treated with 50|xl of

20mg/ml Xgal and 25fjJ of 200mg/mi Isopropyl (3-D-1-thiogalactopyranoside (IPTG). Colonies were blue/white selected for the presence of an insert.
Isolation of Plasmid DNA and Sequencing
5
Selected white colonies were picked and expanded. The DNA was
isolated with QIAprep Spin Miniprep Kit (Qiagen). A control digest was
performed with EcoRI (both forward and reverse PCR primers contained an
EcoRI site). Inserts in plasmids yielding the expected digestion pattern were
10 sequenced (Biotech Core).
RhD1, RhD2 and RhD3 coding and amino acid sequences
The amino acid sequences of the heavy chain (HC) and light chain (LC) 15 of RhD1, RhD2 and RhD3, and the corresponding nucleotide sequences encoding said heavy and light chains are set out in the accompanying sequence listing, as further explained below.
The sequences were analyzed with the help of IMGT databases and 20 software (imgt.cines.fr). More specifically:
the sequences of constant regions were determined from the
IMGT/GENE-DB database of genomic Ig sequences
(http://imqt.cines.fr/IMGT GENE-DB/GENEIect?livret=OV by selecting the species, locus, gene type, group (skipped subgroup) and functionality (e.g. 25 species: Homo sapiens, locus: IGH, gene type: constant, group: IGHC, functionality: functional), and searching the database - from the resulting list, the desired isotype (e.g. IGG1) was selected in order to identify appropriate IMGT/LIGM-DB reference sequence(s) for comparison with the RhD sequence;
the variable regions were determined by subtracting the constant regions; 30 and
the CDRs were determined using IMGT/V-QUEST tool (http://imgt.cines.fr/IMGT vquest/share/textes/V by selecting the immunoglobulin species (human), uploading the nucleotide sequence of the complete antibody

chain, or just its variable region, in FASTA format, and analyzing the sequence using IMGTA/-QUEST default settings.
For further information on IMGT/V-QUEST tool and 1MGT/GENE-DB see 5 also:
Lefranc M.-P., Giudicelli V., Kaas Q., Duprat E., Jabado-Michafoud J.,
Scaviner D., Ginestoux C, Clement O., Chaume D. and Lefranc G. IMGT, the
international ImMunoGeneTics information system. Nucl. Acids Res., 2005, 33,
D593-D597;
10 Giudicelli V., Chaume D. and Lefranc M.-P. IMGTA/-QUEST, an
integrated software for immunoglobulin and T cell receptor V-J and V-D-J rearrangement analysis. Nucl. Acids Res. 2004, 32, W435-W440; and,
Giudicelli V., Chaume D. and Lefranc M.-P. IMGT/GENE-DB: a comprehensive database for human and mouse immunoglobulin and T cell 15 receptor genes. Nucl. Acids Res. 2005, 33, D256-D261.
V-BASE (a database of alt human germline variable region sequences; http://vbase.mrc-cpe.cam.ac.uk/) can afso be used to determine, or corroborate, the ends of a variable region. Under Alignments, one can find germline 20 sequences of the signal peptides, V-segments, D-segments (if applicable), and J-segments of all heavy and light chains. It will be apparent from the fMGT analysis what segments are employed in a given antibody chain. One can then reference the particular J-segment in V-BASE to determine the exact ending.
25 SEQ ID NO: 1 is the nucleotide sequence of the coding region of RhD1
HC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-448 encode the variable region, of which nucleotides 133-156 encode CDR1, nucleotides 208-231 encode CDR2, and nucleotides 346-414 encode CDR3. Nucleotides 449-1437 encode the constant region (this being a gammal, or lgG1, constant
30 region). The amino acid sequence of RhD1 HC is given as SEQ ID NO: 2.
SEQ ID NO: 3 is the nucleotide sequence of the coding region of RhD1 LC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-388 encode the variable region, of which nucleotides 133-159 encode CDR1, nucleotides

211-219 encode CDR2, and nucleotides 328-357 encode CDR3. Nucleotides 389-705 encode the constant region (this being a lambda constant region). The amino acid sequence of RhD1 LC is given as SEQ ID NO: 4.
5 SEQ ID NO: 5 is the nucleotide sequence of the coding region of RhD2
HC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-448 encode the variable region, of which nucleotides 133-156 encode CDR1, nucleotides 208-231 encode CDR2, and nucleotides 346-414 encode CDR3. Nucleotides 449-1437 encode the constant region (this being a gammal, or lgG1, constant 10 region). The amino acid sequence of RhD2 HC is given as SEQ ID NO: 6.
SEQ ID NO: 7 is the nucleotide sequence of the coding region of RhD2 LC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-388 encode the variable region, of which nucleotides 133-159 encode CDR1, nucleotides 15 211-219 encode CDR2, and nucleotides 328-357 encode CDR3. Nucleotides 389-705 encode the constant region (this being a lambda constant region). The amino acid sequence of RhD2 LC is given as SEQ ID NO: 8.
SEQ ID NO: 9 is the nucleotide sequence of the coding region of RhD3 20 HC. Nucleotides 1-57 encode the signal peptide. Nucleotides 58-448 encode the variable region, of which nucleotides 133-162 encode CDR1, nucleotides 214-234 encode CDR2, and nucleotides 349-414 encode CDR3. Nucleotides 449-1578 encode the constant region (this being a gamma3, or lgG3, constant region). The amino acid sequence of RhD3 HC is given as SEQ ID NO: 10. 25
SEQ ID NO: 11 is the nucleotide sequence of the coding region of RhD3 LC. Nucleotides 1-66 encode the signal peptide. Nucleotides 67-391 encode the variable region, of which nucleotides 145-162 encode CDR1, nucleotides 214-222 encode CDR2, and nucleotides 331-360 encode CDR3. Nucleotides 30 392-711 encode the constant region (this being a kappa constant region). The amino acid sequence of RhD3 LC is given as SEQ ID NO: 12.

Alignments of amino acid sequences of RhD1 -RhD3
The amino acid sequences of RhD1-RhD3 were aligned with the ClustalW program (www.ebi.ac.uk/Tools/clustalw). using the default parameters 5 from the website- The resulting alignments of HCs and LCs are depicted in Figures 1 and 2, respectively. The variable region of each sequence has been underlined, and the CDRs highlighted in bold (the first occurring CDR, reading the sequences left to right and top to bottom, being CDR1, the second being CDR2, and the third being CDR3). Where the same amino acid occurs in all 10 three chains as aligned, this is identified by a "*" below the relevant amino acid in the bottom sequence (that of RhD3).
Similarly, GAP (http://qenome.cs.mtu.edu/aliqn/align.htmQ using default parameters (Max Match = 11; Min Mismatch = -4; Gap-Open Penalty = 10; Gap-
15 Extension Penalty = 2) can be used to align and determine percentage identity between individual pairs of sequences or sections thereof. When so compared, the RhD1 and RhD2 light chain variable regions are 94% identical (104 matches, 6 mismatches, 0 gaps, similarity score of 540), CDR1 regions are 88% identical (8 matches, 1 mismatch, 0 gaps, similarity score of 43), CDR2 regions are 100%
20 identical (3 matches, 0 mismatches, 0 gaps, similarity score of 16), and CDR3 regions are 90% identical (9 matches, 1 mismatch, 0 gaps, similarity score of 43). The RhD1 and RhD2 heavy chain variable regions are 94% identical (123 matches, 7 rrrtSYTtetohes, 0 gaps, swmtarity score ot 650), CORA tenons are S7% identical (7 matches, 1 mismatch, 0 gaps, similarity score of 37), CDR2 regions
25 are 100% identical (8 matches, 0 mismatches, 0 gaps, similarity score of 41), and CDR3 regions are 95% identical (22 matches, 1 mismatch, 0 gaps, similarity score of 131).
Expression vectors
30
Two plasmid expression vectors, designated pCB3 and pCB11, were constructed for expressing the antibody heavy and light chains in CHO dhfr-cells.

pCB3
This plasmid is illustrated in Figure 3. The components of this plasmid are as listed in Table 2.
Table 2 - Components of expression vector pCB3

Vector component Short form Function Source
Human Elongation Factor 1 a with first intron EF1a Promoter Promoter of expression Human genomic DNA (Clontech)
Ampicillin resistance gene (plactamase) AMPr Plasmid propaSation in bacteria Commercial vector (pBluescript; Stratagene)
Origin of replication pUCori Plasmid replication in bacteria Commercial vector (pBluescript; Stratagene)
Simian virus polyadenylate signal SV40E poly(A) Transcription termination Commercial vector (pSV40; BRL/lnvitrogen)
Simian virus 40E promoter sequence SV40E Promoter Promoter of expression Commercial vector (pSV40; BRL/lnvitrogen)
Bovine growth hormone polyadenylate signal BGH PolyfA) Transcription termination Commercial vector (BRL/lnvitrogen)
Dihydrofolate reductase gene DHFR DHFR selection marker Murine cDNA (Sierra Biosource, Inc.)
10 pCB11
This plasmid is illustrated in Figure 4. The components of this plasmid are as listed in Table 3.

Table 3 - Components of expression vector DCB11

Vector component Short form Function Source
Human Elongation Factor 1 a with first intron EF1a Promoter Promoter of expression Human genomic DNA (Clontech)
Ampicillin resistance gene (^lactamase) AMPr Plasmid propagation in bacteria Commercial vector
(pBluescript;
Stratagene)
Origin of replication pUCori Plasmid replication in bacteria Commercial vector
(pBluescript;
Stratagene)
Simian virus polyadenylate signal SV40E poly(A) Transcription termination Commercial vector
(pSV4Q,
BRL/lnvitrogen)
Neomycin
phosphotransferase
(Mutant) neo Mutant Antibiotic selection marker Commercial vector (pSV-Neo; BRLyinvitrogen) modified by Sierra Biosource, Inc.
Simian virus 40E promoter sequence SV40E Promoter Promoter of expression Commercial vector
(pSV40;
BRL/lnvitrogen)
Bovine growth hormone polyadenylate signal BGH Poly(A) Transcription termination Commercial vector (BRL/lnvitrogen)
Insertion of recombinant immunoglobulin genes into expression vectors
A second PCR was used to amplify the HCs and LCs with appropriate restriction sites added so that the fragments could be inserted into expression vectors. The design of the gene-specific forward primers was based on obtained sequences. The consensus Kozak motif (GCCACC), known to increase the efficiency of eukaryotic translation, was included in each forward primer (table 5).

The primers (SEQ (D NOs: 20 to 27) for Insertion of RhD1-RhD3 HCs and LCs into expression vectors were as follows.
RhD1 HC: 5 Forward gene-specific primer (GSP):
5'-ATCGTCTAGAGCCACCATGGACTGGACCTGGAGGTTCC-3'
RhD2 HC:
Forward GSP:
10 5'-ATCGTCTAGAGCCACCATGGACTGGACCTGGAGGTTCC-3'
RhD3 HC: Forward GSP:
5'-ATCGTCTAGAGCCACCATGGACACACTTTGCTACACACTCC-3' 15
The reverse primer used for all heavy chains:
5'-TGACGAATTCCACTCATTTACCCGGAGACAGG-3'
RhD1-RhD2LCs: 20 Forward GSP:
5'-ATCGTCTAGAGCCACCATGGCCTGGGCTCTGCTATTC-3'
Reverse primer
5'-ACTGGAATTCGAACCTATGAACATTCTGTAGGGG-3' 25
RhD3 LC: Forward GSP:
5'-ATCGTCTAGAGCCACCATGGACATGAGGGTCCCCG-3'
30 Reverse primer:
5'-GACTGAATTCCTAACACTCTCCCCTGTTGAAGC-3'
The PCR cycle for insertion of RhD1-RhD3 HCs and LCs into expression vectors comprised the following steps:

94°C 2min
94°C 20s 5 55°C 20s 35x
72°C 2min (1min for RhDl, RhD2 LC) /
72°C 10min
4°C hold
10
Construction of lgG3 variant of RhD1 antibody
An lgG3 variant of RhDl was designed as a chimera between the 15 variable region of RhD1 and the constant region of RhD3. The chimerization took advantage of the identical 5' ends of the RhD1 (lgG1) and RhD3 (lgG3) constant regions. The reverse primer specific for variable domain of RhD1 was designed to overlap three codons of the constant region and to introduce silent mutations that created an Nhel restriction site. Identical modification was 20 introduced into the RhD3 constant region 5' end by the forward primer. The Nhel restriction site allowed for convenient in-frame cloning of amplified RhDl variable domain in front of the RhD3 constant region. This was performed in two steps.
First, the constant region of lgG3 HC from RhD3 antibody was amplified,
25 cut with Xbal and EcoRI enzymes, and ligated into Xbal/EcoRI-digested pCB3
vector. In the second step, this intermediary plasrnfd was re-cut with Xbal and
Nhel endonucleases, and the amplified variable region of RhD1, digested with
the same enzymes, was inserted.
30 The primers (SEQ ID NO: 28-31) used for Construction of lgG3 variant of
RhD were as follows.

Primers used for amplification of RhD3 constant region;
Forward:
5'-ATCGTCTAGAGTCAGCTAGCACCAAGQGCCCATCGGTCTTCC-3' 5 Reverse:
5'-TGACGAATTCCACTCATTTACCCGGAGACAGG-3' Primers used for amplification of RhD1 variable dom,ajn:
10 Forward:
5'-ATCGTCTAGAGCCACCATGGACTGGA£cTGGAGGTTCC-3' Reverse:
5*-GATGCTAGCTGAGGAGACGGTGATCGTGG-3'
15 The PCR cycle for constructing the lgG3 variant 0f RhD1 comprised the
following steps:
94°C 2min
20 94°C 20s 55°C 20s 35x
72°C 2min /
72DC 10min
25 4°C hold
PCR enzyme: PfuUltra High-Fidelity thermostable DNA-polymerase (Stratagene).
30
Expression vectors containing cloned antibody Cjenes
The RhD1 HC, RhD1 LC, RhD2 HC, RhD£ LC, RhD3 HC, RhD3 LC, RhD1V3C HC (chimera composed of the RhD1 hea^y chain variable domain and

RhD3 heavy chain constant region) coding sequences as inserted into the expression vectors, including also the added Kozak motifs and restriction sites, are given as SEQ ID NOs: 32, 33, 34, 35, 36, 37, and 38, respectively. Figure 5 is a map of pCB3 illustrating the location of the inserted anti-RhD antibody heavy 5 chain, and Figure 6 is a map of pCB11 illustrating the location of the inserted anti-RhD antibody light chain (the location of insertion being the same, regardless of the specific RhD1, RhD, RhD3 or RhD1V3C heavy or light chain being expressed).
10 Gene optimization
Coding sequences of RhD1 and RhD3 antibodies were optimized by GENEART AG using proprietary algorithms. The optimized coding sequences for RhD1 HC, RhD1 LC, RhD3 HC, and RhD3 LC are given as SEQ ID NOs: 39, 15 40, 41 and 42, respectively.
Cloning of optimized RhD1 genes into expression vectors
The optimized genes for RhD1 were subcloned into pCB expression 20 vectors. To add the restriction sites necessary for cloning, the coding regions were amplified by PCR using the primers listed below. Each amplified fragment was inserted in the respective vector and verified by sequencing.
The primers (SEQ ID NOs: 43 to 46) that were used for appending the 25 restriction sites compatible with the pCB expression vectors to the optimized RhD1 genes are as follows.
Optimsed RhDIHC:
Forward:
30 5'-ATCGTCTAGAGCCACCATGGACTGGACCTG-3'
Reverse:
5'-ATCGGGATCCTCATCACTTGCCGGGGGAC-3'

Table 5 - Components of freezing media
Freezing medium 1:

Components Vendors Catalog # Volume per 100 ml
Gamma-irradiated dFBS HyClone SH30079.33 95 mL
dimethyl sulfoxide (DMSO) Sigma D2438 5mL
Freezing medium 2:

Component Vendors Catalog # Volume per 100 ml
Gamma-irradiated
FBS HyClone SH30070.03 90 mL
DMSO Sigma D2438 10 mL
Maintenance of cells
10
Dihydrofolate reductase (DHFR)-deficient CHO DXB11 cells were grown in Host Cell Growth Medium 1 or 2 (Table 4) and were split every 3-4 days.
Cell density and viability measurements
15
Viable cell density and cell viability was determined using the Trypan Blue exclusion method and a hemocytometer (Hausser Scientific).
Stable transfection and amplification in methotrexate (MTX)
20
CHO DXB11 cells were co-transfected with equal amounts of plasmid DNA coding for the light and heavy chains of the human IgG (Table 6). Transfections were performed using Lipofectamine 2000 reagent (Invitrogen)

following the manufacturer's recommendation. Stable transfectants were selected using Transfectant Selection Medium (Table 4).
Table 6 - Conditions for a typical transfection ofCHO DXB11 cells

Vessel Amount of HC Amount of LC Amount of
DNA DNA Lipofectamine 2000
T75 flask or 15^ig 15^g 30-75|J
10cm dish
Transfected cells were cultured for 2 days at 37°C and 5% C02 in Host Cell Growth Medium 1 or 2 prior to initiation of the selection process by replacing 10 the Growth Medium with Transfectant Selection Medium (Table 4).
During the selection process, the spent medium was removed and
replaced with fresh medium whenever necessary. After the selection process
was completed and the transfected cefls resumed growing, the cells were either
15 - transferred into the Transfectant Selection Medium (Table 4) containing
various levels of MTX (Calbiochem) for amplification of antibody genes, or
- subcloned (see below). In this case, 12 best-producing clones were selected and pooled for further amplification in MTX,
20 Single cell cloning
In order to select single-cell clones, stably transfected cells were plated in
an appropriate number of flat-bottom 96-well plates at 0.5-1 cell per well. During
the process, the cell growth and health was monitored under the microscope.
25 Cells were cultured for approximately two weeks prior to selection of the best
producing clones by screening with ELISA.

Enzyme-linked Immunosorbent Assay (ELISA)
The antibody titers during all stages of cell line development were evaluated with the Human IgG ELISA Quantitation Kit (Bethyf Laboratories) 5 according to manufacturer's instructions. Shortly, the Nunc Maxisorp ELISA plates were coated with Fc-specific goat anti-human IgG polyclonal antibody in phosphate-buffered saline (PBS). Plates were incubated overnight at 4°C. Next day, the plates were washed three times and blocked for 1 hour with powdered non-fat milk dissolved in the wash buffer. After a washing step, samples and
10 standards were pipetted onto the plates and incubated at room temperature for 1 hour, followed by three washes. Secondary antibody conjugated to horseradish peroxidase (HRP) was then added to each well and the plates were incubated again at room temperature for 1 hour. Plates were washed three times with wash buffer, rinsed once with distilled water, and tapped dry.
15 Tetramethylbenzidine (TMB)-containing substrate was added to each well and color was allowed to develop for 15 minutes at room temperature. The reaction was stopped by sulfuric acid and the plates were read on a plate reader (Bio-Rad, Molecular Dynamics, or Dynex Technologies) at 450nm. The data was analyzed with a software package supplied with the plate reader.
20
Expression of recombinant antibodies from cell pools stably transfected with non-optimized cDNAs
The scheme of transfections (performed according to Table 6) and 25 designations of the transfected cells are provided in Table 7.
Table 7 - Designated name for transfected pools.

HC DNA in pCB3 LCDNAinpCBH Name of Stable Pool Recombinant IgG Isotype
RhD1 gamma RhD1 lambda RhD1 lgG1
RhD1V3C gamma RhD1 lambda RhD4 igG3
RhD2 gamma RhD2 lambda RhD2 IgGi
RhD3 gamma RhD3 kappa RhD3 tgG3

Generally, a better expression was reached when the transfected cells were subcloned after the selection process, the clones were ranked for antibody production by ELiSA, and only the pools of 12 best-producing clones were amplified in MTX. Amplification of selected but non-subcloned transfectants 5 yielded pools exhibiting lower productivity, albeit in shorter time. One typical scheme of MTX amplification is shown below:
- Selected cells (OnM MTX) were transferred in parallel to Transfectant
Selection Medium containing 50nM or 100nM MTX (Step 1)
- Cells recovered from Step 1 were expanded and split into 200nM and
10 500nM MTX (Step 2)
- Cells that have survived Step 2 were expanded and subjected to
amplification in 1000nM MTX (Step 3)
At each step, the antibody productivity was assessed by ELISA (Table 8).
Table 8 - Examples of productivity of unamplified and amplified pools of 12 best clones

Pools of 12 best clones MTX level (nM) Antibody expression levels after 7 day culture (jig/ml)
RhD1 0 10.8
RhD1 50 5.66
RhD1 200 6.44
RhD1 500 9.12
RhD1 1000 27.8

RhD2 0 9.25
RhD2 50 12.25
RhD2 100 12.75
RhD2 200 18.4
.
RhD3 0 4.08

RhD3 200 3.14
RhD3 500 6.85

RhD4 0 1.2
RhD4 0 2
The pools yielding the best antibody titers were expanded in tissue-culture flasks in Transfectant Selection Medium (without MTX and Geneticin and 5 containing low bovine IgG FBS instead of regular FBS). The supernatants from these cultures were collected and used for purification of the antibodies.
Expression of RhD1 and RhD3 antibodies by transfected and amplified 10 clonal cell populations adapted to serum-free media
As the levels of antibody expression obtained from the cell pools (Table 8) were still not as high as desired, the transfection, selection and amplification process was carried out anew, this time employing a subcloning step (as 15 described above) after each amplification step, in addition to after the initial selection step, so as to obtain clonal cell lines (single cell clones) expressing amplified levels of anti-RhD antibody.
More specifically, CHO DXB11 cells were transfected with plasmids 20 encoding the heavy and light chains of either RhD1 or RhD3. Transfection and selection of stably transfected cells was carried out in essentially the same manner as described above. Transfected cells were then subcloned, and the resulting clones screened for antibody production. The most productive clonal cell lines were amplified. After amplification, the cells were again subcloned, and 25 the most productive clones subjected to a further round of amplification and subcloning. The selection media, and the amplification media used for the first and second amplification steps, are listed in Table 9.

The final best producing clonal cell lines (obtained after both rounds of amplification) were adapted to suspension growth in commercial serum-free media (IS CH0_CD4™, Irvine Scientific). This task was performed either in the shake fia&ks &» rrr specifier dottles ay seeding the cells In a t: 1 mixture of the final 5 amplification media (Table 9) and a serum-free media containing the same level of MTX, and then gradually increasing the proportion of the serum-free media over a period of four to six weeks until the cells were fully capable of growing in 100% serum-free medium.
10 The maximum productivities of the best producing RhD1 and RhD3 clonal
cells lines, before and after the adaptation to serum-free media, are listed in Table 9. The supernatants from these cultures were again collected and used for purification of the antibodies.

Table 9 - Selection and amplification media for five selected RhD clones. Included are productivity data before and after the adaptation to serum-free media.

5


Recombinant Clone: RhD1 Clone 1 RhD1 Clone 6 Rh03 Clonel RhD3 Clone 4
Gene Optimization: Yes Yes No No
Selection and
Amplification Media.
The
composition of the
Transfectant Selection Medium is listed in Table 4. Selection: Transfectant
Selection Medium Transfectant
Selection
Medium Transfectant Selection Medium 20nM MTX Transfectant Selection Medium 20nM MTX

Amplification Stepl: Transfectant Selection Medium NoG418 300nM MTX Transfectant Selection Medium NoG418 300nM MTX Transfectant Selection Medium NoG418 200nM MTX Transfectant
Selection Medium NoG418 200nM MTX

Amplification Step 2: Transfectant
Selection
Medium
NoG418
2,400nM
MTX Transfectant
Selection
Medium
NoG418
1(200nM
MTX Transfectant Selection Medium NoG418 800nM MTX Transfectant Selection Medium NoG418 800nM MTX
Antibody Productivity Before
adaptation to serum-free media 87 ng/ml 100 fig/ml 128 \xQlm\ 87 ng/ml

After
adaptation to serum-free
media 419ng/ml 431 ng/ml 320 ^g/rnl 326 ng/ml

Antibody purification
The pH of the culture supernatants was adjusted to pH 7.2 with 1N NaOH. Each supernatant was filtered through a 0.2u filter and loaded on a 5 protein A column pre-equilibrated in phosphate-buffered saline (PBS). The column was washed with PBS to remove all the unbound material from the culture supernatant. The antibody bound to the protein A column was eluted with 0.1M Glycine (pH 2.5). The eluate was neutralized with 2M Tris buffer adjusted to pH 8.0. The eluate containing monoclonal antibody was dialyzed against
10 PBS. The anti-RhD antibody concentration was determined by agglutination assay using D positive erythrocytes. The antibody concentration was determined spectrophotometrically at 280 nm using an optical density value of 1.4 OD for a 1 mg/mi solution based on the molar extinction coefficient for human monoclonal antibody.
15
Anti-D quantitation by hemagglutination assay
The anti-RhD antibody levels in the supernatants and purified antibody 20 were quantified by measuring the agglutination of bromeiain-treated RhD positive erythrocytes using the Technicon Autoanalyzer system as previously described by Gunson et. a! (H. H. Gunson, P. K. Phillips, and F. Stratton J. din. Path., 1972, 25, 198-205. Polyclonal Anti-RhD antibodies from NIBSC (2nd International standard 01/572) were used as a standard. 25
Briefly, bromeiain-treated RhD positive red cells are incubated with various concentrations of anti-RhD antibodies. The cells are allowed to agglutinate over a period of time. The agglutinated cells are removed in the autoanalyzer and the rest of the erythrocytes are lysed using detergent. The 30 optical density of the released hemoglobin is measured spectrophotometrically. The anti-D concentrations of the samples are calculated using a standard graph obtained from various concentrations of the Anti-D standard.

Flow cytometry assay
Each human anti-RhD monoclonal antibody was serially diluted 1 in 3
down from 0.5 mg/ml to prepare the total of 15 dilutions. Each dilution was
5 added to 1-5x105 RhD positive or RhD negative human red blood cells (RBCs),
with otherwise matching genotypes, pretreated with papain to make the antigenic
components of RhD more accessible to the antibodies. An anti-human IgG
antibody labelled with Fluorescein Isothiocyanate (FITC) was used as a
secondary antibody to stain antibodies bound to the RBCs. 10
The samples were analyzed on the FACSort instrument (Becton-
Dickinson). The RBC population was gated for based on the forward- and side
scatter parameters. Fluorescence of RhD negative samples was considered a
background, since these cells lack the RhD antigen that is targeted by anti-RhD
15 antibodies. RhD negative cells incubated with a particular concentration of antibody therefore served as a negative control for RhD positive cells incubated with the same antibody dilution. The specific fluorescence and the percentage of RhD positive cells bound by anti-RhD antibody (and stained with FITC labelled anti-human IgG) was then determined, for each dilution of anti-RhD antibody,
20 based on the difference between the level of fluorescence in the RhD positive and RhD negative samples. For each anti-RhD antibody, the percentage of positive cells bound by the antibody was plotted against the logarithm of the antibody concentration, and EC50 was estimated from this chart. This provided basic information about the binding affinity and specificity of the antibodies for
25 the RhD antigen.
ADCC assay
The effectiveness of the anti-RhD antibodies in eliminating RhD-positive 30 red blood cells in vivo, and thus utility of the antibodies in preventing immunization of an RhD-negative individual exposed to RhD-positive blood, was gauged via an antibody-dependant cellular toxicity (ADCC) assay.
The ADCC assay was based on the method described by Miescher et. al. 35 (British Journal of Haematology 2000 111:157-166). RhD positive erythrocytes

were treated with papain and subsequently labeled with the fluorescent dye 5-(and 6) carboxyfluorescein diacetate succinimidyl ester. The labeled erythrocytes were preincubated with varying concentrations (0.1-50 ng/ml) of anti-RhD antibodies for 1 hr. Peripheral blood mononuclear cells (PBMCs) were 5 added to the erythrocyte suspension and incubated for 18 hrs in a C02 incubator at 37°C. The extent of the target cell lysis at the end of incubation was determined by measuring the release of the dye from lysed RBCs into supernatant with a fluorometer. The percentage of cytotoxicity was calculated according to the following formula: 10
FCex
p — 'Cmed
% specific lysis = x 100
rC(jet — rCmed
15 where
Fcexp = fluorescence of samples
FCdet = maximum fluorescence control (obtained by lysing the RBCs with a
detergent (1% Triton-X100))
Fcjned = background fluorescence control (spontaneous release of the dye from 20 RBCs in the absence of PBMCs and antibody)
The percentage of cytotoxicity was then plotted against the logarithm of antibody concentration at which the erythrocytes were preincubated, and this data used to calculating the EC50, i.e. the effective concentration of antibody
25 causing 50% of the maximum specific lysis achievable by that antibody. By way of example, Fig. 7 is a plot of percentage cytotoxicity again antibody concentration generated from the results of an ADCC assay using an NIBSC standard (anti-RhD polyclonal antibodies). This dose-response dependence theoretically yields a sigmoid curve with a near-linear middle region. To perform
30 a linear approximation in this region, a straight line can be fitted to the pertinent data points by linear regression using a suitable software package (such as, for example, Microsoft Excel™). Fig. 8, for example, is a linear regression performed on the relevant data points from Fig. 7. An equation representing this straight line can then be used to calculate the EC50. For example for the data in

Formulations

The purified monoclonal anti-RhD antibodies can be formulated for
administration via any suitable route. Typica/fy, the antibodies are administered
5 via injection. In such circumstances, the antibody is typically formulated as a
liquid suspension of the antibodies in a suitable buffer solution. Exemplary
buffers include:
phosphate-buffered saline (20 mM phosphate buffer (pH 6.8) containing
150mMNaCI);and
10 glycine saline buffer (0.3 M glycine containing 0.15 M NaCI adjusted to
pH 6.5).
Preferred formulations comprise both monoclonal antibodies having an IgG 1 constant region and monoclonal antibodies having an IgG 3 constant 15 region. Thus, formulations comprising RhD1 antibodies (which are of the IgG 1 isotype) in combination with RhD3 antibodies (which are of the IgG 3 isotype) and/or RhD4 antibodies (which consist of the RhD1V3C heavy chain and RhD1 light chain) are preferred.
20
Dated this 26th day of December. 2008

25
30 To
The Controller of Patents The Patent Office, At Mumbai
We Claim:
1. An isolated anti-RhD monoclonal antibody comprising:
a) a heavy chain variable region having first, second and third CDRs which are identical or
substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 2, and
a light chain variable region having a first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4;
two CDRs are substantially identical if they have amino acid sequences that are at least 80% identical and/or differ in no more than one amino acid.
2. The antibody as claimed in claim 1, comprising:
a) a heavy chain variable region which is at least 80% identical to the variable region of SEQ ID
NO: 2 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 2, and
a light chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 4 and has first, second and third CDRs which are identical or substantially identical to the respective first, second, and third CDRs of SEQ ID NO: 4;.
3. The antibody as claimed in claim 2, wherein the antibody comprises: a heavy chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 2, and has first, second and third CDRs which are identical to the respective first, second and third CDRs of SEQ ID NO: 2; and a light chain variable region which is at least 80% identical to the variable region of SEQ ID NO: 4, and has first, second and third CDRs which are identical to the respective first, second and third CDRs of SEQ ID NO: 4.
4. The antibody as claimed in any one of claims 2 to 3, wherein the respective variable regions are at least 90% identical.
5. The antibody as claimed in claim 4, wherein the respective variable regions are at least 95% identical.
6. The antibody as claimed in claim 5, wherein the respective variable regions are identical.
7. The antibody as claimed in any one or more of the preceding claim, wherein the antibody comprises a light chain constant domain and a heavy chain constant domain.
8. The antibody as claimed in claim 7, wherein the antibody comprises a heavy chain constant region.

9. The antibody as claimed in claim 7 or 8, wherein said heavy chain constant domain or region is an IgG constant domain or region.
10. The antibody as claimed in claim 9, wherein said IgG constant domain or region is an IgG 1 or IgG 3 constant domain or region.
11. An isolated polynucleotide encoding the light and/or heavy chain of an antibody as claimed in any one or more of the preceding claim.
12. An expression vector including coding sequences encoding the light and heavy chains of an antibody as claimed in any one of claims 1 to 11.
13. An expression system including coding sequences encoding the light and heavy chains of an antibody as claimed in any one of claims 1 to 11, the expression system comprising:
a first expression vector including the coding sequence encoding the light chain; and
a second expression vector including the coding sequence encoding the heavy chain.
14. A cell transformed with an expression vector or system as claimed in claim 12 or 13 wherein said cell is a prokaryotic cell
15. A method of manufacturing monoclonal antibodies, comprising cultivating recombinant cells transformed with an expression vector or system as claimed in claim 12 or 13, and recovering the monoclonal antibody from the culture medium.
16. A pharmaceutical composition comprising a monoclonal antibody as claimed in any one of claims 1 to 10 and a pharmaceutically acceptable carrier

Documents:

2730-MUM-2008-ABSTRACT(24-12-2009).pdf

2730-MUM-2008-ANNEXURE TO FORM 3(21-1-2011).pdf

2730-MUM-2008-CLAIMS(24-12-2009).pdf

2730-MUM-2008-CLAIMS(AMENDED)-(17-1-2012).pdf

2730-MUM-2008-CLAIMS(AMENDED)-(23-7-2012).pdf

2730-MUM-2008-CLAIMS(MARKED COPY))-(17-1-2012).pdf

2730-MUM-2008-CORRESPONDENCE(10-5-2012).pdf

2730-MUM-2008-CORRESPONDENCE(19-9-2011).pdf

2730-MUM-2008-CORRESPONDENCE(21-1-2010).pdf

2730-MUM-2008-CORRESPONDENCE(21-1-2011).pdf

2730-MUM-2008-CORRESPONDENCE(23-11-2011).pdf

2730-MUM-2008-CORRESPONDENCE(24-12-2009).pdf

2730-MUM-2008-CORRESPONDENCE(28-1-2010).pdf

2730-mum-2008-correspondence(31-12-2008).pdf

2730-mum-2008-correspondence.pdf

2730-MUM-2008-DEED OF ASSIGNMENT(24-12-2009).pdf

2730-MUM-2008-DESCRIPTION(COMPLETE)-(24-12-2009).pdf

2730-mum-2008-description(provisional)-(31-12-2008).pdf

2730-mum-2008-description(provisional).doc

2730-mum-2008-description(provisional).pdf

2730-MUM-2008-DRAWING(24-12-2009).pdf

2730-mum-2008-drawing.pdf

2730-MUM-2008-FORM 1(19-9-2011).pdf

2730-MUM-2008-FORM 1(23-7-2012).pdf

2730-mum-2008-form 1.pdf

2730-MUM-2008-FORM 13(19-9-2011).pdf

2730-MUM-2008-FORM 13(23-7-2012).pdf

2730-MUM-2008-FORM 18(24-12-2009).pdf

2730-mum-2008-form 2(24-12-2009).pdf

2730-mum-2008-form 2(31-12-2008).pdf

2730-MUM-2008-FORM 2(TITLE PAGE)-(24-12-2009).pdf

2730-mum-2008-form 2(title page)-(31-12-2008).pdf

2730-mum-2008-form 2(title page).pdf

2730-mum-2008-form 2.doc

2730-mum-2008-form 2.pdf

2730-mum-2008-form 26.pdf

2730-MUM-2008-FORM 3(19-9-2011).pdf

2730-MUM-2008-FORM 3(21-1-2010).pdf

2730-MUM-2008-FORM 3(24-12-2009).pdf

2730-MUM-2008-FORM 3(28-1-2010).pdf

2730-mum-2008-form 3(31-12-2008).pdf

2730-mum-2008-form 3.pdf

2730-MUM-2008-FORM 5(24-12-2009).pdf

2730-MUM-2008-FORM PCT-ISA-210(21-1-2011).pdf

2730-MUM-2008-OTHER DOCUMENT(19-9-2011).pdf

2730-MUM-2008-OTHER DOCUMENT(24-12-2009).pdf

2730-MUM-2008-PCT-ISA-237(19-9-2011).pdf

2730-MUM-2008-PETITION UNDER RULE 137(17-1-2012).pdf

2730-MUM-2008-POWER OF ATTORNEY(23-11-2011).pdf

2730-MUM-2008-REPLY TO EXAMINATION REPORT(17-1-2012).pdf

2730-MUM-2008-REPLY TO HEARING(23-7-2012).pdf

2730-mum-2008-sequence listing.pdf

abstract1.jpg


Patent Number 254428
Indian Patent Application Number 2730/MUM/2008
PG Journal Number 44/2012
Publication Date 02-Nov-2012
Grant Date 02-Nov-2012
Date of Filing 31-Dec-2008
Name of Patentee BHARAT SERUMS AND VACCINES LTD.
Applicant Address 16TH AND 17TH FLOOR, HOECHST HOUSE, NARIMAN POINT, MUMBAI-400 021.
Inventors:
# Inventor's Name Inventor's Address
1 DAFTARY GAUTAM VINOD 16TH AND 17TH FLOOR, HOECHST HOUSE, NARIMAN POINT, MUMBAI-400 021.
PCT International Classification Number C07K16/34; A61K39/395; C12N15/13
PCT International Application Number N/A
PCT International Filing date
PCT Conventions:
# PCT Application Number Date of Convention Priority Country
1 NA