NZ576119A - Combination therapy involving egf inhibitors - Google Patents
Combination therapy involving egf inhibitorsInfo
- Publication number
- NZ576119A NZ576119A NZ576119A NZ57611907A NZ576119A NZ 576119 A NZ576119 A NZ 576119A NZ 576119 A NZ576119 A NZ 576119A NZ 57611907 A NZ57611907 A NZ 57611907A NZ 576119 A NZ576119 A NZ 576119A
- Authority
- NZ
- New Zealand
- Prior art keywords
- areg
- egf
- inhibitor
- sequence
- antibody
- Prior art date
Links
Landscapes
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Provided is the use of an inhibitor of HB-EGF and an inhibitor of AREG in the preparation of a medicament formulated for simultaneous or sequential use in the combination treatment of neoplastic disease; wherein, in said combination treatment, said inhibitor of HBEGF and said inhibitor of AREG act synergistically; wherein said inhibitor of HB-EGF is an antibody molecule which binds said HB-EGF or a nucleic acid molecule which inhibits expression of HB-EGF; and wherein said inhibitor of AREG is an antibody molecule which binds AREG or a nucleic acid molecule which inhibits expression of AREG. Further provided are compositions comprising the two inhibitors and specific amino acid sequences of the antibodies therein.
Description
1
2
3
4
6
7
8
9
11
12
13
14
16
17
18
19
21
22
23
24
26
27
28
29
31
32
1 Received at IPONZ on 01.06.2011
Combination Therapy Comprising an Inhibitor of HB-EGF and an Inhibitor of AREG
Field of the Invention
The present invention relates to cancer treatment. In particular it relates to methods of determining susceptibility to resistance to anti-cancer drugs,
methods for overcoming such resistance and combination therapies for the treatment of cancer.
Background to the Invention
Cancer is the leading cause of mortality in the Western countries . A large number of chertiotherapeutic agents have been developed over the last 50 years to treat cancers. The majority of chemotherapeutic agents can be divided into: alkylating agents, antimetabolites, anthracyclines, plant alkaloids, topoisomerase inhibitors, and antitumour agents. All of these drugs affect cell division, or DNA synthesis and function in some way.
The effectiveness of particular chemotherapeutic agents varies between cancers, between patients and over time in Individual patients. Cancerous cells exposed to a chemotherapeutic agent may develop resistance to such an agent, and quite often cross-resistance to several other antineoplastic agents as well. Moreover, the narrow therapeutic index of many chemo therapeut ic agents further limits their use. Accordingly, it is often necessary to change treatments of patients with cancer if the first or second line therapy is not sufficiently effective or ceases to be sufficiently effective. In many cases
2
1 combinations of particular treatments have been .
2 found to be particularly effective.
3
4 For example, colorectal cancer is one of the most
currently diagnosed cancers in Europe and one with
6 the poorest 5 year survival rates. For more than 40
7 years, inhibitors of thymidylate synthase, for
8 example 5-Fluorouracil (5-Fu), have been the
9 treatment of choice for this cancer. Thymidylate
synthase inhibitors act by causing DNA damage due to
11 misincorporation of FUTP into RNA and DNA (Longley
12 et al Nat Rev Cancer, 3:330-338, 2003; Backus et al
13 Oncol research 2000;12 (5) :231-9) . More recently new
14 chemotherapeutic agents have been introduced to the
clinic, for example the topoisomerase I inhibitors
16 (e.g. irinotecan: CPT-11) and DNA damaging agents
17 (e.g. oxaliplatin: alkylating agents).
18 ■
19 These chemotherapeutics agents, 5-Fu included, can
be used alone but it is common that clinical regimes
21 incorporate a combination. Indeed combined
22 chemotherapy has shown promising results by
23 improving the response rates in patients by acting
24 on the tumors through different pathways.
Nevertheless many patients still cannot be treated
26 through these regimes because of drug resistance
27 either acquired or inherent. In vitro and in vivo
28 studies have demonstrated that increased TS
29 expression correlates with increased resistance to
5-FU (Johnston et al, Cancer Res., 52: 4306-4312,
31 1992). Other upstream determinants of 5-FU
3
1 chemosensitivity include the 5-FU-degrading enzyme
2 dihydropyrimidine dehydrogenase and 5-FU-anabolic
3 enzymes such as orotate phosphoribosyl transferase
4 (Longley et al Nat Rev Cancer, 3:330-338, 2003).
6 The use of antimetabolites e.g. tomudex (TDX) and
7 platinum containing compounds e.g. oxaliplatin is
8 similarly limited by resistance.
9
Further, the choice of chemotherapy is further
11 complicated by cancer type and, for example, whether
12 or not the cancer is associated with a p53 mutation.
13 For example, as described in W02005/053739, whereas
14 the combination of platinum based chemotherapeutics
with antiFas antibodies was shown to have a
16 synergistic cytotoxic effect in tumours with wild
17 type p53, such synergy was not seen in p53 mutant
18 cells.
19
5-Fu, CPT-11 and oxaliplatin remain front line
21 therapies, but the development of non responsive
22 tumours or chemotherapy resistant cancer remains a
23 major obstacle to successful chemotherapy. Due to
24 the importance of early treatment of cancers, there
is a clear need for tools which enable prediction of
26 whether a particular therapy, either single or
27 combination, will be effective against particular
28 tumours in individual patients. Moreover, there
29 remains the need for new treatment regimes to
increase the repertoire of treatments available to
31 the physician.
4
1 Summary of the Invention
2
3 The present inventors have investigated proteins
4 upregulated in response to treatment with different
classes of chemotherapy and have surprisingly shown
6 that a variety of genes encoding peptide growth
7 factors of the Epidermal Growth Factor (EGF) family
8 are overexpressed in a number of different tumour
9 cell line models of cancer, from a number of
different types of cancer, following in vivo
11 challenge with different physiologically relevant
12 doses of different classes of chemotherapy.
13
14 Further investigation by the inventors has
surprisingly shown that combinations of inhibitors
16 of different EGFs results in a surprisingly dramatic
17 reduction in tumour cell growth and proliferation
18 compared to the reduction when inhibitors of a
19 single EGF were tested.
21 Accordingly, in a first aspect of the present
22 invention, there is provided a method of treating
23 • neoplastic disease in a subject, said method
24 comprising the simultaneous, sequential or separate,
administration to said subject of an effective
26 amount of (i) an inhibitor of a first EGF and
27 (ii) an inhibitor of a second EGF, wherein said
28 first and second EGF are different EGFs.
29
In a second aspect of the invention, the invention
31 provides a pharmaceutical composition comprising (i)
32 an inhibitor of a first EGF and
1 (ii) an inhibitor of a second EGF, wherein said
2 first and second EGFs are different EGFs.
3
4 . A third aspect of the invention provides kit
comprising, in combination for simultaneous,
6 separate, or sequential use in the treatment of
7 neoplastic disease,
8 (i) an inhibitor of a first EGF and
9 (ii) an inhibitor of a second EGF, wherein said
■ first and second EGF are different EGFs.
11
12 Any EGF may be used in the first, second or third
13 aspects of the invention. Thus the first and second
14 EGFs may each be independently selected from the
group consiting of HB-EGF, AREG, TGF, EREG, BTC, NRG
16 1, NRG2, NRG3, and NRG4.
17
18 In.one embodiment, the first EGF is HB-EGF and said
19 second EGF,is selected from the group consisting of
AREG, TGF, EREG, BTC, and NRG3.
21
22 In a particular embodiment, the first EGF is HB-EGF
23 and said second EGF is AREG.
24
Any inhibitors of an EGF may be used. EGF inhibitors
26 which may be used in the present invention include
27 any molecule which reduces expression of the gene
28 encoding the EGF or antagonizes the EGF protein.
29 Such molecules may include, but are not limited to,
antibodies, antibody fragments, immunoconjugates,
31 small molecule inhibitors, peptide inhibitors,
32 specific binding members, non-peptide small organic
6
1 molecules, nucleic acid molecules which inhibit EGF
2 expression, such as siRNA, antisense molecules or
3 oligonucleotide decoys.
4
' In one embodiment, the inhibitors of said first and
6 second EGFs are different. In a particular
7 embodiment, the inhibitor of the first EGF is not an
8 inhibitor of the second EGF and vice versa.
9
In one embodiment of the invention, the inhibitor of
11 each EGF is a specific inhibitor of that EGF, i.e.
12 not an inhibitor of another EGF.
13
14 In one embodiment, said inhibitor of said first EGF
is an antibody which binds said first EGF or a
16 nucleic acid molecule which inhibits EGF expression.
17
18 As described above, in one embodiment, said first
19 EGF is HB-EGF.
21 In one such embodiment the inhibitor of the first
22 EGF is an siRNA having sense and antisense sequences
23 shown as Sequence ID Nos 1 and 2 respectively:
24
.25 Sequence ID No: 1:
2 6 GAAAAUCGCUUAUAUACCUUU
27 Sequence ID No: 2:
2 8 AGGUAUAUAAGCGAUUUUCUU
29
In another such embodiment the inhibitor of the HB-
31 EGF is an siRNA having sense and antisense sequences
32 shown as Sequence ID Nos 3 and 4 respectively:
33
34 Sequence ID No: 3:
7
1 UGAAGUUGGGCAUGACUAAUU
2 Sequence ID No: 4:
3 UUAGUCAUGCCCAACUUCAUU
4
In another such embodiment the inhibitor of the HB-
6 EGF is an siRNA having sense and antisense sequences
7 shown as Sequence ID Nos 5 and 6 respectively:
. 8 '
9 Sequence ID No: 5:
GGACCCAUGUCUUCGGAAAUU
11 Sequence ID No: 6:
12 UUUCCGAAGACAUGGGUCCUU
13
14 In another such embodiment the inhibitor of the HB-
EGF is an siRNA having sense and antisense sequences
16 shown as Sequence ID Nos 7 and 8 respectively:
17
18 Sequence ID No: 7:
19 GGAGAAU G CAAAUAU GUAUU
Sequence ID No: 8:
21 UCACAUAUUUGCAUUCUCCUU
22
23 In one embodiment, the inhibitor of HB -EGF
24 comprises a pool of two, three or four of the siRNA
molecules (wherein each molecule comprises the sense
26 and complementary antisense molecule) shown above
27 i.e. two, three or four of the sense/antisense pairs
28 selected from the group consisting of Sequence ID
29 No: 1/Sequence ID No: 2, Sequence ID No: 3/Sequence
ID No: 4, Sequence ID No: 5/Sequence ID No: 6, and
31 Sequence ID No: 7/Sequence ID No: 8.
32
33
34 In one embodiment, said inhibitor of said second EGF
is an antibody which binds said second EGF or a
36 nucleic acid molecule which inhibits EGF expression.
37
8.
1 In one embodiment, said second EGF is AREG. In such
2 an embodiment, an antibody which may be used as the
3 inhibitor of AREG is the anti-AREG antibody 6E11 1E9
4 1C6. The VH and VL sequences of the 6E11 1E9 1C6
antibody have been determined by the inventors and
6 are described infra.
1
8 In one embodiment of the invention, siRNA molecules
9 which may be used in the invention as an inhibitor
of AREG is an siRNA having sense and antisense
11 sequences shown as Sequence ID Nos 9 and 10
12 respectively:
13 Sequence ID No: 9
14 UGAUAACGAACCACAAAUAUU
Sequence ID No: 10
16 UAUUUGUGGUUCGUUAUCAUU
18 In another such embodiment the inhibitor of AREG is
19 an siRNA' having sense and antisense sequences shown
as Sequence ID Nos 11 and 12 respectively:
21
22. Sequence ID No: 11
2 3 UGAGUGAAAUGCCUUCUAGUU
24 Sequence ID No: 12
. CUAGAAGGCAUUUCACUCAUU
26
27 In another such embodiment the inhibitor of AREG is
28 an siRNA having sense and antisense sequences shown
29 as Sequence ID Nos 13 and 14 respectively:
31 Sequence ID No: 13
32 GUUAUUACAGUCCAGCUUAUU
33 Sequence ID No: 14
34 UAAGCUGGACUGUAAUAACUU
36 In another such embodiment the inhibitor of AREG is
37 an siRNA having sense and" antisense sequences shown
38 as Sequence ID Nos 15 and 16 respectively:
9
1
2 Sequence ID No: 15
3 GAAAGAAACUUCGACAAGAUU
4 Sequence ID No: 16
■ UCUUGUCGAAGUUUCUUUCUU
6
7 In one embodiment, the inhibitor of AREG comprises a
8 pool of two, three or four of the siRNA molecules
9 (wherein each molecule comprises the sense and
complementary antisense molecule) shown above i.e.
11 two, three or four of the sense/antisense pairs
12 selected from the group consisting of Sequence ID
13 No: 9/Sequence ID No: 10, Sequence ID No:
14 11/Sequence- ID No: 12, Sequence ID No: 13/Sequence
ID No: 14, and Sequence ID No: 15/Sequence ID No:
16 16.
17
18 As described in the Examples, the inventors have
19 developed the antibodies with specificity for AREG,
which may be used in the invention. The antibodies
21 have found to be particularly efficacious.
22
23 Accordingly, in a fourth aspect of the invention,
24 there is provided an antibody molecule having
binding specificity for AREG, wherein the antibody
26 molecule is the 6E11 1E9 1C6 antibody, or a fragment
27 thereof.
28
29 The VH and VL domain sequences of the 6E11 1E9 1C6
antibody has been determined by the inventor and are
31 as follows.
32
33 6E11 1E9 1C6 VH sequence (Sequence ID No: 27) :
1 MECNWILPFILSVTSGVYSQVQLQQSGAELARPGASVKLSCKASGYTFTRYW
2 MQWIKQRPGQGLEWIGAIYPGNGDIRYTQKFKGKATLTADKSSSTAYMQLSS
3 LASE D SAVYYCARGTTP S SYWGQGTLVTVSAAKTTAP SVYP LAPVCGDTTGS
4 SVTLGCLVKGYF
6 6E11 1E9 1C6 VL sequence (Sequence ID No: 28)
7
8 MMSPAQFLFLLVLWIRETSGDWMTQTPLTLSVSIGQPASISCKSSQSLLDS
9 DGKTYLNWLLQRPGQSPKRLIYLVSKLDSGVPDRFTGSGSGTDFTLKISRVE
AEDLGVYYCWQGTHFPWTFGGGTKLEIKRADAAPTVSIFPPSSEQLTSGGAS
11 WCFLNNFYPK'
12
13 Thus, in one embodiment of the invention the
14 antibody molecule having binding specificity for
AREG of arid for use in the invention is an antibody
16 molecule comprising at least one of the CDRs of the
17 6E11 1E9 1C6 VH region and/or at least one of the
18 CDRs of the 6E11 1E9 1C6 VL region. In one
19 embodiment, the antibody molecule comprises all
three of the CDRS of the 6E11 1E9 1C6 VH region
21 and/or all three of the CDRS of the 6E11 1E9 1C6 VL
22 region.
23
24 In one embodiment, the specific binding member
comprises an antibody variable domain (which may be
26 VH or VL) having the VH domain sequence shown above,
27 or an antibody variable domain (which may be VL or
28 VH) having the antibody VL domain sequence shown
29 above, or both.
11
1 The antibody molecule may be a whole antibody. In
2 one alternative embodiment, the antibody molecule
3 may be an antibody fragment such as an scFv.
4
The provision of the antibody molecules of the
6 present invention enables the development of related
7 antibodies which also inhibit tumour cell growth and
8 which optionally have similar or greater binding
9 specificity.
11 Accordingly, further encompassed within the scope of
12 this aspect of the present invention are antibody
13 molecules comprising an antibody variable domain (VH
14 or VL) having the 6E11 1E9 1C6 VH sequence shown
above in which 5 or less, for example 4, 3, 2, or 1
16 amino acid substitutions have been made in at least
17 one CDR and wherein the specific binding member
18 retains the ability to inhibit the tumour cell
19 growth. Also encompassed by the invention are
antibody molecules comprising an antibody variable
21 domain (VL or VH) having the 6E11 1E9 1C6 VL
22 sequence shown above in which 5 or less, for example
23 4, 3, 2, or 1 amino acid substitutions have been
24 made in at least one CDR and wherein the specific
binding member retains the ability to inhibit the 2 6 tumour cell growth.
27
28 The method of any one of the first to third aspects
29 of the invention may further comprise the
simultaneous, sequential or separate, administration
31 to said subject of an effective amount of (iii) a
12
1 chemotherapeutic agent.
2
3 In one embodiment, the chemotherapeutic agent is
4 selected from the group consisting of
antimetabolites, topoisomerase inhibitors,
6 alkylating agents, anthracyclines, and plant
7 alkaloids.
8
9 The inventors have further shown that particular
combinations of EGF inhibitors with topoisomerase
11 inhibitors attenuate tumour cell growth to an extent
12 greater than could be predicted from the effects of
13 each inhibitor alone.
14
Accordingly, in a sixth aspect of the invention,
16 there is provided a method of treating neoplastic
17 disease in a subject, said method comprising the
18 simultaneous, sequential or separate, administration
19 to said subject of an effective amount of (i) an
inhibitor of an EGF, wherein said inhibitor is a
21 nucleic acid molecule which inhibits EGF expression
22 or an anti EGF antibody, and wherein said EGF is HB-
23 EGF or AREG, and (ii) a topoisomerase inhibitor.
24
In a seventh aspect of the invention, there is
26 provided a pharmaceutical composition for the
27 treatment of cancer, said composition comprising an
28 effective amount of (i) an inhibitor of an EGF ,
29 wherein said inhibitor is a nucleic acid molecule
which inhibits EGF expression or an anti EGF
31 antibody, and wherein said EGF is HB-EGF or AREG,
32 and (ii) a topoisomerase inhibitor.
2
3
4
6
7
8
9
11
12
13
14
16
17
18
19
21
22
23
24
26
27
28
29
31
32
Received at IPONZ on 29.02.2012
13
Ail eighth aspect of the invention provides comprising, in combination for simultaneous,
separate, or sequential use in the treatment of neoplastic disec itive amount of (i) an inhibitor of an EGF, wherein said inhibitor is a nucleic acid molecule w hibits EGF expression or an anti EGF antibody, and wherein said EGF is HB-EGF or AREG, and (ii) a topoisomerase inhibitor.
In one embodiment of any one of the sixth, seventh or eighth aspects of the invention, the EGF is HB-EGF . In another embodiment, the EGF is AREG.
In another aspect of the invention there is provided use of an inhibitor of HB-EGF and an inhibitor of AREG in the preparation of a medicament foririu 1 for simultaneous or sequ.ent.ial use in. the combination treatment of neoplastic disease;
wherein, in said combin i treatment, said
Lbitor of HB-EGF and said, inhibitor of AREG act synerg ally;
wherein said inhibitor of HB-EGF is an antibody molecule which binds said HB-EGF or a nu : acid molecule which inhibits expression of HB-EGF; and wherein said inhibitor of AREG is an antibody molecule which binds AREG or a nucleic acid molecule which inhibits express! i.
In another aspect of the invention there is provided use of an inhibitor of HB-EGF and an inh
AREG in the preparation of a rrw ment for the combination treatment of neoplastic disease, wherein
1
2
3
4
6
7
8
9
11
12
13
14
16
17
18
19
21
22
23
24
26
27
28
29
31
32
Received at IPONZ on 29.02.2012
13a said medicament is formulated for administration according to a dosage regime comprising administering said inhibitor of HB-EGF to a patient, where said inhibitor of AREG was administered within the previous 72 hours, or said inhibitor of AREG is administered simultaneously or within the subsequent 72 hours;
wherein, with said dosage regime, said inhibitor of
HB-EGF and said inhibitor of AREG act syne wherein said inhibitor of HB-EGF is an antibody molecule which binds said HB-EGF or a nu acid molecule which inhibits expression of HB-EGF; and wherein j inhibitor of AREG is an antibody molecule which binds AREG or a nucleic acid molecule w libits expression of AREG.
In another aspect of the invention there is provided combination treatment products comprising an inhibitor of HB-EGF and an inhibitor of AREG as a combined preparation for simultaneous or sequential use in the combination treatment of neoplastic disease; wherein, in said combination treatment,
said inhibitor of HB-EGF and said inhibitor of AREG act synergistically;
wherein said inhibitor of HB-EGF is an antibody molecule which binds said HB-EGF or a nu acid molecule which inhibits expression of HB-EGF; and wherein said inhibitor of AREG is an antibody molecule which binds AREG or a nucleic acid molecule which inhibits expression of AREG.
1
2
3
4
6
7
8
9
11
12
13
14
16
17
18
19
Received at IPONZ on 02.02.2012
13b
In these aspects of the invention, any t- merase inhibitor may be used. In a particular embodiment,
the topoisomerase inhibitor is CPT-11. In another embodiment, the topoisomerase inhibitor is an active metabolite of CPT-ll, for example SN-38.
In one embodiment, wherein the EGF is AREG, the EGF inhibitor is the anti-AREG antibody 6E11 1E9 1C6.
The methods of the invention may be used to treat any neoplastic disease. In a particular embodiment, the neoplastic disease is cancer. For example, neoplastic diseases which may be tr< using the compositions and methods of the invention include, but are not limited to, colorectal cancer, breast cancer, lung cancer, prostate cancer, hepatocellular cancer, lymphoma, leukaemia, gastric cancer, pancreatic cancer, cervical cancer, ovarian cancer, liver cancer, renal cancer, thyroid cancer,
14
1 melanoma, carcinoma, head and neck cancer, and skin
2 cancer.
3
4 In one particular embodiment, the neoplastic disease
is colorectal cancer.
6
7 In another embodiment, the neoplastic disease is
8 breast cancer.
9
In another another embodiment, the neoplastic
11 disease is lung cancer.
12
13 As described in the Examples, the inventors have
14 shown that certain EGFs are upregulated by
chemotherapies in p53 mutant tumour cells as well as
16 in p53 wild type tumours. This is particularly
17 surprising given that resistance to chemotherapy has
18 previously been shown to be largely dependent on p53
19 status.
21 In a particular embodiment, the neoplastic disease
22 is a cancer comprising a p53 mutation.
23
24 Further provided by the invention in a ninth aspect
is a method of inducing and/or enhancing expression
26 of a gene encoding an EGF protein in a cell or
27 tissue; said method comprising administration of a
28 topoisomerase inhibitor to said cell or tissue,
29 wherein said EGF is selected from the group
consisting of AREG, TGF, EREG, BTC, and NRG3.
1 The demonstration by the present inventors that
2 expression of EGFs are upregulated in response to
3 treatment with diverse topoisomerase inhibitors
4 suggests that the therapeutic effect of treatment
with these chemotherapies may, in certain patients,
6 be compromised by the upregulation of EGFs.
7
8 Thus, the invention may be used in assays to
9 determine whether or not treatment with a
topoisomerase inhibitor e.g. CPT-11 or an analogue
11 thereof may be effective in a particular patient.
12
13 Thus, in a tenth aspect of the present invention,
14 there is provided an in vitro method for evaluating
the response of tumour cells from a subject to the
16 presence of a topoisomerase inhibitor to predict
17 response of the tumour cells in vivo to treatment
18 with the topoisomerase inhibitor, which method
19 comprises:
(a) providing a sample of tumour cells from a
21 subject;
22 (b) exposing a portion of said sample of tumour .
23 cells to said topoisomerase inhibitor;
24 (c) comparing expression of one or more genes
encoding one or more EGFs wherein said EGF is
26 selected from the group consisting of AREG, TGF,
27 EREG, BTC, and NRG3 in said portion of the sample of
28 tumour cells exposed to said topoisomerase inhibitor
29 with expression of said gene(s) in a control portion
of said sample which has not been exposed to said
31 topoisomerase inhibitor; wherein enhanced expression
32 in the portion of sample exposed to said
16
1 topoisomerase inhibitor is indicative of decreased
2 sensitivity to said topoisomerase inhibitor.
3
4 The invention further represents a tool for
prognosis and diagnosis of a subject afflicted with
6 a tumour. For the purpose of prognosis, determining
7 the expression level of a gene before and after
8 chemotherapeutic treatment would identify if the
9 subject will respond to a combinatory treatment
approach. For the purpose of diagnosis the
11 expression profile of a tumours genetic response to
12 chemotherapy would identify which combination
13 therapy would be most effective for that tumour.
3.4
Thus, an eleventh aspect of the invention provides a
16 method of prognosis for evaluating the response of a
17 patient to combination therapy comprising a
18 topoisomerase inhibitor and an inhibitor of an EGF,
19 said method comprising (a) determining expression of
a gene encoding an EGF in an in vitro sample
21 containing tumour cells obtained from a subject
22 prior to treatment with said chemotherapeutic
23 treatment
24 (b) determining expression of said gene encoding
said EGF, wherein said EGF is selected, from the
26 group consisting of AREG, TGF, EREG, BTC, and NRG3,
27 in an in vitro sample containing tumour cells
28 obtained from a subject after treatment with said
29 chemotherapeutic treatment;
(c) comparing expression in (b) with expression in
31 (a), wherein enhanced expression in (b) compared to
32 (a) is indicative that the patient may benefit from
17
1 combination therapy comprising a topoisomerase
2 inhibitor and an inhibitor of said EGF.
3
4 In the tenth or eleventh aspects of the invention
the expression of gene(s) encoding one or more EGFs
6 may be determined. For example, the expression of
7 genes encoding at least two, for example three, four
8 or five of AREG, TGF, EREG, BTC, and NRG3 may be
9 determined.
-11 In another embodiment of the tenth or eleventh
12 aspects of the invention the expression of genes
13 encoding at least two, for example three, or four of
14 TGF, EREG, BTC, and NRG3 may be determined.
16 In aspects of the invention involving the
17 determination of expression of a gene encoding an
18 EGF, the expression of any gene encoding said EGF in
19 the subject may be determined.
21 For example, in an embodiment in which the EGF is
22 AREG, the gene may be NM_001657.' In an embodiment,
23 in which the EGF is HB-EGF, the gene may be
24 NM_0 0194 5.
26 In an embodiment of the invention, expression of
27 said gene in the sample exposed to said
28 chemotherapeutic agent is considered to be enhanced
29 if the expression is at least 1.5-fold, preferably
•at least 2-fold, more preferably at least 5-fold,
31 that of the one or more genes in the control portion
18
1 of said sample which has not been exposed to said
2 chemotherapeutic agent.
3
4 In the present application, unless the context
demands otherwise, where reference is made to a'
6 chemotherapeutic agent and an EGF modulator, the
7 chemotherapeutic agent and the EGF modulator are
8 different agents. Generally, the chemotherapeutic
9 agent will have a' different mode of action from the
EGF'modulator. In one embodiment, the
11 chemotherapeutic agent will not inhibit the EGF.
12
13 In a further aspect of the invention, there is
14 provided the use of an inhibitor of a first EGF in
the preparation of a medicament for simultaneous,
16 separate or sequential use with an inhibitor of a
17 second EGF for the treatment of neoplastic disease;
18 wherein said first and second EGFs are different
19 EGFs.
21 Another aspect of the invention provides the use of
22 an inhibitor of a second EGF in the preparation of a
23 medicament for simultaneous, separate or sequential
24 use with an inhibitor of a first EGF for the
treatment of neoplastic disease; wherein said first
26 and second EGFs are different EGFs.
27
28 Another aspect which is provided is the use of an
29 inhibitor of an EGF,
wherein said'inhibitor is a nucleic acid molecule
31 which inhibits EGF expression or an anti EGF
32 antibody, and wherein said EGF is HB-EGF or AREG,
19
1 in the preparation of a medicament for the
2 simultaneous, separate or sequential use with a
3 topoisomerase inhibitor in the treatment of a
4 neoplastic disease.
6 Further provided is the use of a topoisomerase
7 inhibitor in the preparation of a medicament for
8 simultaneous, separate or sequential use with an
9 inhibitor of an EGF in the treatment of a neoplastic
disease,
11 wherein said inhibitor of an EGF is a nucleic acid
12 molecule which inhibits EGF expression or^an anti
13 EGF antibody, and wherein said EGF is HB-EGF or
14 AREG.
16 Preferred and alternative features of each aspect of
17 the invention are as for each of the other aspects
18 mutatis mutandis unless the context demands
19 otherwise.
2 0
21 Detailed Description
22
23 As described above and in the Examples, the present
24 invention is based on the demonstration that
expression of various EGF genes and proteins are
26 upregulated in tumour cells in the presence of
27 certain chemotherapies and that particular
28 combinations of EGF inhibitors and chemotherapeutic
29 agents as well as particular combinations of two or
more EGF inhibitors demonstrate superadditive
31 effects in the attenuation of tumour cell growth.
1 Assays
2
3 As described above, in one embodiment, the present
4 invention relates to methods of screening samples
comprising tumour cells for expression of EGF genes
6 in order to determine suitability for treatment
7 using particular chemotherapeutic agents.
8
9 The methods of the invention may involve the
determination of expression of any gene encoding an
11 EGF. The EGF-family of peptide growth factors' is
12 made up of 10 members which have the ability to
13 selectively bind the ErrB receptors (ErrBl or EGF
14 receptor, ErrB2 or Her2, ErrB3 and ErrB4).
16 In one embodiment of the invention, the EGF is a
17 ligand of ErbB-1, for example, amphiregulin (AREG),
18 TGF, Epiregulin (EREG) or BTC.
19
In another embodiment, the EGF is a ligand of ErbB-
21 4, for example NRG3
22
23 Accession details are provided for each of these 2'4 genes below.
Gene
Accession No
BTC
NM_0 0172 9
HB-EGF
: NM_0 0194 5
AREG
NM_0 01657
TGFA
NM_0 032 3 6
EREG
NM_0 014 32
NRG 3
NM_0 01010848
21
1
2 The expression of any gene encoding an EGF of
3 interest may be determined.
4
For example, where the EGF is AREG, the Areg gene
6 may be NM_001657.
7
8 In a particular embodiment of the invention, the
9 gene is Areg having accession no: NM_00.1657. In
another particular embodiment of the invention, the
11 gene is the HB-EGF gene having accession no:
12 NM_0 0194 5.
13
14 The expression of each gene may be measured using
any technique known in the art. Either mRNA or
16 protein can be measured as a means of determining
17 up-or down regulation of expression of a gene.
18 Quantitative techniques are preferred. However semi-
19 quantitative or qualitative techniques can also be
used. Suitable techniques for measuring gene
21 products include, but are not limited to, SAGE
22 analysis, DNA microarray analysis, Northern blot,
23 Western blot, immunocytochemical analysis, and
24 ELISA. "
26 In the methods of the invention, RNA can be detected
27 using any of the known techniques in the art.
28 Preferably an amplification step is used as the 2 9 amount of RNA from the sample may be very small.
Suitable techniques may include RT-PCR,
31 hybridisation of copy mRNA (cRNA) to an array of
32 nucleic acid probes and Northern Blotting.
22
1
2 For example, when using mRNA detection, the method
3 may be carried out by converting the isolated mRNA
4 to cDNA according to standard methods; treating the
converted cDNA with amplification reaction reagents
6 (such as cDNA PCR reaction reagents) in a container
7 along with an appropriate mixture of nucleic acid
8 primers; reacting the contents of the container to
9 produce amplification products; and analyzing the
amplification products to detect the presence of
11 gene expression products of one or more genes
12 encoding Areg in the sample. Analysis may be
13 accomplished using Northern Blot analysis to detect
14 the presence of the gene products in the
amplification product. Northern Blot analysis is
16 known in the art. The analysis step may be further
17 accomplished by quantitatively detecting the
18 presence of such gene products in the amplification
19 products, and comparing;the quantity of product
detected against a panel of expected values for
21 known presence or absence in normal and malignant
22 tissue derived using similar primers.
23
24 Primers for use in methods of the invention will of
course depend on the gene(s), expression of which is
26 being determined. In one embodiment of the
27 invention, one or more of the following primer sets
28 may be used:
29
Forward : TTTTTTGGATCCAATGACACCTACTCTGGGAAGCGT (SEQ
31 ID No:17)
23
1 Reverse : TTTTTTAAGCTTAATTTTTTCCATTTTTGCCTCCC(SEQ ID
2 No:18)
3 And Exon Spanning
4 Forward : TTTTTTGGATCCCTCGGCTCAGGCCATTATGCTGCT(SEQ
ID No:19)
6 Reverse : TTTTTTAAGCTTTACCTGTTCAACTCTGACTG(SEQ ID
7 No:20)
8
9 Forward 5'-TTTCTGGCTGCAGTTCTCTCGGCACT-3'(SEQ ID
No:21)
11 Reverse 5'-CCTCTCCTATGGTACCTAAACATGAGAAGCCCC-3'(SEQ
12 ID No:22)
13
14 In e.g. determining gene expression in carrying out
methods of the invention, conventional molecular
16 biological, microbiological and recombinant DNA
17 techniques known in the art may be employed.
18 Details of such techniques are described in, for
19 example, Current Protocols in Molecular Biology/ 5th
ed.,Ausubel et al. eds., John Wiley & Sons, 2005
21 and, Molecular Cloning: a Laboratory Manual: 3rd
22 edition Sambrook et al., Cold Spring Harbor
23 Laboratory Press, 2001.
24
The assays of the invention may be used to monitor
26 disease progression, for example using biopsy
27 samples at different times. In such embodiments,
28 instead of comparing the expression of EGF against a
29 control sample which has not been exposed to said
chemotherapeutic agent, the expression of the EGF is
31 compared against a sample obtained from the same
32 tissue at an earlier time point, for example from
24
1 days, weeks or months earlier.
2
3 The methods of the invention may be used to
4 determine the suitability for treatment of any
suitable cancer with a chemotherapeutic agent e.g.
6 CPT-11 or analogues thereof. For example the
7 methods of the invention may be used to determine
8 the sensitivity or resistance to treatment of
9 cancers including, but not limited to,
gastrointestinal, such as colorectal, te, head and
11 neck cancers.
12
13 In a particular embodiment of the invention, the
14 methods of the invention may be used to determine
the sensitivity or resistance to treatment of
16 colorectal cancer.
17
18 In another particular embodiment of the invention,
19 the methods of the invention may be used to
'determine the sensitivity or resistance to treatment
21 of lung cancer.
22
23 In another particular embodiment of the invention,
24 the methods of the invention may be used to
determine the sensitivity or resistance to treatment
26 of breast cancer.
27
28 The nature of the tumour or cancer will determine
29 the nature of the sample which is to be used in the
methods of the invention. The sample may be, for
31 example, a sample from a tumour tissue biopsy, bone
32 marrow biopsy or circulating tumour cells in e.g.
1 blood. Alternatively, e.g. where the tumour is a
2 gastrointestinal tumour, tumour cells may be
3 isolated from faeces samples. Other sources of
4 tumour cells may include plasma, serum,
cerebrospinal fluid, urine, interstitial fluid,
6 ascites fluid etc.
7
8 For example, solid tumour samples collected in
9 complete tissue culture medium with antibiotics.
Cells may be manually teased from the tumour
11 specimen or, where necessary, are enzymatically
12 disaggregated by incubation with collagenase/DNAse
13 and suspended in appropriate media containing, for
14 example, human or animal sera.
16 In other embodiments, biopsy samples may be isolated
17 and frozen or fixed in fixatives such as formalin.
18 The samples may then be tested for expression levels
19 of genes at a later stage.
21 In determining treatment, it may e desirable to
22 determine p53 status of a cancer. For example, p53
23 status may be useful as it may dictate the type of
24 chemotherapy which should be used in combination
with particular EGF proteins. p3 status may be 26' detemined using conventional methods. For example,
27 the use of immunohistochemistry may be used to
28 identify hotspot mutations while gene sequencing or
29 other DNA analysis methodologies may also be
employed. This analysis may suitably be performed on
31 isolated tumour tissue.
26
1 Chemotherapeutic Agents
2 Chemotherapeutic agents may be used in certain
3 embodiments of the the present invention. For
4 example agents which may be used include
antimetabolites, including thymidylate synthase
6 inhibitors, nucleoside analogs, platinum cytotoxic
7 agents, topoisomerase inhibitors or antimicrotubules
8 agents. Examples of thymidylate synthase inhibitors
9 which may be used in the invention include 5-FU, MTA
and TDX. An example of an antimetabolite which may
11 be used is tomudex (TDX). Examples of platinum
12 cytotoxic agents which may be used include cisplatin
13 and oxaliplatin.
14
Chemotherapeutic agents which may be used in the
16 present invention in addition or instead of the
17 specific agents recited above, may include
18 alkylating agents; alkyl sulfonates; aziridines;
19 ethylenimines; methylamelamines; nitrogen mustards;
nitrosureas; anti-metabolites; folic acid analogues;
21 purine analogs; pyrimidine analogs; androgens; anti-
22 adrenals; folic acid replenishers; aceglatone;
23 aldophosphamide glycoside; aminolevulinic acid;
24 amsacrine; bestrabucil; bisantrene; edatraxate;
defofamine; demecolcine; diaziquone; elfomithine;
26 elliptinium acetate; etoglucid; gallium nitrate;
27 hydroxyurea; lentinan; ionidamine; mitoguazone;
28 mitoxantrone
29
In particular embodiments of the invention, the
31 chemotherapeutic agent is a topoisomerase inhibitor.
27
1 Any suitable topoisomerase inhibitor may be used in
2 the present invention. In a particular embodiment,
3 the topoisomerase inhibitor is a topoisomerase I
■ 4 inhibitor, for example a camptothecin. A suitable
topoisomerase I inhibitor, which may be used in the
6 present invention is irenotecan (CPT-11) or its
7 active' metabolite SN-38. CPT-11 specifically acts in
8 the S phase of the cell cycle by stabilizing a
9 reversible covalent reaction intermediate, referred
to as a cleavage or cleavage complex and may also
11 induces G2-M cell cycle arrest.
12
13 In certain embodiments of the invention, the
14 chemotherapeutic agent is a fluoropyrimidine e.g. 5-
FU.
16
17 Where reference is made to specific chemotherapeutic
18 agents,, it should be understood that analogues
19 including biologically active derivatives and
substantial equivalents thereof, which retain the
21 antitumour activity of the specific agents, may be
22 used.
23
24 EGF Inhibitors
As described above, the inventors have found that
26 ^combinations of two or more inhibitors of EGFS may
27 be used to obtain a dramatically enhanced tumour
28 cell growth attenuating effect. In certain
29 embodiments of the invention, any molecule' which
reduces expression of an EGF gene or antagonizes the
31 'EGF protein may be used as the EGF inhibitor. In
28
1 particular embodiments, the EGF is HB-EGF, AREG,
2 TGF, EREG, BTC, or NRG3.
3
4 In one embodiment, inhibitors of HB-EGF and of AREG
are used.
6
7 EGF inhibitors may include, but are not limited to,-
8 antibodies, antibody fragments, immunoconjugates,
9 small molecule inhibitors, peptide inhibitors,
specific binding members, non-peptide small organic
11 molecules, antisense molecules, aptamers, or
12 oligonucleotide decoys.
13
14 Any Erbl or EGF receptor inhibitor should indirectly
inhibit AREG activity. Suitable inhibitors include,
16 but are not limited to, PD169540 (a pan-ErbB
17 inhibitor) and IRESSA (an ErbBl-specific inhibitor).
18
19 Other suitable inhibitors may include CTyrphostin AG
1478 (a selective and potent inhibitor of EGF-R
21 kinase) which indirectly inhibits TGF-alpha; ZM
22 252868 is an Epidermal growth factor (EGF) receptor-
23 specific tyrosine kinase inhibitor which inhibits
24 TGF-alpha actions in ovarian cancer cells (Simpson
et al, British Journal of Cancer, 7 9(7-8):1098-103,
26 1999).
27
28 A suitable inhibitor of HB-EGF may include CRM197 .
29
In one embodiment, an indirect inhibitor of the EGF-
31 receptor may be utilised.
32
29
1 In another embodiment, the inhibitor a direct
2 inhibitor of the EGF is used. In particular,
3 embodiments, a direct inhibitor is an antibody
4 molecule which binds EGF or a nucleic acid molecule
which inhibits expression of said EGF.
6
7 In one embodiment, the inhibitor is an anti EGF
8 antibody.
9
The inventors have developed some novel antibodies
J
11 for use in the present invention. In a particular,
12 embodiment, an antibody of or for use in the
13 invention is an antibody molecule having binding
14 specificity for AREG, wherein the antibody molecule
is the 6E11 1E9 1C6 antibody, or a fragment thereof.
16
17 Antibody molecules of or for use in the invention
18 herein include antibody fragments and "chimeric"
19 antibodies in which a portion of the heavy and/or
light chain is identical with or homologous to
21 corresponding sequences in antibodies derived from a
22 particular species or belonging to a particular
23 antibody class or subclass, while the remainder of
24 the chain (s) is identical with or homologous to
corresponding sequences,in antibodies derived from
26 another species or belonging to another antibody
27 class or subclass, as well as fragments of such
28 antibodies, so long as they exhibit the desired
29 biological activity (see U. S. Patent No. 4, 816,
567 ; and Morrison et al., Proc. Natl. Acad. Sci.
31 USA, 81 : 6851-6855 (1984)). Chimeric antibodies of
32 interest herein include "primatized"antibodies
1 comprising variable domain antigen-binding sequences
2 derived from a non-human primate(e. g. Old World
3 Monkey, Ape etc), and human constant region
4 sequences.
6 An antibody molecule for use in the invention may be
7 a bispecific antibody or bispecific fragment. For
8 example, the antibody molecule or fragment may have
9 specificity for HB-EGF and for AREG. For example, In
one embodiment, a bispecific antibody molecule for
11 use in the present invention may comprise a first .
12 heavy chain and a first light chain from the anti
13 6E11 1E9 1C6 and an additional antibody heavy chain
14 and light chain with binding specificity for HB-EGF.
A number of methods are known in the art for the
16 production of antibody bispecific antibodies and
17 fragments. For example, such methods include the
18 fusion of hybridomas or linking of Fab' fragments
19 (for example, see Songsivilai & Lachmann, Clin. Exp.
Immunol. 79: 315-321 (1990), Kostelny et al., J.
21 Immunol. 148:1547-1553 (1992)). In another
22 embodiement, bispecific antibodies may be formed as
23 "diabodies".
24
Antibody molecules, such as antibodies and antibody
26 fragments, for use in the present invention may be
27 produced .in any suitable way, either naturally or
28 synthetically. Such methods may include, for
29 example, traditional hybridoma techniques (Kohler
and Milstein (1975) Nature, 256 :495-499),
31 recombinant DNA techniques (see e.g. U. S. Patent
32 No. 4,816, 567), or phage display techniques using
31
1 antibody libraries (see e.g. Clackson et al. (1991)
2 Nature, 352: 624-628 and Marks et al. (1992) Bio/
3 Technology, 10: 779-783). Other antibody production
4 techniques are described in Using Antibodies: A
Laboratory Manual, eds. Harlow and Lane, Cold Spring
6 Harbor Laboratory, 1999.
7
8 Traditional hybridoma techniques typically involve
9 the immunisation of a mouse or other animal with an
antigen in order to elicit production of lymphocytes
11 capable of binding the antigen. The lymphocytes are
12 isolated and fused with a a myeloma cell line to
13 form hybridoma cells which are then cultured in
14 conditions which inhibit the growth of the parental
myeloma cells but allow growth of the antibody
16 producing cells. The hybridoma may be subject to
17 genetic mutation, which may or may not alter the
18 binding specificity of antibodies produced.
19 Synthetic antibodies can be made using techniques
known in the art (see, for example, Knappik et al,
21 J. Mol. Biol. (2000) 296, 57-86 and Krebs et al, J.
22 Immunol. Meth. (2001) 2154 67-84.
23
24 Modifications may be made in the VH, VL or CDRs of
the binding members, or indeed in the FRs using any
26 suitable technique known in the art. For example,
27 variable VH and/or VL domains may be produced by
28 introducing a CDR, e.g. CDR3 into a VH or VL domain
29 lacking such a CDR. Marks et al. (1992) Bio/
Technology, 10: 779-783 describe a shuffling
31 technique in which a repertoire of VH variable
32
1 domains lacking CDR3 is generated and is then
2 combined with a CDR3 of a particular antibody to
3 produce novel VH regions. Using analogous
4 techniques, novel VH and VL domains comprising CDR
derived sequences of the present invention may be
6 .produced.
7
8 Accordingly, antibodies and antibody fragments for
9 use in the invention may be produced by a method
comprising: (a) providing a starting repertoire of
11 nucleic acids encoding a variable domain, wherein
12 the variable domain includes a CDRl, CDR2 or CDR3
13 to be replaced or the nucleic acid lacks an encoding
14 region for such a CDR; (b) combining the repertoire
with a donor nucleic acid encoding an amino acid
16 sequence such that the donor nucleic acid, is
17 inserted into the CDR region in the repertoire so as
18 to provide a product repertoire of nucleic acids
19 encoding a variable domain; (c) expressing the
nucleic acids of the product repertoire; (d)
21 ' selecting a specific antigen-binding fragment
22 specific for said target; and (e) recovering the
23 specific antigen-binding fragment or nucleic acid
24 encoding it. The method may include an optional step
of testing the specific binding member for ability
26 to inhibit the activity of said target.
27
28- Alternative techniques of producing antibodies for
29 use in the invention may involve random mutagenesis
of gene(s) encoding the VH or VL domain using, for'
31 example, error prone PCR (see Gram et al, 1992,
33
1 P.N.A.S. 89 3576-3580. Additionally or
2 alternatively, CDRs may be targeted for mutagenesis '3 e.g. using the'molecular evolution approaches
4 described by Barbas et al 1991 PNAS 3809-3813 and
Scier 1996 J Mol Biol 263 551-567.
6
7 An antibody for use in the invention may be a•
8 "naked" antibody (or fragment therof) i.e. an
9 antibody (or fragment thereof) which is not
conjugated with an "active therapeutic agent". An
11 "active therapeutic agent" is a molecule or atom
12 which is conjugated to a antibody moiety (including
13 antibody fragments, CDRs etc) to produce a
14 conjugate. Examples of such "active therapeutic
agents" include drugs, toxins, radioisotopes,
16 immunomodulators, chelators, boron compounds, dyes
17 etc.
18
19 An EGF inhibitor for use in the invention may be in
the form of an immunoconjugate, comprising an
21 antibody fragment conjugated to an "active
22 therapeutic agent". The therapeutic agent may be a
23 chemotherapeutic agent or another molecule.
V
24
Methods of producing immunoconjugates are well known
26 in the art; for example, see U. S. patent No.
27 5,057,313, Shih et al., Int. J. Cancer 41: 832-839
28 (1988); Shih et al., Int. J.Cancer 46: 1101-1106
29 (1990), Wong, Chemistry Of Protein Conjugation And
Cross-Linking (CRC Press 1991); Upeslacis et al.,
34
1 "Modification of Antibodies by Chemical Methods,"in
2 Monoclonal Antibodies: Principles And Applications,
3 Birch et al. (eds.), pages 187-230 (Wiley-Liss, Inc.
4 1995); Price, "Production and Characterization of
Synthetic Peptide-Derived Antibodies," in Monoclonal '6 Antibodies: Production, Engineering And Clinical
7 Application, Ritter et al.(eds.), pages 60-84
8 (Cambridge University Press 1995) .
9
The antibody molecules for use in the invention may
11 comprise further modifications. For example the
12 antibody molecules can be glycosylated, pegylated,
13 or linked to albumin or a nonproteinaceous polymer.
14
Antisense/siRNA
16
17 Inhibitors of EGF and inhibitors of HB-EGF for use
18 in the present invention may comprise nucleic acid
19 molecules capable of modulating gene expression, for
example capable of down regulating expression of a
21 sequence encoding an EGF protein. Such nucleic acid
22 molecules may include, but are not limited to
23 antisense molecules, short interfering nucleic acid
24 (siNA), for example short interfering RNA (siRNA),
double-stranded RNA (dsRNA), micro RNA, short
26 hairpin RNA (shRNA), nucleic acid sensor molecules,
27 allozymes, enzymatic nucleic acid molecules, and
28 triplex oligonucleotides and any other nucleic acid 2 9 molecule which can be used in mediating RNA
interference "RNAi" or gene silencing in a sequence-
31 specific manner (see for example Bass, 2001, Nature,
1 411, 428-429; Elbashir et al., 2001, Nature, 411,
2 494-498; WO 00/44895; WO 01/36646; WO 99/32619; WO
3 00/01846; WO 01/29058; WO 99/07409; and WO 00/44914;
4 Allshire, 2002, Science, 297, 1818-1819; Volpe et
al., 2002, Science, 297, 1833-1837; Jenuwein, 2002,
6 Science, 297, 2215-2218; Hall et al., 2002, Science,
7 297, 2232-2237; Hutvagner and Zamore, 2002, Science,
8 297, 2056-60; McManus et al., 2002, RNA, 8, 842-850;
9 Reinhart et al., 2002, Gene & Dev., 16, 1616-1626;
and Reinhart & Bartel, 2002, Science, 297, 1831).
11
12 An "antisense nucleic acid", is a non-enzymatic
13 nucleic acid molecule that binds to target RNA by
14 means of RNA-RNA or RNA-DNA or RNA-PNA (protein
nucleic acid; Egholm et al., 1993 Nature 365, 566)
16 interactions and alters the activity of the target
17 RNA (for a review, see Stein and Cheng, 1993 Science
18 261, 1004 and Woolf et al., U.S. Pat. No.
19 5,849,902). The antisense molecule may be
complementary to a target sequence along a single
21 contiguous sequence of the antisense molecule or may
22 be in certain embodiments, bind to a substrate such
23 that the substrate, the antisense molecule or both
24 can bind such that the antisense molecule forms a
loop such that the antisense molecule can be 2 6 complementary to two or more non-contiguous
27 substrate sequences or two or more non-contiguous
28 sequence portions of an antisense molecule can be
29 complementary to a target sequence, or both.
Details of antisense methodology are known in the
31 art, for example see Schmajuk et al., 1999, J. Biol.
32 Chem., 274, 21783-21789, Delihas et al., 1997,
36
1 Nature, 15, 751-753, Stein et al., 1997, Antisense
2 N. A. Drug. Dev., 7, 151, Crooke, 2000, Methods
3 Enzymol., 313, 3-45; Crooke, 1998, Biotech. Genet.
4 Eng. Rev., 15, 121-157, Crooke, 1997, Ad.
Pharmacol., 4 0, l-'4 9.
6
7 A "triplex nucleic acid" or "triplex
8 oligonucleotide" is a polynucleotide or
9 oligonucleotide that can bind to a double-stranded
DNA in a sequence-specific manner to form a triple-
11 strand helix. Formation of such triple helix
12 structure has been shown to modulate transcription
13 of the targeted gene (Duval-Valentin et al., 1992,
14 Proc. Natl. Acad. Sci. USA, 89, 504).
16 Aptamers
17
18 Aptamers are nucleic acid (DNA and RNA)
19 macromolecules that bind tightly to a specific
molecular target. They can be produced rapidly
21 through repeated rounds of in vitro selection for
22 example by SELEX (systematic evolution of ligands
23 by exponential enrichment) to bind to various
24 molecular targets such as small molecules, proteins,
nucleic acids etc ( see Ellington and Szostak,
26 Nature 346 (6287) :818-822 (1990), Tuerk and Gold,
27 Science 249 (4968) :505-510 (1990) U.S. Patent No.
28 6, 867, 289; U.S.. Patent No. 5, 567, 588, U.S. Patent
29 No. 6, 699, 843).
31 In addition to exhibiting remarkable specificity,
32 aptamers generally bind their targets with very high
37
1 affinity; the majority of anti-protein aptamers have
2 equilibrium dissociation constants (Kds) in the
3 picomolar (pM) to low nanomolar (nM) range.
4 Aptamers are readily produced by chemical synthesis,
possess desirable storage properties, and elicit
6 little or no immunogenicity in therapeutic
7 applications.
8
9 Non-modified aptamers are cleared rapidly from the
bloodstream, with a half-life of minutes to hours,
11 ■ mainly due to nuclease degradation and renal
12 clearance a result of the aptamer's inherently low
13 molecular weight. However, as is known in the art,
14 modifications, such as 2'-fluorine-substituted
pyrimidines, polyethylene glycol (PEG) linkage, etc.
16 (can be used to adjust the half-life of the
17 molecules to days or weeks as required..
18
19 Peptide aptamers are proteins that are designed to
interfere with other protein interactions inside
21 cells. They consist of a variable peptide loop
22 attached at both ends to a protein scaffold. This
23 double structural constraint greatly increases the
24 binding affinity of the peptide aptamer to levels
comparable to an antibody's (nanomolar range). The
26 variable loop length is typically comprised of 10 to
27 20 amino acids, and the scaffold may be any protein
28 which has good solubility and compacity properties.
29 Aptamers may comprise any deoxyribonucleotide or
ribonucleotide or modifications of these bases, such
31 as deoxythiophosphosphate (or phosphorothioate),
32 which have sulfur in place of oxygen as one of the
38
1 non-bridging ligands bound to the phosphorus.
2 Monothiophosphates aS have one sulfur atom and are
3 thus chiral around the phosphorus center.
4 Dithiophosphates are substituted at both oxygens and
are thus achiral. Phosphorothioate nucleotides are
6 commercially available or can be synthesized by
7 several different methods known in the art.).
8
9 Treatment
11 "Treatment" or "therapy" includes any regime that
12 can benefit a human or non-human animal. The
13 treatment may be in respect of an existing condition
14 or may be prophylactic (preventative treatment).
Treatment may include curative, alleviation or
16 prophylactic effects.
17 ■
18 "Treatment of cancer" includes treatment of
19 conditions caused by cancerous growth and/or
vascularisation and includes the treatment of
21 neoplastic growths or tumours. Examples of tumours
22 that can be treated using the invention are, for
23 instance, sarcomas, including osteogenic and soft
24 tissue sarcomas, carcinomas, e.g., breast-, lung-,
bladder-, thyroid-, prostate-, colon-, rectum-,
26 pancreas-, stomach-, liver-, uterine-, prostate ,
27 cervical and ovarian carcinoma, non-small cell lung
28 cancer, hepatocellular carcinoma, lymphomas,
29 including Hodgkin and non-Hodgkin lymphomas,
neuroblastoma, melanoma, myeloma, Wilms tumor, and
31 leukemias, including acute lymphoblastic leukaemia
39
1 and acute myeloblastic leukaemia, astrocytomas,
2 gliomas and retinoblastomas.
. 3
(
4 The invention may be particularly useful in the
treatment of existing cancer and in the prevention
6 of the recurrence of cancer after initial treatment
7 or surgery.
8
9 Pharmaceutical Compositions
11 Pharmaceutical compositions according to the present
12 invention, and for use in accordance with the
13 present invention may comprise, in addition to
14 active ingredients, e.g (i) a chemotherapeutic agent
and/or an EGF inhibitor or (ii) an inhibitor of a
16 first EGF and an inhibitor of a second EGF, a
17 pharmaceutically acceptable excipient, a carrier,
18 buffer stabiliser or other materials well known to
19 those skilled in the art (see, for example,
(Remington: the Science and Practice of Pharmacy,
21 21st edition, Gennaro AR, et al, eds., Lippincott
22 Williams & Wilkins, 2005.). Such materials may
23 include buffers such as acetate, Tris, phosphate,
24 citrate, and other organic acids ; antioxidants;
preservatives; proteins, such as serum albumin,
26 gelatin, or immunoglobulins ; hydrophilic polymers
27 such aspolyvinylpyrrolidone ; amino acids such as
28 glycine, glutamine, asparagine, histidine, arginine,
29 or lysine ; carbohydrates; chelating agents;
tonicifiers; and surfactants.
40
1
2 The pharmaceutical compositions may also contain one
3 or more further active compound selected as
4 necessary for the particular indication being
treated, preferably with complementary activities
6 - - that do not adversely affect the activity of the
7 composition of the invention. For example, in the
8 'treatment of cancer, in addition to one or more EGF
9 inhibitors and/or a chemotherapeutic agent, the
formulation or kit may comprise an additional
11 component, for example a second or further EGF
12 inhibitor, a second or further chemotherapeutic
13 agent, or an antibody to a target other than the EGF
14 to which the said inhibitors bind, for example to a
growth factor which affects the growth of a
16 particular cancer.
17
18 The active ingredients (e.g. EGF inhibitors, for
19 example HB-EGF inhibitors, AREG inhibitors, and/or
chemotherapeutic agents) may be administered via
21 microspheres, microcapsules liposomes, other
22 microparticulate delivery systems. For example,
23 active ingredients may be entrapped within .
24 microcapsules which may be prepared, for example, by
coacervation techniques or by interfacial
26 polymerization, for example, hydroxymethylcellulose
27 or gelatinmicrocapsules and poly-
28 (methylmethacylate) microcapsules, respectively, in
29 colloidal drug delivery systems (for example,
liposomes, albumin microspheres, microemulsions,
31 - nano-particles and nanocapsules) or in
41
1 macroemulsions. For further details, see Remington:
2 the Science and Practice of Pharmacy, 21st edition,
3 Gennaro AR, et al, eds., Lippincott_Williams &
4 Wilkins, 2005.
6 Sustained-release preparations may be used for
7 delivery of active agents. Suitable examples of
8 sustained-release preparations include semi-
9 . permeable matrices of solid hydrophobic polymers
" containing the antibody, which matrices are in the
11 form of shaped articles, e. g. films, suppositories
12 or microcapsules. Examples of sustained-release
13 matrices include polyesters, hydrogels (for example,
14 poly (2-hydroxyethyl-methacrylate) , or poly
(vinylalcohol)), polylactides (U. S. Pat. No. 3,
16 773, 919), copolymers of L-glutamic acid andy ethyl-
17 Lglutamate,non-degradable ethylene-vinyl acetate,
18 degradable lactic acid-glycolic acid copolymers, and
19 poly-D- (-)-3-hydroxybutyric acid.
21 As described above nucleic acids may also be used in
22 methods of treatment. Nucleic acid for use in the
23 invention may be delivered to cells of interest
24 using any suitable technique known in the art.
Nucleic acid (optionally contained in a vector) may
26 be delivered to a patient's cells using in vivo or
27 ex vivo techniques. For in vivo techniques,
28 transfection with viral vectors (such as adenovirus,
29 Herpes simplex I virus, or adeno-associated virus)
and lipid-based systems (useful lipids for lipid-
31 mediated transfer of the gene are DOTMA, DOPE and
32 DC-Choi, for example) may be used (see for example,
42
1 Anderson et al., Science 256 : 808-813 (1992). See
2 also WO 93/25673 ).
3
4 In ex vivo techniques, the nucleic acid is
introduced into isolated cells of the patient with
6 the modified cells being administered to the patient
7 either directly or, for example, encapsulated within
8 porous membranes which are implanted into the
9 patient (see, e. g. U. S. Patent Nos. 4, 892, 538 10 and 5, 283, 187). Techniques available for
11^ introducing nucleic acids into viable cells may
12 include the use of retroviral vectors, liposomes,
13 electroporation, microinjection, cell fusion, DEAE-
14 dextran, the calcium phosphate precipitation method,
etc.
16
17 The EGF inhibitors and/or chemotherapeutic agent may
18 be administered in a localised manner to a tumour
19 site or other desired site or may be delivered in a
manner in which it targets tumour or other cells.
21 Targeting therapies may be used to deliver the
22 active agents more specifically to certain types of
23 cell, by the use of targeting systems such as
24 antibody or cell specific ligands. Targeting may be
desirable for a variety of reasons, for example if
26 the agent is unacceptably toxic, or if it would
27 otherwise require too high a dosage, or if it would
28 not otherwise be able to enter the target cells.
29
Administration
43
1
2 In embodiments of the invention, where an EGF
3. inhibitor and a chemotherapeutic agent are used in
4 treatment, the EGF inhibitor may be administered
simultaneously, separately or sequentially with the
6 chemotherapeutic agent. Likewise, in embodiments of
7 the invention, where a first EGF inhibitor and a
8 second EGF inhibitor are used in treatment together,
9 the first EGF inhibitor may be administered
simultaneously, separately or sequentially with the
11 second EGF inhibitor. Where administered separately
12 or sequentially, they may be administered within any
13 suitable time period e. g. within 1, 2, 3, 6, 12,
14 24, 48 or 72 hours of each other. In preferred
embodiments, they are administered within 6,
16 preferably within 2, more preferably within 1, most
17 preferably within 20 minutes of each other.
18
19 Kits
21 The invention further extends to various kits for
22 the treatment of cancer or the killing of tumour
23 cells. The kits may optionally include instructions
24 for the administration of each component, e.g. EGF
inhibitor and chemotherapeutic agent, or first EGF
26 inhibitor and second EGF inhibitor, separately,
27 sequentially or simultaneously.
28
2 9 Dose
44
1 The EGF inhibitors and/or chemotherapeutic agents of
2 and for use in the invention are suitably
3 administered to an individual in a "therapeutically
4 effective amount", this being sufficient to show
benefit to the individual. The actual dosage
6 regimen will depend on a number of factors ' including
7 the condition being treated, its severity, the
8 patient being treated, the agents being used, and
9 will be at the discretion of the physician.
11 In one embodiment of the methods, compositions or
12 kits, in which an EGF inhibitor and a
13 chemotherapeutic agent is used, the EGF inhibitor
14 and chemotherapeutic agent are administered in doses
which produce a potentiating ratio. Likewise, in
16 one embodiment of the methods, compositions or kits,
17 in which a first EGF inhibitor and a second EGF
18 inhibitor is used, the EGF inhibitor and
19 chemotherapeutic agent are administered in doses
which produce a potentiating ratio.
21
22 The term "potentiating ratio" in the context of the
23 present invention is used to indicate that two
24 components, e.g. EGF inhibitors,chemotherapeutic
agents etc. are present in a ratio such that the
26 cytotoxic activity of the combination is greater
27 than that of either component alone or of the
28 additive activity that would be predicted for the
29 combinations based on the activities of the
individual components.
45
1 Thus in a potentiating ratio, the individual
2 components act synergistically.
3
4 Synergism may be defined using a number of methods.
6 In one method, synergism may be determined by
7 calculating the combination index (CI) according to
8 the method of Chou and Talalay. CI values of 1, < 1,
9 and > 1 indicate additive, synergistic and
antagonistic effects respectively.
11
12 . In one embodiment of the invention, the EGF
13 inhibitor and the chemotherapeutic agent are present
14 in concentrations sufficient to produce a CI of less
than 1, such as less than 0.85. Likewise, in another
16 embodiment of the invention, the first EGF inhibitor
17 and the second EGF inhibitor are present in
18 concentrations sufficient to produce a CI of less
19 than 1, such as less than 0.85.
21 Synergism is preferably defined as an RI of greater
22 than unity using the method of Kern as modified by
23 Romaneli (1998a, b). The RI may be calculated as the
24 ratio of expected cell survival (Sep, defined as the
product of the survival observed with component A
26 alone and the survival observed with component B
27 alone) to the observed cell survival (Sobs) for the
28 combination of A and B(RI=Se/Sobs). Synergism may
29 then be defined as an RI of greater than unity.
31 In one embodiment of the invention, the EGF
32 inhibitor and the chemotherapeutic agent ( or the
46
1 first EGF inhibitor and the second EGF inhibitor)
2 are provided in concentrations sufficient to produce
3 an RI of greater than 1.5, such as greater than 2.0,
4 for example greater than 2.25.
6 Thus in one embodiment the combined medicament^
7 produces a synergistic effect when used to treat
8 tumour cells.
9
The optimal dose can be determined by physicians
11 based on a number of parameters including, for
12 example, age, sex, weight, severity of the condition
13 being treated, the active ingredient being
14 administered and the route of administration.
16 The invention will now be described further in the
17 following non-limiting examples with reference made
18 to the accompanying drawings in which:
19
Figure 1A illustrates analysis of AREG and beta
21 actin RNA expression in RKO +/+ with / without
22 either a 48 hour CPT11 treatment or 5-Fu treatment.
23 RNA levels were analyzed following 35 cycle of PCR
24 to determine relative differences in expression
between treated and untreated samples;
26
27 Figure IB illustrates analysis of AREG and beta
28 actin RNA expression in HCT116 +/+ with / without a
29 48 hour CPTll treatment. RNA levels were analyzed
following 35 cycles of PCR to determine relative
47
1 differences in expression between treated and
2 untreated samples;
3
4 Figure 2 illustrates western blot analysis of AREG
and gamma tubulin protein expression in HCT116 +/+
6 and RKO +/+ with / without a 48 hour CPT11 or 5-Fu
7 treatment;
8
9 Figure 3 illustrates confocal microscopy image of
AREG and Actin protein in HCT 116 +/+ with or
11 without CPT-11 treatment;
12
13 Figure 4 illustrates analysis of AREG protein
14 - expression in H630 p53 mutant colorectal cancer cell
lysates following a 48 hour CPT11 treatment. Western
16 blots were probed using an anti-amphiregulin
17 antibody. Enhanced AREG expression was observed
18 following chemotherapy (A and B illustrate two
19 separate experiments).
21
22 Figure 5 illustrates analysis of AREG and beta actin
23 RNA expression in H460 lung cancer cells with /
24 without either a 48 hour CPT11 treatment or 5-Fu
treatment. RNA levels were analyzed following 35
26 cycle of PCR to determine relative differences in
27 expression between treated and untreated samples.
28
48
1 Figure 6 illustrates AREG upregulation following
2 chemotherapeutic challenge in A) HT29, B) HCT116 and
3 C) MDA-MB231 cells. Cells were treated with/without
4 chemotherapy for 48 hours. RT-PCR was performed with
1 pg of total RNA using primer pairs specific for
6 the human AREG gene or GAPDH. The PCR products were
7 separated on 1.5% agarose gel electrophoresis and
8 visualized by ethidium bromide staining.
9 .
Figure 7a illustrates specific AREG silencing by
11 siRNA in HCT116. HCT116 cells were transfected with
12 AREG specific siRNA (lOnM), or a control siRNA
13 (lOnM). AREG and Beta-actin gene expression were
14 measured by RT-PCR RNA 72 hrs after transfection.
Figure 7b) illustrates specific HB-EGF silencing by
16 siRNA in HCT116. HCT116 cells were transfected with
17 HB-EGF specific siRNA (lOnM), or a control siRNA
18 (lOnM). HB-EGF and Beta-actin gene expression were
19 measured by RT-PCR RNA 72 hrs after transfection
21 Figure 8 illustrates HCT116 cell proliferation
22 following AREG/HB-EGF silencing by siRNA and/or
23 Chemotherapy treatment. Cells were transfected with
24 AREG siRNA (lOnM), HB-EGF siRNA (lOnM) or a control
siRNA (lOnM). Transfected cells were treated with no
26 drug or 3.5pM CPT-11. Cell proliferation was
27 analysed by MTT assay 48hr after
28 transfection/chemotherapy
29
49
1 Figure 9 illustrates specific AREG silencing by
2 siRNA in HT29 colorectal cancer cells. Cells were
3 transfected with AREG specific siRNA (lOnM), or a
4 control siRNA (lOnM). AREG and GAPDH gene expression
were measured by RT-PCR RNA 72 hrs after
6 transfection.
7
8 Figure 10 illustrates HT29 cell proliferation
9 following AREG silencing by siRNA and/or
Chemotherapy treatment. Cells were transfected with
11 AREG siRNA (lOnM) or a control siRNA (lOnM).
12 Transfected cells were treated with no drug or 3.5yM
13 CPT-11. Cell proliferation was analysed by MTT assay
14 48hr after transfection/chemotherapy.
16 Figure 11 illustrates Specific AREG silencing by
17 siRNA in MDA-MB231 cells. Cells were transfected
18 with AREG specific siRNA (lOnM), or a control siRNA
19 (lOnM). AREG and GAPDH gene expression were measured
by RT-PCR RNA 72 hrs after transfection.
21
22 Figure 12 illustrates MDA-MB2 31 cell proliferation
23 following AREG silencing by siRNA and/or
24 chemotherapy treatment. Cells were transfected with
AREG siRNA (lOnM), or a control siRNA (lOnM) and
26 varying doses of 5-FU. Cell proliferation was
27 analysed by MTT assay 48hr after
28 transfection/chemotherapy.
29
50
1 Figure 13 illustrates MDA-MB231 cell proliferation
2 following AREG/HB-EGF silencing by siRNA. Cells were
3 transfected with AREG siRNA (lOnM), HB-EGF siRNA
4 (lOnM) or a control siRNA (lOnM). Cell proliferation
was analysed by MTT assay 48hr after
6 transfection/chemotherapy.
7
8 Figure 14 illustrates amplification of amphiregulin
9 fragments from cDNA library. Amphiregulin was
amplified from kidney cDNA and PCR reaction was
11 analysed on 1.5% agarose gel stained with ethidium
12 bromide
13
14 Figure 15 illustrates colony PCR of AREG fragments.
PCR amplification was carried out colonies to
16 identify colonies that had the amphiregulin fragment
17 successful cloned into the expression vector. PCR
18 reaction was analysed on 1.5% agarose gel stained
19 with ethidium bromide. Any positive colonies were
selected for sequence and expression analysis
21
22 Figure 16 Panel A) illustrates the elution profile
23 of the purification of AREG recombinant protein from
24 500ml culture volume. Pellet from culture was
resuspended in 8M Urea and then purified by mature
26 of the 6xHistidine tag. The elution samples were
27 collected and analysed by SDS -PAGE (Panel B). The
28 gel was stained with comassie blue.
51
1 Figure 17 illustrates ELISA result of AREG
2 monoclonal antibodies produced. Monoclonal
3 antibodies were tested by ELISA against recombinant
4 AREG protein and a negative control produced in
similar method.
6
\
7 Figure 18 illustrates western blot analysis of
8 monoclonal test bleeds. The monoclonal test bleeds
9 were tested by western blot against the recombinant
amphiregulin protein and a negative control protein.
11 Equal amounts of protein were loaded on SDS-PAGE
12 gel.
13
14 Figure 19 illustrates western blot analysis of AREG
monoclonal antibodies against recombinant protein.
16 Recombinant AREG protein and a negative control
17 protein were run on SDS-PAGE gel. The gel was
18 transferred to nitrocellulose membrane and probed
19 with the AREG monoclonal antibodies.
21 Figure 20 illustrates western blot analysis of AREG
22 monoclonal antibodies against whole cell lysated
23 from colorectal cell lines HCT116 and HT29. Whole
24 cell lysates from HCT116 and HT29 cell lines were
prepared and ran on SDS-PAGE. Blots were probed with
26 AREG monoclonal antibodies a) 6E11 1E9 2D8 and b)
27 6E11 1E9 1C6. ( NB 6E11 1E9 2D8 has been
28 subsequently shown to be the sanme antibody as 6E11
29 1E9 1C6) .
52
1 Figure 21 illustrates confocal microscopy image of
2 AREG and actin protein in HCT 116 +/+ stained with
3 6E11 1E9 1C6 Monoclonal antibody
4
Figure 22 illustrates confocal microscopy image of
6 AREG and actin protein in HCT 116 +/+ stained with
7 6E11 1E9 2D8 Monoclonal antibody
8
9 Figure 23 illustrates FACS analysis in HCT116
colorectal cancer cell line treated with or without
11 2 . 5pM irinotecan for 48 hours.
12
13 Figure 24 illustrates FACS analysis in HCT116 cells
14 treated with or without 2.5pM irinotecan for 48
hours. Following treatment cells were stained with
16 AREG monoclonal antibodies and analysed by FACS
17
18 Figure 25 illustrates FACS analysis in H460 lung
19 carcinoma cell line treated with or without 2.5pM
irinotecan for 48 hours. Following treatment cells
21 were stained with AREG monoclonal antibodies and
22 analysed by FACS
23
24 Figure 26 illustrates MDA MB231 cell proliferation
after treatment with AREG antibody. Cell
26 proliferation was analysed by MTT assay 48hr after
27 treatment
28
53
1 Figure 27 illustrates MDA MB231 cell proliferation
2 after treatment with AREG antibody.' Cell
3 .proliferation was analysed by MTT assay 48hr after
4 treatment.
6 Figure 28 illustrates HCT116 cell proliferation
7 after treatment with AREG antibody. Cell
8 proliferation was analysed by MTT assay 48hr after
9 treatment.
11 Figure 2 9 illustrates MDA MB231 cell proliferation
12 after treatment. with AREG antibody. Cell
13 proliferation was analysed by MTT assay 48hr after
14 treatment.
16 Figure 30 illustrates HCT116 cell proliferation
17 after treatment with AREG antibody. Cell
18 proliferation was analysed by MTT assay 48hr after
19 treatment.
21 Figure 31 illustrates HCT116 cell proliferation
22 after treatment with AREG antibody. Cell
23 proliferation was analysed by MTT assay 48hr after
24 treatment
26 Figure 32 illustrates H460 lung carcinoma cell
27 proliferation following treatment with different
28 concentrations of AREG (6E11 1E9 1C6) antibody or an
29 isotype control antibody. Cells were seeded 24 hours
54
1 before treatment with either 25nM, 50nM or lOOnM
2 antibody. Cell viability was assayed 48 hours after
3 treatment by MTT assay.
4
Figure 33 illustrates HCT116 cell proliferation
6 following HB-EGF silencing by siRNA and/or treatment
7 with Anti AREG antibodies (6E11 1E9 2D8 & 6E11 1E9
8 1C6). Cells were transfected with HB-EGF siRNA
9 (50nM) or a control siRNA (50nM) .- Cell proliferation 10' was analysed by MTT assay 72hr after transfection.
11
12 Materials and Methods
13
14 Cell lines and culture conditions
16 The HCT116 (p53 wild type) human colorectal
17 adenocarcinoma cell line was maintained in McCoys
18 • (Invitrogen, UK). The RKO (p53 wild type) colorectal
19 carcinoma cell line, the MDA-MB231 human breast
carcinoma cell line and the HT2 9 human colorectal
21 carcinoma cell line were each maintained in
22 Dulbecco's Modified Eagle's Medium (DMEM,
23 Invitrogen, UK).
24 colorectal cell lines were maintained in Dulbecco's
Modified Eagle's Medium (DMEM, Invitrogen, UK). The
26 HH630 (p53 mutant) colorectal cancer cell lines were
27 maintained in Dulbecco's Modified Eagle's,Medium
28 (DMEM, Invitrogen, UK). The H460 (p53 mutant) lung
29 cancer cell lines were maintained in RPMI media
55
1 (Sigma Aldrich, UK). All medium was supplemented
2 with 10% FCS (normal (Invitrogen, UK) or dialysed
3 (Autogene Bioclear, UK)), 1% pen / strep, 1% L-
4 ' Glutamine (All Invitrogen, UK).
6 Xenograft models
7 6-8 week old female SCID mice were implanted with 2
8 x 106 HCT116+/+ human colorectal adenocarcinoma
9 cells into each flank. HCT116 cells in a log phase
of growth were harvested, washed in PBS and
11 resuspended in HBSS. They were mixed with equal
12 volumes of matrigel to give a final concentration of
13 5 x 106 cells / ml. Mice were randomly separated
14 into treatment groups on day 5 after implantation
and treated with different doses of chemotherapy. 5-
16 Fu (70 mgs/kg), CPT11 (70 mgs/kg) or saline control
17 solution and the mice have been sacrificed at
18 different time points (24 and 48 hrs after
19 injections). All drugs were administered through a
bolus injection. Animals were sacrificed at various
21 time points and tumours were removed for analysis
22
23 Microarray analysis
24 Approximately lOpg total RNA was isolated from
tumour cells and was used as the starting material
26 for preparation of probes. The microarray analysis
27 was carried using an Affymetrix U133 plus 2.0
28 GeneChip®. Probes were prepared as per the
29 manufacturers recommendations.
56
1 After RNA extraction, samples were reverse
2 transcribed into cDNA which was then purified on a
3 column prior to labelling. The probes were then
4 amplified and labelled using Oligo(dT)-primed in
vitro transcription generating high-yield,
6 biotinylated targets from the 3'-end. The cRNA was
7 fragmented to obtain optimal assay sensitivity and
8 then subjected to quality control to confirm that
9 fragment sizes range from 35-200 nucleotides. cRNA
was quantified on a spectrophotometer and the
11 quality of fragmented cRNA checked on a bioanalyser.
12 For the next stage the fragmented cRNA was
13 hybridised to the array for 16 hours at 45°C.
14 Following this the array was washed and stained with
streptavidin-phycoerythrin (SAPE) using a fluidics
16 station and scanned using a GeneChip® Scanner 3000.
17 Stained images were then analysed.
18
19 Initially the data was scaled using the Affymetrix®
GCOS (Genechip® Operating System) software, to
21 assess quality metrics. The data was then normalized
22 against a control sample. After normalisation the
23 data was filtered removing all genes where the noise
24 level obscured signal and were fold change was
greater than 2-fold. Finally a confidence filter
26 where the t-test p-value were used to filter the
27 genes to derive lists of statistically robust data.
28
29 Each treatment type and timepoint was carried out in
triplicate and statistics and filtering were applied
31 to the whole data set from each condition.
57
1
2 Chemotherapy treatment
3 a- Cell culture
4 Cells in a log phase of growth were seeded into T75
flask at -20% confluence and incubated overnight to
6 allow adherence to the plate. Wells were treated
7 with CPT11 (Irinotecan) and 5-Fu (Flourouracil) at
8 7 . 5|iM concentration for 48 hours. Chemotherapy was
9 substituted with saline in control flasks. After
different time exposure to chemotherapy the cells
11 were harvested, washed 3 times in PBS and total RNA
12 was isolated using the RNA TAT-60 reagent according
13 to the manufacturer instructions.
14 Reverse transcription was performed with 2.5 yg of
RNA using a High' Capacity cDNA Archive kit (Applied
16 Biosystem) according to the instruction of the
17 manufacturer.
18
19 b- organs
For in vivo toxicity study, mice were inoculated
21 with 2 x 106 HCT116+/+. human colorectal
22 adenocarcinoma cells into each flank. Mice were
23 randomly separated into treatment groups on day 5
24 after implantation and treated or not with 5Fu
(15mg/kg daily or 70mg/kg twice weekly). All drugs
26 were administered through a bolus injection. Animals
27 were sacrificed at various time points and organs
28 and tumor were removed for analysis.
58
1 Semi quantitative RT-PCR
2 Semi quantitative RT-PCR was performed using a PTC
3 225 Gradient Cycler (MJ Research Incorporated. The
4 PCR mixture, in a final volume of 25 pL, contains
12.5pL of 2x Biomix (Bioline,UK), 2 pL of primers
6 (10 pmol/L), 1 pL of cDNA and 9.5 pL of dH20) . PCR
7 conditions were initial denaturation step of' 95C for
8 10 minutes, followed by 35 cycles of 95°C for 30 sec
9 for denaturation; as annealing step 55°C for 30 sec;
and extension at 72°C for 90 sec, with a final
11 extension of 72°C for 10 minutes. 5 pi of amplified
12 product reactions was loaded onto a 1.5% agarose gel
13 (0.001% ethidium bromide) which was ran at 90V for
14 30 to 40 minutes prior to analysis on a UV box. A
Beta-actin control PCR amplification was performed
16 for each cDNA to check the level of cDNA charged in
17 the PCR mix.
18
19 For Example 2, Total RNA was isolated from cells '
following the RNA STAT-60 manufacturer's protocol
21 (Biogenesis, Poole, U.K.). RT-PCR was performed with
22 1 pg of total cell RNA using a Promega ImProm-II™
23 Reverse Transcription System (Promega, Southhampton,
24 UK). PCR was performed using primer pairs specific
for human AREG ( Forward 5'-
26 TTTTTTGGATCCCTCGGCTCAGGCCATTATGCTGCT-3'(SEQ ID
27 No:19), Reverse 5'-TTTTTTAAGCTTTACCTGTTCAACTCTGACTG-
28 3' (SEQ ID No:20)), human HB-EGF (Forward 5'-
29 TTTCTGGCTGCAGTTCTCTCGGCACT-3' (SEQ ID No:21),
Reverse 5'-CCTCTCCTATGGTACCTAAACATGAGAAGCCCC-3'(SEQ
31 ID No:22)), human GAPDH as a control (Forward
59
1 5'ACCACAGTCCATGCCATCAC-3' (SEQ ID No:23), Reverse 5'
2 TCCACCACCCTGTTGCTGTA-3'(SEQ ID No:24)) and human
3 Beta-actin as a control (Forward 5'-
4 ATCTGGCACCACACCTTTACAATGAGCTGCG-3' (SEQ ID No:25),
Reverse 5'-CGTCATACTCCTGCTTGCTGATCCACATCTGC-3'(SEQ
6 ID No:2 6)) .
7 The PCR products were separated on 1.5% agarose gel
8 and visualized by ethidium bromide staining.
9
Western blotting
11 a- Cell lysis
12 HCT 116, RKO, HT 29 and H630 are human colorectal
13 carcinoma cell lines. After different time exposure
14 to chemotherapy the cells were harvested, and
washed in 1 X PBS. The cell pellet was then lysed
16 in a' suitable amount of 1 X RIPA lysis buffer (150mM
17 NaCl, lOmM Tris at pH 7.2, 0.1% SDS, 1.0% triton X-
18 100 and 5mM.EDTA) supplemented with protease
19 inhibitors. The cell lysate was briefly vortexed,
incubated on ice for 10 min and then centrifuged at
21 12,000rpm to remove cell debris. Following
22 centrifugation, the lysate supernatant was removed
23 to a fresh eppendorf tube.
24
b- Quantitation of Whole Cell Lysates (WCL)
26 Protein concentrations were assayed using the BCA
27 Protein Assay (Pierce) according to the
28 manufacturer's instructions. The absorbance of each
29 sample at 620nm was assayed using a microplate
reader. A standard BSA curve was plotted for each
60
1 experiment and the protein concentration of each
2 sample calculated.
3
4 c- Preparation of Whole Cell Lysate (WCL) protein
samples
6 To each sample an equal volume of 5 X Western sample
7 buffer and 10% .of the final volume of |3-
8 mercaptoethanol (Sigma) was added. The samples were
9 denatured at 95°C for 5 minutes and placed on ice
prior to loading onto SDS polyacrylamide gel (SDS
11 PAGE). lOpl of pre-stained protein molecular weight
12 marker was loaded into one well of the gel.
13
14 d- Electro-transfer of proteins to Polyvinylidene
Flouride (PVDF) membrane
16 After electrophoresis the gel was placed in 1 X
17 Western transfer buffer. A piece of PVDF (Millipore)
18 was soaked in 100% methanol for 30 seconds then
19 washed with deionised H2O and equilibrated in 1 X
transfer buffer.
21
22 The above were then assembled into a Trans-blot SD
23 semi-dry transfer cell (Bio-Rad) as follows: One
24 piece of blotting paper soaked in transfer buffer,
the PVDF membrane, followed by the gel, then one
26 piece of blotting paper soaked in transfer buffer.
27 The proteins were then electrophoresed onto the
28 membrane at 20V for 90min.
29
61
1 e- Immunoblotting (Western blotting)
2 Following transfer, the PDVF membrane was washed
3 three times for 10 minutes with 1 X PBS/0.1% Tween
4 before being blocked for 1 hour in 1 X PBS/5% milk.
Once blocked the membrane was incubated with the
6 appropriate primary antibody at the relevant
7 dilution, in 1 X PBS/0.1 Tween/5% milk for 1 hour.
8 Following incubation the membrane was washed three
9 times with 1 X PBS/0.1% Tween before being probed
with the appropriate secondary antibody (Bio-Rad) at
•11 a dilution 1:5000 in 1 X PBS/5% milk for 1 hour. The
12 membrane was subsequently washed three times with 1
13 X PBS/0.1% Tween for 10 minutes each before
14 visualisation using Super Signal detection method
(Pierce), as described by the manufacturers. Protein
16 bands were detected by exposure to autoradiograph
17 which was subsequently developed. If detection of a
18 second protein was required from the same
19 immunoblot, the membrane was placed in western
stripping buffer, incubated for 30 min in a 50°C
21 rocking incubator. Following membrane stripping it
22 was washed in 1 X PBS/0.1% Tween, 5 times, for 10
23 min periods. The membrane was reprobed, as before
24 with the appropriate antibodies.
2 6 RNA interference
27 AREG, HB-EGF and Control siRNAs and Dharmafect 4
28 transfection reagent were obtained from Dharmacon,
29 (Lafayette, CO,USA).
62
1 For HB-EGF, the siRNAs used had the following
2 sequences:
3 1 Sense sequence
4 GAAAAUCGCUUAUAUACCUUU (Sequence ID No: 1)
lAnti-sense sequence
6 AGGUAUAUAAGCGAUUUUCUU(Sequence ID No: 2)
7
8 2 Sense Sequence
9 UGAAGUUGGGCAUGACUAAUU(Sequence ID No: 3)
2 Anti-sense sequence
11 UUAGUCAUGCCCAACUUCAUU(Sequence ID No: 4)
12
13 3 Sense sequence
14 GGACCCAUGUCUUCGGAAAUU(Sequence ID No: 5)
3 Antisense sequence
16 UUUCCGAAGACAUGGGUCCUU(Sequence ID No: 6)
17
18 4 Sense Sequence
19 GGAGAAUGCAAAUAUGUAUU(Sequence ID No: 7)
4 Anti-sense Sequence
21 UCACAUAUUUGCAUUCUCCUU (Sequence ID No: 8)
22
23 For AREG, the siRNA sequences used had the following
24 sequences:
1 Sense Sequence
26 UGAUAACGAACCACAAAUAUU(Sequence ID No: 9)
27 1 Anti-Sense Sequence
28 UAUUUGUGGUUCGUUAUCAUU(Sequence ID No: 10)
29
2 Sense Sequence
31 UGAGUGAAAUGCCUUCUAGUU(Sequence ID No: 11)
32 2 Anti-sense Sequence
33 CUAGAAGGCAUUUCACUCAUU(Sequence ID No: 12)
34
3 Sense Sequence
36 GUUAUUACAGUCCAGCUUAUU(Sequence ID No: 13)
37 3 Anti-Sense Sequence
38, UAAGCUGGACUGUAAUAACUU(Sequence ID No: 14)
39
40 4 Sense Sequence
41 GAAAGAAACUUCGACAAGAUU(Sequence ID No: 15)
42 4 Anti-sense Sequence
43 UCUUGUCGAAGUUUCUUUCUU(Sequence ID No: 16)
44
63
1 Cells were seeded at 5 000 cells per well in a 96
2 well plate or 5x 105 cells per well in a 6 well
3 plate. The cells were cultured for 24 hours before
4 transfection. The siRNA was made up to lOOnM in
serum free DMEM and left for 5 minutes at room
6 temperature. The Dharmafect transfection reagent was
7 also made up in the serum free DMEM and incubated
8 for 5 minutes at room temperature. The transfection
9 reagent was added to the siRNA and incubated at room
temperature for 20 minutes. The media was removed
11 from the plate wells and antibiotic free DMEM was
12 added to the wells. After 20 minutes the siRNA was
13 added dropwise to the wells. The plates were
14 incubated at 37DC for 48 hours.
16 MTT assay
17 Cell viability was assessed by 3-(4, 5-
18 dimethylthiazol-2-yl)-2,5-diphenyltetrazolium
19 bromide(MTT, Sigma) assay (Mosmann, T. 1983. J.
Immunol. Methods 65:55-63). To assess chemotherapy/
21 siRNA interactions 5000 cells were seeded per well
22 on 96 well plate. After 24 hours cells were
23 transfected with siRNA and treated with various
24 chemotherapeutic agents at different concentrations.
After 48 hours MTT (l.Omg/ml) was added to each well
26 and cells were incubated at 37°C for 2 hours. The
27 culture media was removed and formazan crystals were
28 reabsorbed in 200pl DMSO. Cell viability was
29 determined by reading the absorbance of each well at
570nm using a microplate reader (Tecan Sunrise,
31 Biorad, UK).
64
1
2 Cloning of Amphiregulin(AREG)
3 The DNA sequence encoding the amphiregulin protein
4 was amplified by PCR from a cDNA library using gene-
specific primers. The AREG gene was cloned into the
6 bacterial expression vector pETlOO allowing the
7 incorporation of a hexahistidine tag onto the N-
8 terminus of the recombinant protein. This construct
9 was then used to transform competent TOPIOF' E.coli
cells (Invitrogen). Positive transformants were
11 selected by colony PCR using vector-specific primers
12 flanking the multiple cloning site.
13
14 Expression of recombinant AREG protein
The positive clones were propagated overnight at 37
16 °C in 5 mis of Luria-Bertani (LB) broth supplemented
17 with 50 |J,M ampicillin. A 300 |_ll aliquot of this
18 culture was retained for inoculation of secondary
19 cultures and the remainder of the sample was
miniprepped using the Qiagen miniprep kit and the
21 sequence verified by DNA sequencing.
22
23 Three secondary cultures were inoculated to allow
24 visualisation of protein expression. The cultures
were induced with IPTG (final concentration 1 mM)
26 when the cultures had an OD of 0.2, 0.5 and 1.0
27 (A55o) respectively and then left for 4 hrs at 37 °C.
28 The cells were then harvested by centrifugation at
29 4000 rpm for 15 mins and the pellet resuspended in 1
ml of PBS/0.1 % Igepal supplemented with 1 p.1 of
31 lysonase. Samples were then analysed by SDS-PAGE and
32 western blotting to confirm expression of the
65
1 protein. The SDS-PAGE gel was stained overnight in
2 coomassie blue and destained the following day.
3
4 The recombinant AREG protein was then expressed in
500 mis of LB broth supplemented with ampicillin,
6 using the secondary culture as an inoculant and
7 induced with IPTG once the culture had reached the
8 optimal optical density. The culture was centrifuged
9 at 5000 rpm for 15 mins and the pellet retained for
protein purification.
11
12 Protein Purification
13 The induced recombinant protein was solubilised in
14 50 mis of 8 M urea buffer (480g Urea, 29g NaCl,
3.12g NaH2P04 (dihydrate), 0.34g Imidazole)
16 overnight.. The solution was centrifuged at 6000 rpm
17 for 1 hr, after which the supernatant was filtered
18 using 0.8 |am gyrodisc filters before purification.
19
The protein was purified by its N-terminal
21 hexahistidine tag and refolded using on-column
22 . refolding by immobilized metal affinity
23 chromatography. Chelating hi-trap columns (Amersham
24 Biosciences) were charged using 100 mM nickel
sulphate before attachment to the Aktaprime.
26 Refolding takes place by the exchange of the 8 M
27 urea buffer with a 5 mM imidazole wash buffer (29g
28 NaCl, 3.12g NaH2P04 (dihydrate) 0.34g Imidazole, pH
29 8.0 ) and elution of the protein using a 500 mM
imidazole elution buffer (29g NaCl, 3.12g NaH2P04
31 (dihydrate), 34g Imidazole). The elution profile of
66
1 the purified recombinant protein was recorded and
2 can be seen in figure 16.
3
4 The eluted fractions were subjected to SDS-PAGE
analysis to confirm recombinant protein presence in
6 eluted fractions. The gels were stained with
7 coomassie blue overnight and subsequently destained
8 to determine the fractions containing the AREG
9 protein.
11 Antibody generation
12 The refolded protein was used as an immunogen to
13 generate monoclonal antibodies. Five BALB/C mice
14 were immunized at three weekly intervals with 150 |Llg
of purified recombinant protein and the antibody
16 titre was analysed after boosts three and five. A
17 test bleed was taken from each animal and tested at
18 1:1000 dilutions in western blotting against 100 ng
19 of antigen. Blots were developed using 3,3'-
diaminobenzidine (DAB).
21
22 After the fifth boost, the spleen was removed from
23 the mouse and the antibody producing B cells were
24 fused with SP2 myeloma cells following standard
protocols. Eleven days after the hybridoma fusion,
26 the plates were examined for cell growth. Clones
27 were screened by ELISA against recombinant protein
28 and selected positive hybridomas were cloned twice
29 by limiting dilution.
67
1 ELISA
2 The monoclonal antibodies were screened by ELISA to
3 determine which clones should be expanded. Maxi Sorb
4 96 well plates were coated with recombinant antigen
by adding 100 J0.1 of coating buffer (Buffer A: 0.42g
6 sodium bicarbonate/100|il H2O, Buffer B: 0.53g sodium
7 carbonate/100|i.l H2O, pH 9.5) containing the
8 screening antigen to each well (100 ng/well). A
9 control antigen was also used to eliminate non-
specific clones. The plates were incubated at 37 °C
11 for 1 hr to allow the antigen to bind to the well
12 and then blocked for 1 hr at room temperature by
13 adding 200 |0.1 PBS/3 % BSA to each well.
14
The blocking solution was removed from the plates
16 . and 100 |Xl of hybridoma supernatant was added to a
17 positive antigen and a control antigen well. The
18 screening plates were incubated with supernatant for
19 1 hr on a rocker at room temperature. The plates
were washed three times with PBS-T, after which 100
21 |J.l of goat anti-mouse HRP conjugated secondary
22 antibody (1:3000) was added to each well and
23 incubated for 1 hr at room temperature. The plates
24 were washed three times with PBS-T and 100 |Lll of
3, 3' ,5,5'-tetramethylbenzidine (TMB) was added to
26 each well and incubated for 5 mins at 37 °C.
27 Positive wells were indicated by a colour
28 development and the reaction was stopped by addition
29 of 50 |J,1 1M HCL. Plates were read by a
spectrophotometer at 450 nm and samples displaying a
31 positive reading in the screening well (+) with a
68
1 negative reading in the control well (-) were chosen
2 for further work. The cells from the original wells
3 were transferred into a 24 well plate and grown up.
4
Western blotting
6 The supernatants from the hybridoma cell lines were
7 analysed by western blotting to determine the
8 ability of the monoclonal antibodies to detect both
9 recombinant AREG and endogenous native AREG protein
in a range of cancer cell lines, which are
11 representative of AREG expression in cancer.
12 Aliquots of HCT116 and HT29 whole cell lysates (-30
13 (0,g/ml) or recombinant AREG protein were separated by
14 SDS-PAGE and transferred onto Hybond-C Extra
nitrocellulose membrane (Amersham Biosciences). The
16 membrane was blocked by incubation in PBS / 5 %
17 marvel for 1 hr at room temperature, after which it
18 was rinsed briefly in PBS. The monoclonal antibodies
19 were used at a 1:500 or 1:250 dilutions in PBS and
incubated on the membrane overnight at 4°C while
21 gently rocking. The blot were then rinsed three
22 times with PBS / 1 % marvel and 0.1% Tween-20 and
23 then incubated with the goat anti-mouse HRP
24 conjugated secondary antibody at a 1:3000 dilution
for 1 hr at room temperature while shaking. The
26 i blots were then rinsed three times with the PBS / 1
27 % marvel and 0.1 % Tween-20 solution, followed by a
28 short rinse in PBS. The blots were incubated with
29 ECL plus substrate (Amersham Bioscicences) for 5
mins at room temperature prior to analysis on the
31 Kodak imager.
69
1 Flow Cytometry Analysis
2 HCT116 or H460 cells were treated for 48 hours with
3 or without 2.5pM irinotecan. After 48 hours cells
4 were washed in PBS and blocked for 20 minutes in
Normal Goat Serum. 5x10s cells were incubated with
6 AREG antibodies or isotype control for 2 hours and
7 washed in PBS-T. The cells were incubated with a
8 FITC conjugated goat anti-mouse antibody for 1 hour
9 and washed in PBS-T before analysis on BD FACS
canto.
11
12 Results
13
14 Example 1
A xenograft study was set up to examine the genetic
16 response to 2 different chemotherapeutic drugs 24
17 and 48 hours after treatment. Each mouse was
18 implanted with equal volumes of HCT116 cells and
19 each condition was performed in triplicate. 4 groups
of three mice were administered of lOOul CPT-11
21 (70mg/kg), 5-FU (70mg/kg) or saline control. Tumours
22 were then resected after 24h (5-FU) & 48h (CPT-11,
23 5-FU) . Average mass of the tumours did not vary over
24 control and drug treated groups.
26 ' RNA isolated from tumours in each of the 1'2 mice was
27 subjected to microarray analysis to measure mRNA
28 expression levels. Fold change values for drug
29 treated against untreated control is presented.
After 48 hours, the fold change values for AREG mRNA
31 expression in 5FU treated against untreated controls
70
1 was 2.1 with the fold change values for AREG mRNA
2 expression in CPTll treated against untreated
3 controls being 2.2. The data was passed through
4 stringent statistical filters and is considered
statistically robust. The amphiregulin RNA was
6 significantly upregulated greater than 2 fold
7 relative to control.
8
9 Five other ErbB cognate ligands have also been found
to be up-regulated by our micro-array analysis. TGF
11 and HB-EGF protein showed up-regulation when treated
12 by 5-FU. EREG protein showed up-regulation after 48h
13 treatment with both CPT-11 and 5-FU. BTC protein
14 showed up-regulation in all 3 conditions. NRG3 was
upregulated after 48h treatment with both CPT-11 and
16 5-FU. The results are summarised in Table 1.
17
18 The genes were selected for further study as a
19 potential target for antagonists. To validate the
expression data observed in the microarray semi
21 quantitive RT-PCR was carried out for the gene. RT-
22 PCR was carried out on RNA extracted from
23 colorectal cell lines (including HCT116, RKO, HT29 &
24 H630) following exposure to a relevant range of
chemotherapeutic treatments
26
27 Results for the selected target using Q-PCR
28 validated the results observed in the microarray
29 analysis. AREG upregulation was validated in RKO
cell lines 48h after treatment with CPT-11 and 5-FU
71
1 and in HCT116 cells 48h after treatment with 5-FU
2 (FIG 1) .
3
4 To make a suitable target for an AREG inhibitor
preferential upregulation should be observed in
6 tumour tissue when compared to other vital organs.
7 For this experiment the inventors have used mouse
8 homologues of the targets and examined regulation in
9 mice organs, for the gene. None of the targets
analysed displayed upregulation in the mouse organs
11 examined which suggests the chemotherapeutic
12 treatment has a more acute affect on expression in
13 cancer cells than stable tissue.
14
To show that target upregulation observed at the
16 mRNA level was mirrored at the protein level western
17 blot analysis was performed. AREG protein expression
18 in RKO and HCT116 p53 wild type colorectal cancer
19 cell lysates was analysed following a 48 hour CPTll
or 5FU treatment. Western blots were probed using an
21 anti-AREG antibody. Enhanced AREG expression was
22 observed, following CPTll treatment with both the
23 HCT116 and RKO cell lines (FIG 2) .
24 Confocal microscopy was used to analyse the in vitro
effects of CPT-11 treatment (24h and 48h) of HCT116
26 cells on AREG expression levels. The inventors
27 observed increasing levels of expression at each
28 time point when compared to untreated controls (FIG
29 3) .
72
1 Figure 4 illustrates analysis of AREG protein
2 expression in H630 p53 mutant colorectal cancer cell
3 lysates following a 48 hour CPTll treatment. Western 4. blots were probed using an anti-AREG antibody.
Enhanced AREG expression was observed following
6 chemotherapy (A and B illustrate two separate
7 experiments)when compared to controls. This data
8 demonstrates that CPT-11 (or indeed analogues or
9 metabolites thereof) in combination with an ErbB
cognate ligand (as shown to be upregulated) can be
11 used for the treatment of p53 mutant cancers.
12
13 Figure 5 illustrates analysis of AREG and beta actin
14 RNA expression in H460 lung cancer cells with /
without chemotherapy (either a 4 8 hour CPTll
16 treatment or 5-Fu treatment). RNA levels were
17 analyzed following 35 cycle of PCR to determine
18 relative differences in expression between treated
19 and untreated samples. This data shows an enhanced
expression of AREG following both CPT-11 and 5-Fu
21 challenge.
22
23 Table 1 Result of microarray analysis performed on
24 5FU & CPT-11 treated HCT116+/+ cells (In Vivo) for
the different members of the EGF-family protein
73
Gene
24h 5-FU
48h 5-FU
48h CPT-11
EGF
-2 . 4
-3.8
-2.5
(Epidermal
Growth factor)
TGF alpha
1.3
2.2
-2.1
(Transforming
1.8
1.0
-1.2
Growth factor
alpha)
BTC
1.1
1.9
2.4
(Betacellulin)
HB-EGF
1.6
1.6
1.0
(Heparin-
3.1
-1.1
-1.3
Binding EGF-
like growth
factor)
EREG
-1.5 '
1.9
1.5
(Epiregulin)
1.0
2.1
1.5
NRG1
N
0
T
NRG 2
-2 .1
-1.5
-1.5
-2 . 5
-4 . 7
1.5
NRG 3
-2.1
1.6
1.2
NRG 4
-1.1
-1.8
1.2
74
1 This chemotherapy induced up-regulation of AREG has
2 been observed at both the mRNA and protein level
3 using p53 wt and mutant colorectal cancer cell lines
4 (HCT116+/+; RK0+/+ and H630). AREG has also been
shown to be up-regulated at the mRNA level in H460
6 lung cancer cells and MDA breast cancer cells which
7 indicates that this effect may be observed over a
8 range of AREG expressing cancers.
9
As seen from the molecular analysis, these proteins
11 are selectively expressed after chemotherapy
12 treatment in different carcinoma cell lines. This
13 indicates that cancer cells, as a response to a
14 chemotherapy challenge, over-express six different
growth factors of the same family. This response
16 seems to be a way used by the cancer cells to
17 overcome chemotherapy insult. By selectively
18 targeting these proteins, their role in cancer cell
19 survival may be at least reduced and at best
inhibited, which may lead to a reduction in tumour
21 growth. Furthermore, the simultaneous targeting of
22 two or more over expressed ligands (by an antagonist
23 molecule like an antibody) may provide a useful
24 therapeutic strategy.
2 6 Example 2
27 Chemotherapy induced AREG up-regulation in
2 8 colorectal and breast cancer cell lines.
29
AREG up-regulation was further validated in several
31 carcinoma cell lines. In human HT29 colorectal
75
1 cancer cells and human HCT116 colorectal cancer
2 cells AREG mRNA up-regulation was observed after
3 treatment with IC50 dose of CPT 11 (Figure 6A and
4 6B). Moreover, after treatment with IC50 dose of 5-
FU in human MDA-MB231 breast carcinoma cell line up-
6 regulation of AREG mRNA was shown (Figure 6C) .
7
8 Silencing of AREG and HB-EGF in cancer cells
9
siRNA potently down-regulated expression of AREG
11 (Figure 7A) and HB-EGF (Figure 7B) in HCT116
12 . colorectal cell line in comparison to untreated
13 cells, mock transfection and control siRNA. In
14 Figure 9 and Figure 11 respectively, AREG knockdown
is also shown in HT29 colorectal cancer cells and in
16 MDA-MB2 31.
17
18 Synergistic attenuation of cell growth after
19 treatment with siRNA and chemotherapy in colorectal
cancer
21
22 Following confirmation of AREG and HB-EGF silencing
23 by siRNA, MTT assays were performed to investigate
24 the effect of down-regulation of these two genes on
cell growth.
26
27 AREG siRNA alone, HB-EGF siRNA alone and monotherapy
28 of CPT-11 had no significant effect on cell
29 viability compared to untreated cells, mock
76
1 transfection and control siRNA. However, co-
2 treatment of HCT116 with AREG siRNA and CPT-11
3 resulted in synergistic decreases in cell viability.
4 The same effect was observed when AREG siRNA was
replaced with HB-EGF siRNA (Figure 8).
6 In another colorectal cancer cell line, HT29 similar
7 results were obtained as those observed with HCT116.
8 AREG siRNA alone and control siRNA alone had no
9 significant effect on the growth of the cells. The
combination of AREG siRNA and CPT-11 had a
11 synergistic effect on cell viability resulting in
12 the decrease of cell growth (Figure 10) .
13 Collectively these results indicate that down-
14 regulation of AREG/HB-EGF expression in combination
with chemotherapy had a significant effect on the
16 attenuation of cell growth in colorectal cancer.
17
18 Synergy between silencing of AREG and treatment with
19 chemotherapy led to an attenuation of cell growth in
breast cancer
21
22 Following transfection with control siRNA alone,
23 transfection reagent alone (mock) and chemotherapy
24 treatment, a 20% reduction in cell growth was
observed. A further 23% decrease was observed when
26 cells were transfected with AREG siRNA alone.
27 Treatment with varying doses of 5-FU (2.5 - 6pM)
28 showed similar results to that of AREG siRNA alone.
29 However, treatment with 7.5jiM 5-FU in combination
with AREG siRNA lead to a further 20% reduction in
77
1 growth (60% overall reduction in growth in
2 comparison to the untreated, Figure 12).
3
4 When siRNA experiments and MTT assays were performed
using a combination of AREG and HB-EGF the inventors
6 surprisingly observed a marked reduction in cell
7 growth. Experiments were performed in the MDA-MB2 31
8 breast cancer cell line, to assess if knocking down
9 AREG and HB-EGF had any affect on cell viability in
breast cancer cells.
11
12 Remarkably, co-silencing AREG and HB-EGF resulted in
13 a significant decrease on cell viability compared to
14 controls. A decline in cell growth of ~75% was
observed when MDA-MB231 cells were co-treated with
16 target siRNA compared to controls figure 13.
17
18 Development of AREG specific monoclonal antibodies.
19
A panel of murine monoclonal antibodies were raised
21 against recombinant human amphiregulin (Figure 14-
22 16). They were characterised by ELISA, western
23 blotting (whole cell lysates from colorectal cell
24 lines HCT116 and HT29) and confocal microscopy
analysis for demonstration of specific recognition
26 of AREG (Figure 17-22).
27
28 Up-regulation of AREG as shown by FACS analysis 2 9 using AREG monoclonal antibodies on colorectal 30 cancer and lung carcinoma cell lines.
78
1 Flow cytometry analysis of HCT116 cells and H460
2 cells shows cell surface recognition of AREG when
3 assessed using the Anti AREG monoclonal antibodies
4 (Figure 23 shows FACS analysis of HCT116 cells
treated with AREG clones 4G5 and 6E11 and an isotype
6 control). Furthermore, cells treated with 2.5pM
7 irinotecan for 48 hours prior to analysis showed up
8 to a 40% increase in the cell surface expression of
9 AREG. The Anti-AREG clone 6E11 1E9 detected a 20%
increase in AREG expression in HCT116 cells after
11 treatment with irinotecan (Figure 24).
12 FACS analysis has also been carried out on the lung
13 carcinoma cell line H460. Two clones namely 3H5 and
14 3F8 both detected AREG expression on the surface and
up-regulation in expression levels after treatment
16 with 2.5yM irinotecan (Figure 25).
17 These results demonstrate that the inventors' panel
18 of AREG monoclonal antibodies recognise the AREG
19 protein on the surface of the HCT116 cells.
21 Attenuation of cell growth in cancer cells after
22 treatment with AREG monoclonal antibodies.
23 MTT assays were performed on cancer cell lines, MDA-
24 MB231 breast cancer cells (Figure 26 & Figure 27)
and HCT116 colorectal cancer cells (Figure 28) to
26 ascertain the effect of AREG monoclonal antibodies
27 on cell growth. The activities of different clones
28 were screened on the MDA-MDB231 cell line with
29 several showing -40% reduction in cell growth when
compared to untreated cells. In HCT116 cells a more
31 pronounced effect was observed with -60% reduction
32 in the cell viability observed with clones 4G5 and
79
1 6E11. Figure 29 shows similar effects on cell
2 growth in the MDA-MB231 breast cancer cells with
3 AREG clone 6E11 1E9 2D8 ( which has been shown to be
4 the same clone as 6E11 1E9 1C6) . Figure 30 and 31
shows the effect of AREG antibodies in the HCT116
6 cell line. A 65% decrease in cell viability is
7 observed in Figure 30 and Figure 31 shows a 50%
8 decrease in cell growth. Figure 32 shows the effect
9 of the AREG antibody 6E11 1E9 1C6 in the lung cancer
H460 cell line, comparing the effect against a
11 isotypic control antibody and showing that the
12 antibody significantly attenuates cell viability of
13 the lung cancer cells. Together these results show
14 that the AREG monoclonal antibodies have a
significant effect on the cell viability of AREG
16 expressing cancer cells including colorectal, lung
17 and breast carcinoma.
18
19 Synergistic attenuation of growth on colorectal 2 0 cancer cells after treatment with HBEGF siRNA and
21 AREG antibodies.
22 The effect of down-regulation of the HBEGF siRNA in
23 combination with an AREG antibody was investigated
24 by performing an MTT cell viability assay (Figure
33). The HBEGF siRNA alone had a slight reduction on
26 cell growth while the AREG antibody 6E11 1E9 2D8 had
27 -50% reduction in cell growth. When the HBEGF siRNA
28 and 6E11 1E9 2D8 was added in combination a further
29 reduction in cell viability was observed. To see if
this effect was synergistic the RI values as
31 described by Kern 1988 and modified by Romanelli
32 1998 were calculated. The RI value is calculated as
80
1 the ratio of expected survival (Sexp defined as the
2 product of the survival observed with drug A alone
3 and the survival observed with drug B alone) to the
4 observed cell survival (S0bs) for the combination of A
and B. (RI=Sexp/S0bs) • Synergism is defined as RI>1.
6 The RI value for 6E11 1E9 2D8 was approximately 2.
7 However, the RI value for 6E11 1E9 1C6 was
8 calculated as being above 5. Collectively these
9 results show that the combined targeting of both
HBEGF and AREG in the treatment of cancers
11 associated with Erb ligands or EGF (including
12 colorectal, breast and lung) results in a
13 synergistic attenuation of cell growth.
14
The development of non-responsive tumours or
16 chemotherapy resistant cancer remains a major
17 obstacle to successful treatment. There is a clear
18 need for tools which enable prediction of whether a
19 particular therapy either single or combination will
be effective against particular tumours. Moreover,
21 there remains the need for new treatment regimes to
22 increase the repertoire of treatments available.
23 Combined therapies have shown promising results by
24 improving the response rates in patients by acting
on the tumours through different mechanisms, the
26 inventors' data suggests inhibitory molecules, for
27 example antagonist antibodies, specific to AREG and
28 HB-EGF used alone or in combination with
29 chemotherapy could potentially be used to treat a
wide variety of aggressive cancers including
31 colorectal, lung and breast cancer.
81
1
2 All documents referred to in this specification are
3 herein incorporated by reference. Various
4 modifications and variations to the described
embodiments of the inventions will be apparent to
6 those skilled in the art without departing from the
7 scope and spirit of the invention. Although the
8 invention has been described in connection with
9 specific preferred embodiments, it should be
understood that the invention as claimed should not
11 be unduly limited to such specific embodiments.
12 Indeed, various modifications of the described modes
13 of carrying out the invention which are obvious to
14 those skilled in the art are intended to be covered
by the present invention.
16
17
18
Claims (19)
1. Use of an inhibitor of HB-EGF' and an. inhibitor of AREG in the preparation of a medicament formi d for siinultaiieousor sequential use in the combination treatment of neoplastic disease; wh i, in said combination treatment, said inhibitor of HBEGF and said inhibitor of AREG act syn wherein said in! IGF is an antibody molecule which binds said HB-EGF or a nucleic acid molecule w hibits expression of HB-EGF; and wherein said inhibitor of AREG is an antibody molecule which binds AREG or a nucleic acid molecule which inhibits expression of AREG,
2. Use of an inhibitor of HB-EGF and an inh r of AREG in the preparation of a medicament for the combination treatment of neoplastic disease, wherein said medicament is formu ministration according to a dosage regime comprising administering said inhibitor of HB-EGF to a patient, where said inhibitor of .AREG was administered within the previous 72 hours, or said inhibitor of A,REG is administered simultaneously or within the subsequent 72 hours; 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 13 20 21 22 23 24 25 26 27 28 29 30 31 Received at IPONZ on 29.02.2012 83 wherein, with said dosage regime, said inhibitor of HB-EGF and said irxl act synergistically; wherein said inhibitor of HB-EGF is an antibody molecule which binds said HB-EGF or a nucleic acid molecule which inhibits expression of HB-EGF; and wherein said inhibitor of AREG is an antibody molecule which binds AREG or a n* c acid molecule which inhibits expression 3.
3, The use according to claim 1 or claim. 2, wherein said inhibitor of HB-EGF is a first siRNA and /or said inhibitor of AREG is a second siRNA.
4, The use according to claim 1 or claim. 2, wherein the inhi G is an anti AREG antibody molecule,
5. The use according to claim. 4, wherein the inhibitor of AREG is an antibody molecule which comprises a variable region having the amino acid sequence of Sequence ID No: 27 and a variable region, having the amino acid, sequence of S< ce ID No: 28.
6. The use according to any one of claims 1 to 5, wherein the medicament is for simultaneous or sequential use in combination treatment with a 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 Received at IPONZ on 29.02.2012 84 chemotherapeutic agent. 7, The use according to claim 6, wherein the chemotherapeutic agent is selected from the group consisting of antinw , topoisomerase inhibitors, alkylating agents, anthracyclines, and plant alka.; 8, The use according to claim 7, wherein the-; chemotherapeutic agent is CPT-11, 9, The use according to claim 8, wherein the chemotherap : agent is 5-FU. 10, T! ording to any one of the claims 1 to 9, wherein said neopt^r. t ise is selected frona the group cc of colorectal cancer, breast cancer and lung cancer. 11, The use according to any one of claims 1 to 10, wherein, the net dsease is a. cancer comp Lg a p53 mutation. 12, A pharmaceutical composition comprising (i) an inhibitor of HB-EGF and (ii) an inhibitor of AREG,- wherein said inhibitor of AREG is an dy molecule which binds said AREG or a nucleic acid r ale which inhibits expression of said AREG; and wherein said inh >r of HB-EGF is an -body molecule which binds said HB-EGF or 1 2 3 4 5 6
7
8
9
10
11
12 13 14 15 16 17 18 19 20 21 22 23 24 25 26 27 28 29 30 31 32 Received at IPONZ on 29.02.2012 85 a nucleic acid molecule which inhibits expression of HB-EGF.
13. The composition according to claim 12, wherein the inhi 6 is an anti AREG antibody molecule.
14. The composition according to claim 12, whei:ein the inhibitor of AREG is an antibody molecule which comprises a variable region having the ami ce of Sequence ID No: 27 and a variable region having the amino acid sequence of Sequence ID No; 28.
15. The composition according to claim 12, wherein s-ho bnibitor of HB-EGF is a first siRNA and/or said in! of AREG is a second siRNA..
16. The use according to claim 1, or the composition according to claim 12, wherein the inhibitor an antibody molecule comprising at least one of the CDRs of the VH region having the amino acid sequence shown as Sequence ID No: 27 and/or at least one of the CDRs of the VL region having the amino acid sequence shown, as Sequence ID No: 28, wherein the antibody molecule has binding sp G.
17. The use or composition according to claim, 18, wherein the antibody r ale comprises 1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 86 all three of the CDRS of the VH region having the amino acid sequence shown as Sequence ID-No: 27 and/or all three of the CDRS of the VL region having the amino acid sequence shown as Sequence ID No: 28.
18. The use or composition according to claim 17, wherein the antibody molecule comprises a variable region having the amino acid sequence of Sequence ID No: 27.
19. The use or composition according to claim 17 or claim 18, wherein the antibody molecule comprises a variable region having the amino acid sequence of Sequence ID No: 28. Fusion Antibodies Limited eys for the Applicant Aliens Arthur Robinson Patent & Trade Marks Attorneys
Applications Claiming Priority (6)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
GB0620147A GB0620147D0 (en) | 2006-10-11 | 2006-10-11 | Assay method |
GB0621848A GB0621848D0 (en) | 2006-11-02 | 2006-11-02 | Assay method |
GB0711226A GB0711226D0 (en) | 2007-06-09 | 2007-06-09 | Assay method |
GB0711228A GB0711228D0 (en) | 2007-06-09 | 2007-06-09 | Treatment method |
US99514307P | 2007-09-25 | 2007-09-25 | |
PCT/GB2007/050623 WO2008044068A2 (en) | 2006-10-11 | 2007-10-11 | Combination therapy |
Publications (1)
Publication Number | Publication Date |
---|---|
NZ576119A true NZ576119A (en) | 2012-05-25 |
Family
ID=46125075
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
NZ576119A NZ576119A (en) | 2006-10-11 | 2007-10-11 | Combination therapy involving egf inhibitors |
Country Status (1)
Country | Link |
---|---|
NZ (1) | NZ576119A (en) |
-
2007
- 2007-10-11 NZ NZ576119A patent/NZ576119A/en not_active IP Right Cessation
Similar Documents
Publication | Publication Date | Title |
---|---|---|
AU2007306139B2 (en) | Combination therapy | |
EP2281001B1 (en) | Anti-areg/hb-egf antibodies and treatment | |
KR102129636B1 (en) | Methods of treating cancer using pd-l1 axis binding antagonists and vegf antagonists | |
EP3038647B1 (en) | Combination therapy for the treatment of glioblastoma | |
AU2014364520A1 (en) | Methods and assays relating to circulating tumor cells | |
BRPI0712077A2 (en) | prolongation of survival of cancer patients with elevated levels of egf or tgf-alpha | |
JP2015514710A (en) | Diagnosis and treatment of HER3 inhibitors | |
US20220242963A1 (en) | B-cell activating cd73 antibodies | |
KR20140138956A (en) | Combination therapies including inhibitors of the extracellular domain of e-cadherin | |
NZ576119A (en) | Combination therapy involving egf inhibitors | |
ES2502217T3 (en) | NLRR-1 antagonists and their uses | |
US20230416346A1 (en) | C-terminal sparc fragments for treating cancer | |
WO2013148159A1 (en) | Therapeutic methods for peritoneal carcinomatosis | |
WO2013148202A1 (en) | Novel compositions and methods for preventing or treating cancer metastasis |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
PSEA | Patent sealed | ||
RENW | Renewal (renewal fees accepted) | ||
RENW | Renewal (renewal fees accepted) |
Free format text: PATENT RENEWED FOR 1 YEAR UNTIL 11 OCT 2015 BY FISHER ADAMS KELLY Effective date: 20141009 |
|
RENW | Renewal (renewal fees accepted) |
Free format text: PATENT RENEWED FOR 1 YEAR UNTIL 11 OCT 2016 BY RENEWALS TEAM Effective date: 20151008 |
|
LAPS | Patent lapsed |