AU2007279261A1 - Treatment of tumors in pediatric patients with epidermal growth factor receptor antagonists - Google Patents
Treatment of tumors in pediatric patients with epidermal growth factor receptor antagonists Download PDFInfo
- Publication number
- AU2007279261A1 AU2007279261A1 AU2007279261A AU2007279261A AU2007279261A1 AU 2007279261 A1 AU2007279261 A1 AU 2007279261A1 AU 2007279261 A AU2007279261 A AU 2007279261A AU 2007279261 A AU2007279261 A AU 2007279261A AU 2007279261 A1 AU2007279261 A1 AU 2007279261A1
- Authority
- AU
- Australia
- Prior art keywords
- egfr
- antibody
- antibodies
- tumors
- tumor
- Prior art date
- Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
- Abandoned
Links
- 206010028980 Neoplasm Diseases 0.000 title claims description 48
- 102000001301 EGF receptor Human genes 0.000 title description 78
- 108060006698 EGF receptor Proteins 0.000 title description 78
- 238000011282 treatment Methods 0.000 title description 24
- 229940044551 receptor antagonist Drugs 0.000 title description 2
- 239000002464 receptor antagonist Substances 0.000 title description 2
- 238000000034 method Methods 0.000 claims description 31
- 239000002246 antineoplastic agent Substances 0.000 claims description 22
- 239000012634 fragment Substances 0.000 claims description 21
- 239000003446 ligand Substances 0.000 claims description 15
- 229940127089 cytotoxic agent Drugs 0.000 claims description 13
- 239000005557 antagonist Substances 0.000 claims description 12
- 229960005395 cetuximab Drugs 0.000 claims description 12
- 229960004768 irinotecan Drugs 0.000 claims description 11
- UWKQSNNFCGGAFS-XIFFEERXSA-N irinotecan Chemical compound C1=C2C(CC)=C3CN(C(C4=C([C@@](C(=O)OC4)(O)CC)C=4)=O)C=4C3=NC2=CC=C1OC(=O)N(CC1)CCC1N1CCCCC1 UWKQSNNFCGGAFS-XIFFEERXSA-N 0.000 claims description 11
- 206010029260 Neuroblastoma Diseases 0.000 claims description 6
- 230000012010 growth Effects 0.000 claims description 6
- 230000002401 inhibitory effect Effects 0.000 claims description 6
- VBEQCZHXXJYVRD-GACYYNSASA-N uroanthelone Chemical compound C([C@@H](C(=O)N[C@H](C(=O)N[C@@H](CS)C(=O)N[C@@H](CC(N)=O)C(=O)N[C@@H](CS)C(=O)N[C@H](C(=O)N[C@@H]([C@@H](C)CC)C(=O)NCC(=O)N[C@@H](CC=1C=CC(O)=CC=1)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CS)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H]([C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CC=1C2=CC=CC=C2NC=1)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](CCCNC(N)=N)C(O)=O)C(C)C)[C@@H](C)O)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CO)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H](NC(=O)[C@H](CC=1NC=NC=1)NC(=O)[C@H](CCSC)NC(=O)[C@H](CS)NC(=O)[C@@H](NC(=O)CNC(=O)CNC(=O)[C@H](CC(N)=O)NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CS)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)CNC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@H](CO)NC(=O)[C@H]1N(CCC1)C(=O)[C@H](CS)NC(=O)CNC(=O)[C@H]1N(CCC1)C(=O)[C@H](CC=1C=CC(O)=CC=1)NC(=O)[C@H](CO)NC(=O)[C@@H](N)CC(N)=O)C(C)C)[C@@H](C)CC)C1=CC=C(O)C=C1 VBEQCZHXXJYVRD-GACYYNSASA-N 0.000 claims description 6
- 101800003838 Epidermal growth factor Proteins 0.000 claims description 5
- 206010018338 Glioma Diseases 0.000 claims description 5
- 229940116977 epidermal growth factor Drugs 0.000 claims description 5
- 230000005855 radiation Effects 0.000 claims description 5
- 230000004913 activation Effects 0.000 claims description 4
- 102000009024 Epidermal Growth Factor Human genes 0.000 claims 1
- 208000032612 Glial tumor Diseases 0.000 claims 1
- 210000004027 cell Anatomy 0.000 description 25
- 229940122558 EGFR antagonist Drugs 0.000 description 16
- 102000005962 receptors Human genes 0.000 description 13
- 108020003175 receptors Proteins 0.000 description 13
- 101001012157 Homo sapiens Receptor tyrosine-protein kinase erbB-2 Proteins 0.000 description 10
- 229940034982 antineoplastic agent Drugs 0.000 description 10
- 230000000694 effects Effects 0.000 description 10
- 108090000623 proteins and genes Proteins 0.000 description 10
- 102100030086 Receptor tyrosine-protein kinase erbB-2 Human genes 0.000 description 9
- 230000026731 phosphorylation Effects 0.000 description 9
- 238000006366 phosphorylation reaction Methods 0.000 description 9
- 150000003384 small molecules Chemical class 0.000 description 9
- 201000010099 disease Diseases 0.000 description 8
- 208000037265 diseases, disorders, signs and symptoms Diseases 0.000 description 8
- 102000004169 proteins and genes Human genes 0.000 description 8
- 102000004022 Protein-Tyrosine Kinases Human genes 0.000 description 7
- 108090000412 Protein-Tyrosine Kinases Proteins 0.000 description 7
- 239000000427 antigen Substances 0.000 description 7
- 102000036639 antigens Human genes 0.000 description 7
- 108091007433 antigens Proteins 0.000 description 7
- 201000011510 cancer Diseases 0.000 description 7
- 239000003112 inhibitor Substances 0.000 description 7
- 238000006386 neutralization reaction Methods 0.000 description 7
- 241001529936 Murinae Species 0.000 description 6
- 239000003795 chemical substances by application Substances 0.000 description 6
- 229940121647 egfr inhibitor Drugs 0.000 description 6
- 230000005764 inhibitory process Effects 0.000 description 6
- 108090000765 processed proteins & peptides Proteins 0.000 description 6
- 230000019491 signal transduction Effects 0.000 description 6
- AOJJSUZBOXZQNB-TZSSRYMLSA-N Doxorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 AOJJSUZBOXZQNB-TZSSRYMLSA-N 0.000 description 5
- 108060003951 Immunoglobulin Proteins 0.000 description 5
- GTTBEUCJPZQMDZ-UHFFFAOYSA-N erlotinib hydrochloride Chemical group [H+].[Cl-].C=12C=C(OCCOC)C(OCCOC)=CC2=NC=NC=1NC1=CC=CC(C#C)=C1 GTTBEUCJPZQMDZ-UHFFFAOYSA-N 0.000 description 5
- 230000014509 gene expression Effects 0.000 description 5
- 102000018358 immunoglobulin Human genes 0.000 description 5
- -1 morpholin-4-yl-propoxy Chemical group 0.000 description 5
- 230000004044 response Effects 0.000 description 5
- NMWKYTGJWUAZPZ-WWHBDHEGSA-N (4S)-4-[[(4R,7S,10S,16S,19S,25S,28S,31R)-31-[[(2S)-2-[[(1R,6R,9S,12S,18S,21S,24S,27S,30S,33S,36S,39S,42R,47R,53S,56S,59S,62S,65S,68S,71S,76S,79S,85S)-47-[[(2S)-2-[[(2S)-4-amino-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-[[(2S)-2-amino-3-methylbutanoyl]amino]-3-methylbutanoyl]amino]-3-hydroxypropanoyl]amino]-3-(1H-imidazol-4-yl)propanoyl]amino]-3-phenylpropanoyl]amino]-4-oxobutanoyl]amino]-3-carboxypropanoyl]amino]-18-(4-aminobutyl)-27,68-bis(3-amino-3-oxopropyl)-36,71,76-tribenzyl-39-(3-carbamimidamidopropyl)-24-(2-carboxyethyl)-21,56-bis(carboxymethyl)-65,85-bis[(1R)-1-hydroxyethyl]-59-(hydroxymethyl)-62,79-bis(1H-imidazol-4-ylmethyl)-9-methyl-33-(2-methylpropyl)-8,11,17,20,23,26,29,32,35,38,41,48,54,57,60,63,66,69,72,74,77,80,83,86-tetracosaoxo-30-propan-2-yl-3,4,44,45-tetrathia-7,10,16,19,22,25,28,31,34,37,40,49,55,58,61,64,67,70,73,75,78,81,84,87-tetracosazatetracyclo[40.31.14.012,16.049,53]heptaoctacontane-6-carbonyl]amino]-3-methylbutanoyl]amino]-7-(3-carbamimidamidopropyl)-25-(hydroxymethyl)-19-[(4-hydroxyphenyl)methyl]-28-(1H-imidazol-4-ylmethyl)-10-methyl-6,9,12,15,18,21,24,27,30-nonaoxo-16-propan-2-yl-1,2-dithia-5,8,11,14,17,20,23,26,29-nonazacyclodotriacontane-4-carbonyl]amino]-5-[[(2S)-1-[[(2S)-1-[[(2S)-3-carboxy-1-[[(2S)-1-[[(2S)-1-[[(1S)-1-carboxyethyl]amino]-4-methyl-1-oxopentan-2-yl]amino]-4-methyl-1-oxopentan-2-yl]amino]-1-oxopropan-2-yl]amino]-1-oxopropan-2-yl]amino]-3-(1H-imidazol-4-yl)-1-oxopropan-2-yl]amino]-5-oxopentanoic acid Chemical compound CC(C)C[C@H](NC(=O)[C@H](CC(C)C)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](C)NC(=O)[C@H](Cc1c[nH]cn1)NC(=O)[C@H](CCC(O)=O)NC(=O)[C@@H]1CSSC[C@H](NC(=O)[C@@H](NC(=O)[C@@H]2CSSC[C@@H]3NC(=O)[C@H](Cc4ccccc4)NC(=O)[C@H](CCC(N)=O)NC(=O)[C@@H](NC(=O)[C@H](Cc4c[nH]cn4)NC(=O)[C@H](CO)NC(=O)[C@H](CC(O)=O)NC(=O)[C@@H]4CCCN4C(=O)[C@H](CSSC[C@H](NC(=O)[C@@H](NC(=O)CNC(=O)[C@H](Cc4c[nH]cn4)NC(=O)[C@H](Cc4ccccc4)NC3=O)[C@@H](C)O)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N[C@@H](Cc3ccccc3)C(=O)N[C@@H](CC(C)C)C(=O)N[C@@H](C(C)C)C(=O)N[C@@H](CCC(N)=O)C(=O)N[C@@H](CCC(O)=O)C(=O)N[C@@H](CC(O)=O)C(=O)N[C@@H](CCCCN)C(=O)N3CCC[C@H]3C(=O)N[C@@H](C)C(=O)N2)NC(=O)[C@H](CC(O)=O)NC(=O)[C@H](CC(N)=O)NC(=O)[C@H](Cc2ccccc2)NC(=O)[C@H](Cc2c[nH]cn2)NC(=O)[C@H](CO)NC(=O)[C@@H](NC(=O)[C@@H](N)C(C)C)C(C)C)[C@@H](C)O)C(C)C)C(=O)N[C@@H](Cc2c[nH]cn2)C(=O)N[C@@H](CO)C(=O)NCC(=O)N[C@@H](Cc2ccc(O)cc2)C(=O)N[C@@H](C(C)C)C(=O)NCC(=O)N[C@@H](C)C(=O)N[C@@H](CCCNC(N)=N)C(=O)N1)C(=O)N[C@@H](C)C(O)=O NMWKYTGJWUAZPZ-WWHBDHEGSA-N 0.000 description 4
- FDKXTQMXEQVLRF-ZHACJKMWSA-N (E)-dacarbazine Chemical compound CN(C)\N=N\c1[nH]cnc1C(N)=O FDKXTQMXEQVLRF-ZHACJKMWSA-N 0.000 description 4
- 102400001368 Epidermal growth factor Human genes 0.000 description 4
- 108010021625 Immunoglobulin Fragments Proteins 0.000 description 4
- 102000008394 Immunoglobulin Fragments Human genes 0.000 description 4
- 241000699666 Mus <mouse, genus> Species 0.000 description 4
- 241000699670 Mus sp. Species 0.000 description 4
- 102000006747 Transforming Growth Factor alpha Human genes 0.000 description 4
- 101800004564 Transforming growth factor alpha Proteins 0.000 description 4
- 238000003556 assay Methods 0.000 description 4
- 208000011654 childhood malignant neoplasm Diseases 0.000 description 4
- 229940072221 immunoglobulins Drugs 0.000 description 4
- 229960001972 panitumumab Drugs 0.000 description 4
- 239000000126 substance Substances 0.000 description 4
- 238000002560 therapeutic procedure Methods 0.000 description 4
- 210000004881 tumor cell Anatomy 0.000 description 4
- 230000004614 tumor growth Effects 0.000 description 4
- STQGQHZAVUOBTE-UHFFFAOYSA-N 7-Cyan-hept-2t-en-4,6-diinsaeure Natural products C1=2C(O)=C3C(=O)C=4C(OC)=CC=CC=4C(=O)C3=C(O)C=2CC(O)(C(C)=O)CC1OC1CC(N)C(O)C(C)O1 STQGQHZAVUOBTE-UHFFFAOYSA-N 0.000 description 3
- 206010012735 Diarrhoea Diseases 0.000 description 3
- 125000000539 amino acid group Chemical group 0.000 description 3
- 150000001413 amino acids Chemical class 0.000 description 3
- 230000000340 anti-metabolite Effects 0.000 description 3
- 229940100197 antimetabolite Drugs 0.000 description 3
- 239000002256 antimetabolite Substances 0.000 description 3
- 210000004556 brain Anatomy 0.000 description 3
- 150000001875 compounds Chemical class 0.000 description 3
- 229960003901 dacarbazine Drugs 0.000 description 3
- STQGQHZAVUOBTE-VGBVRHCVSA-N daunorubicin Chemical compound O([C@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(C)=O)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 STQGQHZAVUOBTE-VGBVRHCVSA-N 0.000 description 3
- 238000001514 detection method Methods 0.000 description 3
- 231100000371 dose-limiting toxicity Toxicity 0.000 description 3
- 230000006870 function Effects 0.000 description 3
- 210000003128 head Anatomy 0.000 description 3
- 230000001404 mediated effect Effects 0.000 description 3
- 230000002297 mitogenic effect Effects 0.000 description 3
- 230000009467 reduction Effects 0.000 description 3
- 201000009410 rhabdomyosarcoma Diseases 0.000 description 3
- 208000024891 symptom Diseases 0.000 description 3
- 231100000419 toxicity Toxicity 0.000 description 3
- 230000001988 toxicity Effects 0.000 description 3
- KDOPAZIWBAHVJB-UHFFFAOYSA-N 5h-pyrrolo[3,2-d]pyrimidine Chemical compound C1=NC=C2NC=CC2=N1 KDOPAZIWBAHVJB-UHFFFAOYSA-N 0.000 description 2
- FJHBVJOVLFPMQE-QFIPXVFZSA-N 7-Ethyl-10-Hydroxy-Camptothecin Chemical compound C1=C(O)C=C2C(CC)=C(CN3C(C4=C([C@@](C(=O)OC4)(O)CC)C=C33)=O)C3=NC2=C1 FJHBVJOVLFPMQE-QFIPXVFZSA-N 0.000 description 2
- 206010003571 Astrocytoma Diseases 0.000 description 2
- IJGRMHOSHXDMSA-UHFFFAOYSA-N Atomic nitrogen Chemical compound N#N IJGRMHOSHXDMSA-UHFFFAOYSA-N 0.000 description 2
- 208000003174 Brain Neoplasms Diseases 0.000 description 2
- DLGOEMSEDOSKAD-UHFFFAOYSA-N Carmustine Chemical compound ClCCNC(=O)N(N=O)CCCl DLGOEMSEDOSKAD-UHFFFAOYSA-N 0.000 description 2
- CMSMOCZEIVJLDB-UHFFFAOYSA-N Cyclophosphamide Chemical compound ClCCN(CCCl)P1(=O)NCCCO1 CMSMOCZEIVJLDB-UHFFFAOYSA-N 0.000 description 2
- 206010014967 Ependymoma Diseases 0.000 description 2
- 108010054477 Immunoglobulin Fab Fragments Proteins 0.000 description 2
- 102000001706 Immunoglobulin Fab Fragments Human genes 0.000 description 2
- XEEYBQQBJWHFJM-UHFFFAOYSA-N Iron Chemical compound [Fe] XEEYBQQBJWHFJM-UHFFFAOYSA-N 0.000 description 2
- 239000005551 L01XE03 - Erlotinib Substances 0.000 description 2
- GQYIWUVLTXOXAJ-UHFFFAOYSA-N Lomustine Chemical compound ClCCN(N=O)C(=O)NC1CCCCC1 GQYIWUVLTXOXAJ-UHFFFAOYSA-N 0.000 description 2
- 102400001093 PAK-2p27 Human genes 0.000 description 2
- 108091000080 Phosphotransferase Proteins 0.000 description 2
- REFJWTPEDVJJIY-UHFFFAOYSA-N Quercetin Chemical compound C=1C(O)=CC(O)=C(C(C=2O)=O)C=1OC=2C1=CC=C(O)C(O)=C1 REFJWTPEDVJJIY-UHFFFAOYSA-N 0.000 description 2
- 238000010240 RT-PCR analysis Methods 0.000 description 2
- 101710100969 Receptor tyrosine-protein kinase erbB-3 Proteins 0.000 description 2
- 206010039491 Sarcoma Diseases 0.000 description 2
- 108010003723 Single-Domain Antibodies Proteins 0.000 description 2
- NKANXQFJJICGDU-QPLCGJKRSA-N Tamoxifen Chemical compound C=1C=CC=CC=1C(/CC)=C(C=1C=CC(OCCN(C)C)=CC=1)/C1=CC=CC=C1 NKANXQFJJICGDU-QPLCGJKRSA-N 0.000 description 2
- RJURFGZVJUQBHK-UHFFFAOYSA-N actinomycin D Natural products CC1OC(=O)C(C(C)C)N(C)C(=O)CN(C)C(=O)C2CCCN2C(=O)C(C(C)C)NC(=O)C1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)NC4C(=O)NC(C(N5CCCC5C(=O)N(C)CC(=O)N(C)C(C(C)C)C(=O)OC4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-UHFFFAOYSA-N 0.000 description 2
- 229940100198 alkylating agent Drugs 0.000 description 2
- 239000002168 alkylating agent Substances 0.000 description 2
- JKOQGQFVAUAYPM-UHFFFAOYSA-N amifostine Chemical compound NCCCNCCSP(O)(O)=O JKOQGQFVAUAYPM-UHFFFAOYSA-N 0.000 description 2
- 230000000259 anti-tumor effect Effects 0.000 description 2
- 230000006907 apoptotic process Effects 0.000 description 2
- 230000035578 autophosphorylation Effects 0.000 description 2
- 230000000903 blocking effect Effects 0.000 description 2
- 210000000133 brain stem Anatomy 0.000 description 2
- 230000001413 cellular effect Effects 0.000 description 2
- 238000002512 chemotherapy Methods 0.000 description 2
- 229960004397 cyclophosphamide Drugs 0.000 description 2
- 229960000975 daunorubicin Drugs 0.000 description 2
- 238000006731 degradation reaction Methods 0.000 description 2
- 238000011161 development Methods 0.000 description 2
- 230000018109 developmental process Effects 0.000 description 2
- 229960004679 doxorubicin Drugs 0.000 description 2
- XGALLCVXEZPNRQ-UHFFFAOYSA-N gefitinib Chemical group C=12C=C(OCCCN3CCOCC3)C(OC)=CC2=NC=NC=1NC1=CC=C(F)C(Cl)=C1 XGALLCVXEZPNRQ-UHFFFAOYSA-N 0.000 description 2
- SDUQYLNIPVEERB-QPPQHZFASA-N gemcitabine Chemical compound O=C1N=C(N)C=CN1[C@H]1C(F)(F)[C@H](O)[C@@H](CO)O1 SDUQYLNIPVEERB-QPPQHZFASA-N 0.000 description 2
- 208000006359 hepatoblastoma Diseases 0.000 description 2
- 229940022353 herceptin Drugs 0.000 description 2
- 210000004408 hybridoma Anatomy 0.000 description 2
- 238000003364 immunohistochemistry Methods 0.000 description 2
- 238000000338 in vitro Methods 0.000 description 2
- 238000001727 in vivo Methods 0.000 description 2
- 238000001802 infusion Methods 0.000 description 2
- 230000003834 intracellular effect Effects 0.000 description 2
- 210000004072 lung Anatomy 0.000 description 2
- 229960004961 mechlorethamine Drugs 0.000 description 2
- HAWPXGHAZFHHAD-UHFFFAOYSA-N mechlorethamine Chemical compound ClCCN(C)CCCl HAWPXGHAZFHHAD-UHFFFAOYSA-N 0.000 description 2
- 229960001924 melphalan Drugs 0.000 description 2
- SGDBTWWWUNNDEQ-LBPRGKRZSA-N melphalan Chemical compound OC(=O)[C@@H](N)CC1=CC=C(N(CCCl)CCCl)C=C1 SGDBTWWWUNNDEQ-LBPRGKRZSA-N 0.000 description 2
- 239000012528 membrane Substances 0.000 description 2
- CFCUWKMKBJTWLW-BKHRDMLASA-N mithramycin Chemical compound O([C@@H]1C[C@@H](O[C@H](C)[C@H]1O)OC=1C=C2C=C3C[C@H]([C@@H](C(=O)C3=C(O)C2=C(O)C=1C)O[C@@H]1O[C@H](C)[C@@H](O)[C@H](O[C@@H]2O[C@H](C)[C@H](O)[C@H](O[C@@H]3O[C@H](C)[C@@H](O)[C@@](C)(O)C3)C2)C1)[C@H](OC)C(=O)[C@@H](O)[C@@H](C)O)[C@H]1C[C@@H](O)[C@H](O)[C@@H](C)O1 CFCUWKMKBJTWLW-BKHRDMLASA-N 0.000 description 2
- 230000004048 modification Effects 0.000 description 2
- 238000012986 modification Methods 0.000 description 2
- 210000003739 neck Anatomy 0.000 description 2
- 201000008968 osteosarcoma Diseases 0.000 description 2
- 230000002611 ovarian Effects 0.000 description 2
- 210000001672 ovary Anatomy 0.000 description 2
- 238000002823 phage display Methods 0.000 description 2
- 102000020233 phosphotransferase Human genes 0.000 description 2
- 229960003171 plicamycin Drugs 0.000 description 2
- 102000004196 processed proteins & peptides Human genes 0.000 description 2
- 238000001959 radiotherapy Methods 0.000 description 2
- 230000033300 receptor internalization Effects 0.000 description 2
- 229960001052 streptozocin Drugs 0.000 description 2
- ZSJLQEPLLKMAKR-GKHCUFPYSA-N streptozocin Chemical compound O=NN(C)C(=O)N[C@H]1[C@@H](O)O[C@H](CO)[C@@H](O)[C@@H]1O ZSJLQEPLLKMAKR-GKHCUFPYSA-N 0.000 description 2
- 238000007920 subcutaneous administration Methods 0.000 description 2
- 239000000758 substrate Substances 0.000 description 2
- 230000004083 survival effect Effects 0.000 description 2
- 229940120982 tarceva Drugs 0.000 description 2
- RCINICONZNJXQF-MZXODVADSA-N taxol Chemical compound O([C@@H]1[C@@]2(C[C@@H](C(C)=C(C2(C)C)[C@H](C([C@]2(C)[C@@H](O)C[C@H]3OC[C@]3([C@H]21)OC(C)=O)=O)OC(=O)C)OC(=O)[C@H](O)[C@@H](NC(=O)C=1C=CC=CC=1)C=1C=CC=CC=1)O)C(=O)C1=CC=CC=C1 RCINICONZNJXQF-MZXODVADSA-N 0.000 description 2
- 229960000575 trastuzumab Drugs 0.000 description 2
- 229940121358 tyrosine kinase inhibitor Drugs 0.000 description 2
- 239000005483 tyrosine kinase inhibitor Substances 0.000 description 2
- 150000004917 tyrosine kinase inhibitor derivatives Chemical class 0.000 description 2
- MWWSFMDVAYGXBV-MYPASOLCSA-N (7r,9s)-7-[(2r,4s,5s,6s)-4-amino-5-hydroxy-6-methyloxan-2-yl]oxy-6,9,11-trihydroxy-9-(2-hydroxyacetyl)-4-methoxy-8,10-dihydro-7h-tetracene-5,12-dione;hydrochloride Chemical compound Cl.O([C@@H]1C[C@@](O)(CC=2C(O)=C3C(=O)C=4C=CC=C(C=4C(=O)C3=C(O)C=21)OC)C(=O)CO)[C@H]1C[C@H](N)[C@H](O)[C@H](C)O1 MWWSFMDVAYGXBV-MYPASOLCSA-N 0.000 description 1
- FPVKHBSQESCIEP-UHFFFAOYSA-N (8S)-3-(2-deoxy-beta-D-erythro-pentofuranosyl)-3,6,7,8-tetrahydroimidazo[4,5-d][1,3]diazepin-8-ol Natural products C1C(O)C(CO)OC1N1C(NC=NCC2O)=C2N=C1 FPVKHBSQESCIEP-UHFFFAOYSA-N 0.000 description 1
- VSNHCAURESNICA-NJFSPNSNSA-N 1-oxidanylurea Chemical compound N[14C](=O)NO VSNHCAURESNICA-NJFSPNSNSA-N 0.000 description 1
- QXLQZLBNPTZMRK-UHFFFAOYSA-N 2-[(dimethylamino)methyl]-1-(2,4-dimethylphenyl)prop-2-en-1-one Chemical compound CN(C)CC(=C)C(=O)C1=CC=C(C)C=C1C QXLQZLBNPTZMRK-UHFFFAOYSA-N 0.000 description 1
- RONQPWQYDRPRGG-UHFFFAOYSA-N 5,6-bis(4-fluoroanilino)isoindole-1,3-dione Chemical compound C1=CC(F)=CC=C1NC(C(=C1)NC=2C=CC(F)=CC=2)=CC2=C1C(=O)NC2=O RONQPWQYDRPRGG-UHFFFAOYSA-N 0.000 description 1
- IDPUKCWIGUEADI-UHFFFAOYSA-N 5-[bis(2-chloroethyl)amino]uracil Chemical compound ClCCN(CCCl)C1=CNC(=O)NC1=O IDPUKCWIGUEADI-UHFFFAOYSA-N 0.000 description 1
- WYWHKKSPHMUBEB-UHFFFAOYSA-N 6-Mercaptoguanine Natural products N1C(N)=NC(=S)C2=C1N=CN2 WYWHKKSPHMUBEB-UHFFFAOYSA-N 0.000 description 1
- 208000024893 Acute lymphoblastic leukemia Diseases 0.000 description 1
- 208000014697 Acute lymphocytic leukaemia Diseases 0.000 description 1
- 208000003200 Adenoma Diseases 0.000 description 1
- GUBGYTABKSRVRQ-XLOQQCSPSA-N Alpha-Lactose Chemical compound O[C@@H]1[C@@H](O)[C@@H](O)[C@@H](CO)O[C@H]1O[C@@H]1[C@@H](CO)O[C@H](O)[C@H](O)[C@H]1O GUBGYTABKSRVRQ-XLOQQCSPSA-N 0.000 description 1
- 102100038778 Amphiregulin Human genes 0.000 description 1
- 108010033760 Amphiregulin Proteins 0.000 description 1
- 108010024976 Asparaginase Proteins 0.000 description 1
- 102000015790 Asparaginase Human genes 0.000 description 1
- 101800001382 Betacellulin Proteins 0.000 description 1
- 108010006654 Bleomycin Proteins 0.000 description 1
- 206010055113 Breast cancer metastatic Diseases 0.000 description 1
- COVZYZSDYWQREU-UHFFFAOYSA-N Busulfan Chemical compound CS(=O)(=O)OCCCCOS(C)(=O)=O COVZYZSDYWQREU-UHFFFAOYSA-N 0.000 description 1
- OYPRJOBELJOOCE-UHFFFAOYSA-N Calcium Chemical compound [Ca] OYPRJOBELJOOCE-UHFFFAOYSA-N 0.000 description 1
- KLWPJMFMVPTNCC-UHFFFAOYSA-N Camptothecin Natural products CCC1(O)C(=O)OCC2=C1C=C3C4Nc5ccccc5C=C4CN3C2=O KLWPJMFMVPTNCC-UHFFFAOYSA-N 0.000 description 1
- OKTJSMMVPCPJKN-UHFFFAOYSA-N Carbon Chemical compound [C] OKTJSMMVPCPJKN-UHFFFAOYSA-N 0.000 description 1
- 208000005623 Carcinogenesis Diseases 0.000 description 1
- 201000009030 Carcinoma Diseases 0.000 description 1
- 102000052052 Casein Kinase II Human genes 0.000 description 1
- 108010010919 Casein Kinase II Proteins 0.000 description 1
- JWBOIMRXGHLCPP-UHFFFAOYSA-N Chloditan Chemical compound C=1C=CC=C(Cl)C=1C(C(Cl)Cl)C1=CC=C(Cl)C=C1 JWBOIMRXGHLCPP-UHFFFAOYSA-N 0.000 description 1
- PTOAARAWEBMLNO-KVQBGUIXSA-N Cladribine Chemical compound C1=NC=2C(N)=NC(Cl)=NC=2N1[C@H]1C[C@H](O)[C@@H](CO)O1 PTOAARAWEBMLNO-KVQBGUIXSA-N 0.000 description 1
- 208000001333 Colorectal Neoplasms Diseases 0.000 description 1
- 108010047041 Complementarity Determining Regions Proteins 0.000 description 1
- 108091035707 Consensus sequence Proteins 0.000 description 1
- 241000699802 Cricetulus griseus Species 0.000 description 1
- XZMCDFZZKTWFGF-UHFFFAOYSA-N Cyanamide Chemical compound NC#N XZMCDFZZKTWFGF-UHFFFAOYSA-N 0.000 description 1
- UHDGCWIWMRVCDJ-CCXZUQQUSA-N Cytarabine Chemical compound O=C1N=C(N)C=CN1[C@H]1[C@@H](O)[C@H](O)[C@@H](CO)O1 UHDGCWIWMRVCDJ-CCXZUQQUSA-N 0.000 description 1
- 108020004414 DNA Proteins 0.000 description 1
- 230000004544 DNA amplification Effects 0.000 description 1
- 108010092160 Dactinomycin Proteins 0.000 description 1
- 238000002965 ELISA Methods 0.000 description 1
- 238000012286 ELISA Assay Methods 0.000 description 1
- 102000056372 ErbB-3 Receptor Human genes 0.000 description 1
- 208000006168 Ewing Sarcoma Diseases 0.000 description 1
- 208000010201 Exanthema Diseases 0.000 description 1
- 238000012413 Fluorescence activated cell sorting analysis Methods 0.000 description 1
- GHASVSINZRGABV-UHFFFAOYSA-N Fluorouracil Chemical compound FC1=CNC(=O)NC1=O GHASVSINZRGABV-UHFFFAOYSA-N 0.000 description 1
- WQZGKKKJIJFFOK-GASJEMHNSA-N Glucose Natural products OC[C@H]1OC(O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-GASJEMHNSA-N 0.000 description 1
- 102000009465 Growth Factor Receptors Human genes 0.000 description 1
- 108010009202 Growth Factor Receptors Proteins 0.000 description 1
- HTTJABKRGRZYRN-UHFFFAOYSA-N Heparin Chemical compound OC1C(NC(=O)C)C(O)OC(COS(O)(=O)=O)C1OC1C(OS(O)(=O)=O)C(O)C(OC2C(C(OS(O)(=O)=O)C(OC3C(C(O)C(O)C(O3)C(O)=O)OS(O)(=O)=O)C(CO)O2)NS(O)(=O)=O)C(C(O)=O)O1 HTTJABKRGRZYRN-UHFFFAOYSA-N 0.000 description 1
- MCAHMSDENAOJFZ-UHFFFAOYSA-N Herbimycin A Natural products N1C(=O)C(C)=CC=CC(OC)C(OC(N)=O)C(C)=CC(C)C(OC)C(OC)CC(C)C(OC)C2=CC(=O)C=C1C2=O MCAHMSDENAOJFZ-UHFFFAOYSA-N 0.000 description 1
- XDXDZDZNSLXDNA-TZNDIEGXSA-N Idarubicin Chemical compound C1[C@H](N)[C@H](O)[C@H](C)O[C@H]1O[C@@H]1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2C[C@@](O)(C(C)=O)C1 XDXDZDZNSLXDNA-TZNDIEGXSA-N 0.000 description 1
- XDXDZDZNSLXDNA-UHFFFAOYSA-N Idarubicin Natural products C1C(N)C(O)C(C)OC1OC1C2=C(O)C(C(=O)C3=CC=CC=C3C3=O)=C3C(O)=C2CC(O)(C(C)=O)C1 XDXDZDZNSLXDNA-UHFFFAOYSA-N 0.000 description 1
- DGAQECJNVWCQMB-PUAWFVPOSA-M Ilexoside XXIX Chemical compound C[C@@H]1CC[C@@]2(CC[C@@]3(C(=CC[C@H]4[C@]3(CC[C@@H]5[C@@]4(CC[C@@H](C5(C)C)OS(=O)(=O)[O-])C)C)[C@@H]2[C@]1(C)O)C)C(=O)O[C@H]6[C@@H]([C@H]([C@@H]([C@H](O6)CO)O)O)O.[Na+] DGAQECJNVWCQMB-PUAWFVPOSA-M 0.000 description 1
- 102000006992 Interferon-alpha Human genes 0.000 description 1
- 108010047761 Interferon-alpha Proteins 0.000 description 1
- 102000003996 Interferon-beta Human genes 0.000 description 1
- 108090000467 Interferon-beta Proteins 0.000 description 1
- 238000012449 Kunming mouse Methods 0.000 description 1
- FBOZXECLQNJBKD-ZDUSSCGKSA-N L-methotrexate Chemical compound C=1N=C2N=C(N)N=C(N)C2=NC=1CN(C)C1=CC=C(C(=O)N[C@@H](CCC(O)=O)C(O)=O)C=C1 FBOZXECLQNJBKD-ZDUSSCGKSA-N 0.000 description 1
- OUYCCCASQSFEME-QMMMGPOBSA-N L-tyrosine Chemical compound OC(=O)[C@@H](N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-QMMMGPOBSA-N 0.000 description 1
- 239000005411 L01XE02 - Gefitinib Substances 0.000 description 1
- UIARLYUEJFELEN-LROUJFHJSA-N LSM-1231 Chemical compound C12=C3N4C5=CC=CC=C5C3=C3C(=O)NCC3=C2C2=CC=CC=C2N1[C@]1(C)[C@](CO)(O)C[C@H]4O1 UIARLYUEJFELEN-LROUJFHJSA-N 0.000 description 1
- GUBGYTABKSRVRQ-QKKXKWKRSA-N Lactose Natural products OC[C@H]1O[C@@H](O[C@H]2[C@H](O)[C@@H](O)C(O)O[C@@H]2CO)[C@H](O)[C@@H](O)[C@H]1O GUBGYTABKSRVRQ-QKKXKWKRSA-N 0.000 description 1
- 108010000817 Leuprolide Proteins 0.000 description 1
- 206010025323 Lymphomas Diseases 0.000 description 1
- FYYHWMGAXLPEAU-UHFFFAOYSA-N Magnesium Chemical compound [Mg] FYYHWMGAXLPEAU-UHFFFAOYSA-N 0.000 description 1
- 241000124008 Mammalia Species 0.000 description 1
- PWHULOQIROXLJO-UHFFFAOYSA-N Manganese Chemical compound [Mn] PWHULOQIROXLJO-UHFFFAOYSA-N 0.000 description 1
- XOGTZOOQQBDUSI-UHFFFAOYSA-M Mesna Chemical compound [Na+].[O-]S(=O)(=O)CCS XOGTZOOQQBDUSI-UHFFFAOYSA-M 0.000 description 1
- 206010027476 Metastases Diseases 0.000 description 1
- 229930192392 Mitomycin Natural products 0.000 description 1
- 241000699660 Mus musculus Species 0.000 description 1
- NWIBSHFKIJFRCO-WUDYKRTCSA-N Mytomycin Chemical compound C1N2C(C(C(C)=C(N)C3=O)=O)=C3[C@@H](COC(N)=O)[C@@]2(OC)[C@@H]2[C@H]1N2 NWIBSHFKIJFRCO-WUDYKRTCSA-N 0.000 description 1
- 238000000636 Northern blotting Methods 0.000 description 1
- 201000010133 Oligodendroglioma Diseases 0.000 description 1
- 206010061535 Ovarian neoplasm Diseases 0.000 description 1
- 101150038994 PDGFRA gene Proteins 0.000 description 1
- 229910019142 PO4 Inorganic materials 0.000 description 1
- 229930012538 Paclitaxel Natural products 0.000 description 1
- 239000001888 Peptone Substances 0.000 description 1
- 108010080698 Peptones Proteins 0.000 description 1
- 206010035226 Plasma cell myeloma Diseases 0.000 description 1
- ZLMJMSJWJFRBEC-UHFFFAOYSA-N Potassium Chemical compound [K] ZLMJMSJWJFRBEC-UHFFFAOYSA-N 0.000 description 1
- 208000006664 Precursor Cell Lymphoblastic Leukemia-Lymphoma Diseases 0.000 description 1
- 102100029837 Probetacellulin Human genes 0.000 description 1
- ZVOLCUVKHLEPEV-UHFFFAOYSA-N Quercetagetin Natural products C1=C(O)C(O)=CC=C1C1=C(O)C(=O)C2=C(O)C(O)=C(O)C=C2O1 ZVOLCUVKHLEPEV-UHFFFAOYSA-N 0.000 description 1
- 102100029986 Receptor tyrosine-protein kinase erbB-3 Human genes 0.000 description 1
- 102100029981 Receptor tyrosine-protein kinase erbB-4 Human genes 0.000 description 1
- 101710100963 Receptor tyrosine-protein kinase erbB-4 Proteins 0.000 description 1
- 108020004511 Recombinant DNA Proteins 0.000 description 1
- HWTZYBCRDDUBJY-UHFFFAOYSA-N Rhynchosin Natural products C1=C(O)C(O)=CC=C1C1=C(O)C(=O)C2=CC(O)=C(O)C=C2O1 HWTZYBCRDDUBJY-UHFFFAOYSA-N 0.000 description 1
- 240000004808 Saccharomyces cerevisiae Species 0.000 description 1
- 208000021712 Soft tissue sarcoma Diseases 0.000 description 1
- 238000002105 Southern blotting Methods 0.000 description 1
- 241000011102 Thera Species 0.000 description 1
- FOCVUCIESVLUNU-UHFFFAOYSA-N Thiotepa Chemical compound C1CN1P(N1CC1)(=S)N1CC1 FOCVUCIESVLUNU-UHFFFAOYSA-N 0.000 description 1
- JXLYSJRDGCGARV-WWYNWVTFSA-N Vinblastine Natural products O=C(O[C@H]1[C@](O)(C(=O)OC)[C@@H]2N(C)c3c(cc(c(OC)c3)[C@]3(C(=O)OC)c4[nH]c5c(c4CCN4C[C@](O)(CC)C[C@H](C3)C4)cccc5)[C@@]32[C@H]2[C@@]1(CC)C=CCN2CC3)C JXLYSJRDGCGARV-WWYNWVTFSA-N 0.000 description 1
- 208000008383 Wilms tumor Diseases 0.000 description 1
- HCHKCACWOHOZIP-UHFFFAOYSA-N Zinc Chemical compound [Zn] HCHKCACWOHOZIP-UHFFFAOYSA-N 0.000 description 1
- RJURFGZVJUQBHK-IIXSONLDSA-N actinomycin D Chemical compound C[C@H]1OC(=O)[C@H](C(C)C)N(C)C(=O)CN(C)C(=O)[C@@H]2CCCN2C(=O)[C@@H](C(C)C)NC(=O)[C@H]1NC(=O)C1=C(N)C(=O)C(C)=C2OC(C(C)=CC=C3C(=O)N[C@@H]4C(=O)N[C@@H](C(N5CCC[C@H]5C(=O)N(C)CC(=O)N(C)[C@@H](C(C)C)C(=O)O[C@@H]4C)=O)C(C)C)=C3N=C21 RJURFGZVJUQBHK-IIXSONLDSA-N 0.000 description 1
- 208000009956 adenocarcinoma Diseases 0.000 description 1
- 201000001256 adenosarcoma Diseases 0.000 description 1
- 239000002671 adjuvant Substances 0.000 description 1
- 229960005310 aldesleukin Drugs 0.000 description 1
- 108700025316 aldesleukin Proteins 0.000 description 1
- 229960001097 amifostine Drugs 0.000 description 1
- 150000003863 ammonium salts Chemical class 0.000 description 1
- 230000033115 angiogenesis Effects 0.000 description 1
- 230000001093 anti-cancer Effects 0.000 description 1
- 230000000118 anti-neoplastic effect Effects 0.000 description 1
- 238000009175 antibody therapy Methods 0.000 description 1
- 229960003272 asparaginase Drugs 0.000 description 1
- DCXYFEDJOCDNAF-UHFFFAOYSA-M asparaginate Chemical compound [O-]C(=O)C(N)CC(N)=O DCXYFEDJOCDNAF-UHFFFAOYSA-M 0.000 description 1
- VSRXQHXAPYXROS-UHFFFAOYSA-N azanide;cyclobutane-1,1-dicarboxylic acid;platinum(2+) Chemical compound [NH2-].[NH2-].[Pt+2].OC(=O)C1(C(O)=O)CCC1 VSRXQHXAPYXROS-UHFFFAOYSA-N 0.000 description 1
- 230000008901 benefit Effects 0.000 description 1
- WQZGKKKJIJFFOK-VFUOTHLCSA-N beta-D-glucose Chemical compound OC[C@H]1O[C@@H](O)[C@H](O)[C@@H](O)[C@@H]1O WQZGKKKJIJFFOK-VFUOTHLCSA-N 0.000 description 1
- 102000023732 binding proteins Human genes 0.000 description 1
- 108091008324 binding proteins Proteins 0.000 description 1
- 201000000053 blastoma Diseases 0.000 description 1
- 229960001561 bleomycin Drugs 0.000 description 1
- OYVAGSVQBOHSSS-UAPAGMARSA-O bleomycin A2 Chemical compound N([C@H](C(=O)N[C@H](C)[C@@H](O)[C@H](C)C(=O)N[C@@H]([C@H](O)C)C(=O)NCCC=1SC=C(N=1)C=1SC=C(N=1)C(=O)NCCC[S+](C)C)[C@@H](O[C@H]1[C@H]([C@@H](O)[C@H](O)[C@H](CO)O1)O[C@@H]1[C@H]([C@@H](OC(N)=O)[C@H](O)[C@@H](CO)O1)O)C=1N=CNC=1)C(=O)C1=NC([C@H](CC(N)=O)NC[C@H](N)C(N)=O)=NC(N)=C1C OYVAGSVQBOHSSS-UAPAGMARSA-O 0.000 description 1
- 210000004369 blood Anatomy 0.000 description 1
- 239000008280 blood Substances 0.000 description 1
- 210000000988 bone and bone Anatomy 0.000 description 1
- 210000001185 bone marrow Anatomy 0.000 description 1
- 210000000481 breast Anatomy 0.000 description 1
- 229960002092 busulfan Drugs 0.000 description 1
- 239000011575 calcium Substances 0.000 description 1
- 229910052791 calcium Inorganic materials 0.000 description 1
- 229940127093 camptothecin Drugs 0.000 description 1
- VSJKWCGYPAHWDS-FQEVSTJZSA-N camptothecin Chemical compound C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-FQEVSTJZSA-N 0.000 description 1
- 230000036952 cancer formation Effects 0.000 description 1
- 150000001720 carbohydrates Chemical class 0.000 description 1
- 235000014633 carbohydrates Nutrition 0.000 description 1
- 229910052799 carbon Inorganic materials 0.000 description 1
- 150000004649 carbonic acid derivatives Chemical class 0.000 description 1
- 229960004562 carboplatin Drugs 0.000 description 1
- 125000003178 carboxy group Chemical group [H]OC(*)=O 0.000 description 1
- 231100000504 carcinogenesis Toxicity 0.000 description 1
- 229960005243 carmustine Drugs 0.000 description 1
- 230000015556 catabolic process Effects 0.000 description 1
- 238000000423 cell based assay Methods 0.000 description 1
- 230000025084 cell cycle arrest Effects 0.000 description 1
- 230000012292 cell migration Effects 0.000 description 1
- 230000004663 cell proliferation Effects 0.000 description 1
- 210000003679 cervix uteri Anatomy 0.000 description 1
- 238000012512 characterization method Methods 0.000 description 1
- 238000006243 chemical reaction Methods 0.000 description 1
- 229960004630 chlorambucil Drugs 0.000 description 1
- JCKYGMPEJWAADB-UHFFFAOYSA-N chlorambucil Chemical compound OC(=O)CCCC1=CC=C(N(CCCl)CCCl)C=C1 JCKYGMPEJWAADB-UHFFFAOYSA-N 0.000 description 1
- DQLATGHUWYMOKM-UHFFFAOYSA-L cisplatin Chemical compound N[Pt](N)(Cl)Cl DQLATGHUWYMOKM-UHFFFAOYSA-L 0.000 description 1
- 229960004316 cisplatin Drugs 0.000 description 1
- 229960002436 cladribine Drugs 0.000 description 1
- 210000001072 colon Anatomy 0.000 description 1
- 238000002648 combination therapy Methods 0.000 description 1
- 230000002860 competitive effect Effects 0.000 description 1
- 230000000295 complement effect Effects 0.000 description 1
- 210000004748 cultured cell Anatomy 0.000 description 1
- 125000004122 cyclic group Chemical group 0.000 description 1
- 229960000684 cytarabine Drugs 0.000 description 1
- 230000001086 cytosolic effect Effects 0.000 description 1
- 239000000824 cytostatic agent Substances 0.000 description 1
- 230000001085 cytostatic effect Effects 0.000 description 1
- 231100000433 cytotoxic Toxicity 0.000 description 1
- 230000001472 cytotoxic effect Effects 0.000 description 1
- 230000003013 cytotoxicity Effects 0.000 description 1
- 231100000135 cytotoxicity Toxicity 0.000 description 1
- 229960000640 dactinomycin Drugs 0.000 description 1
- 229940107841 daunoxome Drugs 0.000 description 1
- 230000032459 dedifferentiation Effects 0.000 description 1
- 230000002950 deficient Effects 0.000 description 1
- 239000007857 degradation product Substances 0.000 description 1
- 239000000539 dimer Substances 0.000 description 1
- 238000006471 dimerization reaction Methods 0.000 description 1
- AIUDWMLXCFRVDR-UHFFFAOYSA-N dimethyl 2-(3-ethyl-3-methylpentyl)propanedioate Chemical class CCC(C)(CC)CCC(C(=O)OC)C(=O)OC AIUDWMLXCFRVDR-UHFFFAOYSA-N 0.000 description 1
- VSJKWCGYPAHWDS-UHFFFAOYSA-N dl-camptothecin Natural products C1=CC=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)C5(O)CC)C4=NC2=C1 VSJKWCGYPAHWDS-UHFFFAOYSA-N 0.000 description 1
- 229940115080 doxil Drugs 0.000 description 1
- 239000003814 drug Substances 0.000 description 1
- 201000008184 embryoma Diseases 0.000 description 1
- 238000005516 engineering process Methods 0.000 description 1
- 108010087914 epidermal growth factor receptor VIII Proteins 0.000 description 1
- 229940082789 erbitux Drugs 0.000 description 1
- SIHZWGODIRRSRA-ONEGZZNKSA-N erbstatin Chemical compound OC1=CC=C(O)C(\C=C\NC=O)=C1 SIHZWGODIRRSRA-ONEGZZNKSA-N 0.000 description 1
- 229960001433 erlotinib Drugs 0.000 description 1
- 210000003238 esophagus Anatomy 0.000 description 1
- 229940098617 ethyol Drugs 0.000 description 1
- VJJPUSNTGOMMGY-MRVIYFEKSA-N etoposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@H](C)OC[C@H]4O3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 VJJPUSNTGOMMGY-MRVIYFEKSA-N 0.000 description 1
- 229960005420 etoposide Drugs 0.000 description 1
- 238000010228 ex vivo assay Methods 0.000 description 1
- 201000005884 exanthem Diseases 0.000 description 1
- ODKNJVUHOIMIIZ-RRKCRQDMSA-N floxuridine Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(=O)NC(=O)C(F)=C1 ODKNJVUHOIMIIZ-RRKCRQDMSA-N 0.000 description 1
- 229960000961 floxuridine Drugs 0.000 description 1
- 229960000390 fludarabine Drugs 0.000 description 1
- GIUYCYHIANZCFB-FJFJXFQQSA-N fludarabine phosphate Chemical compound C1=NC=2C(N)=NC(F)=NC=2N1[C@@H]1O[C@H](COP(O)(O)=O)[C@@H](O)[C@@H]1O GIUYCYHIANZCFB-FJFJXFQQSA-N 0.000 description 1
- 229960002949 fluorouracil Drugs 0.000 description 1
- 229960005277 gemcitabine Drugs 0.000 description 1
- 239000008103 glucose Substances 0.000 description 1
- 210000002216 heart Anatomy 0.000 description 1
- 229960002897 heparin Drugs 0.000 description 1
- 229920000669 heparin Polymers 0.000 description 1
- MCAHMSDENAOJFZ-BVXDHVRPSA-N herbimycin Chemical compound N1C(=O)\C(C)=C\C=C/[C@H](OC)[C@@H](OC(N)=O)\C(C)=C\[C@H](C)[C@@H](OC)[C@@H](OC)C[C@H](C)[C@@H](OC)C2=CC(=O)C=C1C2=O MCAHMSDENAOJFZ-BVXDHVRPSA-N 0.000 description 1
- 238000005734 heterodimerization reaction Methods 0.000 description 1
- 208000029824 high grade glioma Diseases 0.000 description 1
- 102000051957 human ERBB2 Human genes 0.000 description 1
- 229960000908 idarubicin Drugs 0.000 description 1
- 229960001101 ifosfamide Drugs 0.000 description 1
- HOMGKSMUEGBAAB-UHFFFAOYSA-N ifosfamide Chemical compound ClCCNP1(=O)OCCCN1CCCl HOMGKSMUEGBAAB-UHFFFAOYSA-N 0.000 description 1
- 230000028993 immune response Effects 0.000 description 1
- 238000002649 immunization Methods 0.000 description 1
- 230000003053 immunization Effects 0.000 description 1
- 238000007901 in situ hybridization Methods 0.000 description 1
- 230000002779 inactivation Effects 0.000 description 1
- 150000002475 indoles Chemical class 0.000 description 1
- 230000003993 interaction Effects 0.000 description 1
- 229960001388 interferon-beta Drugs 0.000 description 1
- 102000027411 intracellular receptors Human genes 0.000 description 1
- 108091008582 intracellular receptors Proteins 0.000 description 1
- 238000007918 intramuscular administration Methods 0.000 description 1
- 238000007912 intraperitoneal administration Methods 0.000 description 1
- 238000001990 intravenous administration Methods 0.000 description 1
- 230000009545 invasion Effects 0.000 description 1
- 229940084651 iressa Drugs 0.000 description 1
- 229910052742 iron Inorganic materials 0.000 description 1
- MWDZOUNAPSSOEL-UHFFFAOYSA-N kaempferol Natural products OC1=C(C(=O)c2cc(O)cc(O)c2O1)c3ccc(O)cc3 MWDZOUNAPSSOEL-UHFFFAOYSA-N 0.000 description 1
- 210000003734 kidney Anatomy 0.000 description 1
- 229940043355 kinase inhibitor Drugs 0.000 description 1
- 239000008101 lactose Substances 0.000 description 1
- 210000000867 larynx Anatomy 0.000 description 1
- 208000032839 leukemia Diseases 0.000 description 1
- GFIJNRVAKGFPGQ-LIJARHBVSA-N leuprolide Chemical compound CCNC(=O)[C@@H]1CCCN1C(=O)[C@H](CCCNC(N)=N)NC(=O)[C@H](CC(C)C)NC(=O)[C@@H](CC(C)C)NC(=O)[C@@H](NC(=O)[C@H](CO)NC(=O)[C@H](CC=1C2=CC=CC=C2NC=1)NC(=O)[C@H](CC=1N=CNC=1)NC(=O)[C@H]1NC(=O)CC1)CC1=CC=C(O)C=C1 GFIJNRVAKGFPGQ-LIJARHBVSA-N 0.000 description 1
- 229960004338 leuprorelin Drugs 0.000 description 1
- 230000000670 limiting effect Effects 0.000 description 1
- 239000007788 liquid Substances 0.000 description 1
- 210000004185 liver Anatomy 0.000 description 1
- 229960002247 lomustine Drugs 0.000 description 1
- 239000006166 lysate Substances 0.000 description 1
- 230000002934 lysing effect Effects 0.000 description 1
- 239000011777 magnesium Substances 0.000 description 1
- 229910052749 magnesium Inorganic materials 0.000 description 1
- 230000003211 malignant effect Effects 0.000 description 1
- 201000011614 malignant glioma Diseases 0.000 description 1
- 210000004962 mammalian cell Anatomy 0.000 description 1
- 229910052748 manganese Inorganic materials 0.000 description 1
- 239000011572 manganese Substances 0.000 description 1
- 238000004519 manufacturing process Methods 0.000 description 1
- 239000011159 matrix material Substances 0.000 description 1
- 229950008001 matuzumab Drugs 0.000 description 1
- 230000007246 mechanism Effects 0.000 description 1
- 229960001786 megestrol Drugs 0.000 description 1
- RQZAXGRLVPAYTJ-GQFGMJRRSA-N megestrol acetate Chemical compound C1=C(C)C2=CC(=O)CC[C@]2(C)[C@@H]2[C@@H]1[C@@H]1CC[C@@](C(C)=O)(OC(=O)C)[C@@]1(C)CC2 RQZAXGRLVPAYTJ-GQFGMJRRSA-N 0.000 description 1
- GLVAUDGFNGKCSF-UHFFFAOYSA-N mercaptopurine Chemical compound S=C1NC=NC2=C1NC=N2 GLVAUDGFNGKCSF-UHFFFAOYSA-N 0.000 description 1
- 229960001428 mercaptopurine Drugs 0.000 description 1
- 229960004635 mesna Drugs 0.000 description 1
- 230000009401 metastasis Effects 0.000 description 1
- 206010061289 metastatic neoplasm Diseases 0.000 description 1
- 229960000485 methotrexate Drugs 0.000 description 1
- 239000003226 mitogen Substances 0.000 description 1
- 229960004857 mitomycin Drugs 0.000 description 1
- 229960000350 mitotane Drugs 0.000 description 1
- KKZJGLLVHKMTCM-UHFFFAOYSA-N mitoxantrone Chemical compound O=C1C2=C(O)C=CC(O)=C2C(=O)C2=C1C(NCCNCCO)=CC=C2NCCNCCO KKZJGLLVHKMTCM-UHFFFAOYSA-N 0.000 description 1
- 229960001156 mitoxantrone Drugs 0.000 description 1
- 201000004058 mixed glioma Diseases 0.000 description 1
- ZDZOTLJHXYCWBA-BSEPLHNVSA-N molport-006-823-826 Chemical compound O([C@H]1[C@H]2[C@@](C([C@H](O)C3=C(C)[C@@H](OC(=O)[C@H](O)[C@@H](NC(=O)OC(C)(C)C)C=4C=CC=CC=4)C[C@@]1(O)C3(C)C)=O)(C)[C@@H](O)C[C@H]1OC[C@]12OC(=O)C)C(=O)C1=CC=CC=C1 ZDZOTLJHXYCWBA-BSEPLHNVSA-N 0.000 description 1
- 230000035772 mutation Effects 0.000 description 1
- 201000000050 myeloid neoplasm Diseases 0.000 description 1
- 230000017066 negative regulation of growth Effects 0.000 description 1
- 230000027405 negative regulation of phosphorylation Effects 0.000 description 1
- 210000000653 nervous system Anatomy 0.000 description 1
- 206010061311 nervous system neoplasm Diseases 0.000 description 1
- 210000004498 neuroglial cell Anatomy 0.000 description 1
- 230000003472 neutralizing effect Effects 0.000 description 1
- 208000004235 neutropenia Diseases 0.000 description 1
- 229910052757 nitrogen Inorganic materials 0.000 description 1
- 208000002154 non-small cell lung carcinoma Diseases 0.000 description 1
- 230000002018 overexpression Effects 0.000 description 1
- 229960001592 paclitaxel Drugs 0.000 description 1
- 210000000496 pancreas Anatomy 0.000 description 1
- 230000036961 partial effect Effects 0.000 description 1
- 230000037361 pathway Effects 0.000 description 1
- 229960001744 pegaspargase Drugs 0.000 description 1
- 108010001564 pegaspargase Proteins 0.000 description 1
- WVUNYSQLFKLYNI-AATRIKPKSA-N pelitinib Chemical compound C=12C=C(NC(=O)\C=C\CN(C)C)C(OCC)=CC2=NC=C(C#N)C=1NC1=CC=C(F)C(Cl)=C1 WVUNYSQLFKLYNI-AATRIKPKSA-N 0.000 description 1
- 229960002340 pentostatin Drugs 0.000 description 1
- FPVKHBSQESCIEP-JQCXWYLXSA-N pentostatin Chemical compound C1[C@H](O)[C@@H](CO)O[C@H]1N1C(N=CNC[C@H]2O)=C2N=C1 FPVKHBSQESCIEP-JQCXWYLXSA-N 0.000 description 1
- 235000019319 peptone Nutrition 0.000 description 1
- 229940066779 peptones Drugs 0.000 description 1
- 210000005105 peripheral blood lymphocyte Anatomy 0.000 description 1
- 230000035699 permeability Effects 0.000 description 1
- 230000000144 pharmacologic effect Effects 0.000 description 1
- 235000021317 phosphate Nutrition 0.000 description 1
- 150000003013 phosphoric acid derivatives Chemical class 0.000 description 1
- 238000003566 phosphorylation assay Methods 0.000 description 1
- 239000003757 phosphotransferase inhibitor Substances 0.000 description 1
- DCWXELXMIBXGTH-UHFFFAOYSA-N phosphotyrosine Chemical compound OC(=O)C(N)CC1=CC=C(OP(O)(O)=O)C=C1 DCWXELXMIBXGTH-UHFFFAOYSA-N 0.000 description 1
- NJBFOOCLYDNZJN-UHFFFAOYSA-N pipobroman Chemical compound BrCCC(=O)N1CCN(C(=O)CCBr)CC1 NJBFOOCLYDNZJN-UHFFFAOYSA-N 0.000 description 1
- 229960000952 pipobroman Drugs 0.000 description 1
- 229920001184 polypeptide Polymers 0.000 description 1
- 239000011591 potassium Substances 0.000 description 1
- 229910052700 potassium Inorganic materials 0.000 description 1
- 230000002265 prevention Effects 0.000 description 1
- 229960000624 procarbazine Drugs 0.000 description 1
- CPTBDICYNRMXFX-UHFFFAOYSA-N procarbazine Chemical compound CNNCC1=CC=C(C(=O)NC(C)C)C=C1 CPTBDICYNRMXFX-UHFFFAOYSA-N 0.000 description 1
- 230000008569 process Effects 0.000 description 1
- 238000004393 prognosis Methods 0.000 description 1
- 238000000159 protein binding assay Methods 0.000 description 1
- 230000003947 protein internalization Effects 0.000 description 1
- 230000017854 proteolysis Effects 0.000 description 1
- BWESROVQGZSBRX-UHFFFAOYSA-N pyrido[3,2-d]pyrimidine Chemical compound C1=NC=NC2=CC=CN=C21 BWESROVQGZSBRX-UHFFFAOYSA-N 0.000 description 1
- 235000005875 quercetin Nutrition 0.000 description 1
- 229960001285 quercetin Drugs 0.000 description 1
- 150000003246 quinazolines Chemical class 0.000 description 1
- 238000003653 radioligand binding assay Methods 0.000 description 1
- 206010037844 rash Diseases 0.000 description 1
- 102000027426 receptor tyrosine kinases Human genes 0.000 description 1
- 108091008598 receptor tyrosine kinases Proteins 0.000 description 1
- 210000000664 rectum Anatomy 0.000 description 1
- 230000001105 regulatory effect Effects 0.000 description 1
- 150000003839 salts Chemical class 0.000 description 1
- 230000011664 signaling Effects 0.000 description 1
- 210000003491 skin Anatomy 0.000 description 1
- 210000000813 small intestine Anatomy 0.000 description 1
- 239000011734 sodium Substances 0.000 description 1
- 229910052708 sodium Inorganic materials 0.000 description 1
- 239000007787 solid Substances 0.000 description 1
- 210000000952 spleen Anatomy 0.000 description 1
- 210000002784 stomach Anatomy 0.000 description 1
- 238000006467 substitution reaction Methods 0.000 description 1
- 150000003467 sulfuric acid derivatives Chemical class 0.000 description 1
- 229960001603 tamoxifen Drugs 0.000 description 1
- NRUKOCRGYNPUPR-QBPJDGROSA-N teniposide Chemical compound COC1=C(O)C(OC)=CC([C@@H]2C3=CC=4OCOC=4C=C3[C@@H](O[C@H]3[C@@H]([C@@H](O)[C@@H]4O[C@@H](OC[C@H]4O3)C=3SC=CC=3)O)[C@@H]3[C@@H]2C(OC3)=O)=C1 NRUKOCRGYNPUPR-QBPJDGROSA-N 0.000 description 1
- 229960001278 teniposide Drugs 0.000 description 1
- 238000012360 testing method Methods 0.000 description 1
- 210000001550 testis Anatomy 0.000 description 1
- BPEWUONYVDABNZ-DZBHQSCQSA-N testolactone Chemical compound O=C1C=C[C@]2(C)[C@H]3CC[C@](C)(OC(=O)CC4)[C@@H]4[C@@H]3CCC2=C1 BPEWUONYVDABNZ-DZBHQSCQSA-N 0.000 description 1
- 229960005353 testolactone Drugs 0.000 description 1
- 229960001196 thiotepa Drugs 0.000 description 1
- 210000001541 thymus gland Anatomy 0.000 description 1
- 229960003087 tioguanine Drugs 0.000 description 1
- MNRILEROXIRVNJ-UHFFFAOYSA-N tioguanine Chemical compound N1C(N)=NC(=S)C2=NC=N[C]21 MNRILEROXIRVNJ-UHFFFAOYSA-N 0.000 description 1
- 210000001519 tissue Anatomy 0.000 description 1
- 229960000303 topotecan Drugs 0.000 description 1
- UCFGDBYHRUNTLO-QHCPKHFHSA-N topotecan Chemical compound C1=C(O)C(CN(C)C)=C2C=C(CN3C4=CC5=C(C3=O)COC(=O)[C@]5(O)CC)C4=NC2=C1 UCFGDBYHRUNTLO-QHCPKHFHSA-N 0.000 description 1
- 239000011573 trace mineral Substances 0.000 description 1
- 235000013619 trace mineral Nutrition 0.000 description 1
- 230000009466 transformation Effects 0.000 description 1
- 238000011830 transgenic mouse model Methods 0.000 description 1
- 230000014616 translation Effects 0.000 description 1
- 238000011269 treatment regimen Methods 0.000 description 1
- 239000013638 trimer Substances 0.000 description 1
- 230000004565 tumor cell growth Effects 0.000 description 1
- OUYCCCASQSFEME-UHFFFAOYSA-N tyrosine Natural products OC(=O)C(N)CC1=CC=C(O)C=C1 OUYCCCASQSFEME-UHFFFAOYSA-N 0.000 description 1
- 230000003827 upregulation Effects 0.000 description 1
- 229960001055 uracil mustard Drugs 0.000 description 1
- 210000003932 urinary bladder Anatomy 0.000 description 1
- 210000004291 uterus Anatomy 0.000 description 1
- 239000013598 vector Substances 0.000 description 1
- 229960003048 vinblastine Drugs 0.000 description 1
- JXLYSJRDGCGARV-XQKSVPLYSA-N vincaleukoblastine Chemical compound C([C@@H](C[C@]1(C(=O)OC)C=2C(=CC3=C([C@]45[C@H]([C@@]([C@H](OC(C)=O)[C@]6(CC)C=CCN([C@H]56)CC4)(O)C(=O)OC)N3C)C=2)OC)C[C@@](C2)(O)CC)N2CCC2=C1NC1=CC=CC=C21 JXLYSJRDGCGARV-XQKSVPLYSA-N 0.000 description 1
- 229960004528 vincristine Drugs 0.000 description 1
- OGWKCGZFUXNPDA-XQKSVPLYSA-N vincristine Chemical compound C([N@]1C[C@@H](C[C@]2(C(=O)OC)C=3C(=CC4=C([C@]56[C@H]([C@@]([C@H](OC(C)=O)[C@]7(CC)C=CCN([C@H]67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)C[C@@](C1)(O)CC)CC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-XQKSVPLYSA-N 0.000 description 1
- OGWKCGZFUXNPDA-UHFFFAOYSA-N vincristine Natural products C1C(CC)(O)CC(CC2(C(=O)OC)C=3C(=CC4=C(C56C(C(C(OC(C)=O)C7(CC)C=CCN(C67)CC5)(O)C(=O)OC)N4C=O)C=3)OC)CN1CCC1=C2NC2=CC=CC=C12 OGWKCGZFUXNPDA-UHFFFAOYSA-N 0.000 description 1
- GBABOYUKABKIAF-GHYRFKGUSA-N vinorelbine Chemical compound C1N(CC=2C3=CC=CC=C3NC=22)CC(CC)=C[C@H]1C[C@]2(C(=O)OC)C1=CC([C@]23[C@H]([C@]([C@H](OC(C)=O)[C@]4(CC)C=CCN([C@H]34)CC2)(O)C(=O)OC)N2C)=C2C=C1OC GBABOYUKABKIAF-GHYRFKGUSA-N 0.000 description 1
- 229960002066 vinorelbine Drugs 0.000 description 1
- 238000001262 western blot Methods 0.000 description 1
- 238000012447 xenograft mouse model Methods 0.000 description 1
- 239000011701 zinc Substances 0.000 description 1
- 229910052725 zinc Inorganic materials 0.000 description 1
Classifications
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/395—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
- A61K39/39533—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
- A61K39/39558—Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K31/00—Medicinal preparations containing organic active ingredients
- A61K31/33—Heterocyclic compounds
- A61K31/395—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
- A61K31/435—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
- A61K31/47—Quinolines; Isoquinolines
- A61K31/4738—Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
- A61K31/4745—Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Medicinal Chemistry (AREA)
- Life Sciences & Earth Sciences (AREA)
- Public Health (AREA)
- Veterinary Medicine (AREA)
- Pharmacology & Pharmacy (AREA)
- Animal Behavior & Ethology (AREA)
- General Health & Medical Sciences (AREA)
- Epidemiology (AREA)
- Engineering & Computer Science (AREA)
- Microbiology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Immunology (AREA)
- Mycology (AREA)
- Organic Chemistry (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Biomedical Technology (AREA)
- Oncology (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Description
WO 2008/014386 PCT/US2007/074448 TREATMENT OF TUMORS IN PEDIATRIC PATIENTS WITH EPIDERMAL GROWTH FACTOR RECEPTOR ANTAGONISTS 5 CROSS-REFERENCE TO RELATED APPLICATIONS This application claims the benefit of U.S. provisional patent application Ser. No. 60/833,487, filed on July 27, 2006, the contents of which are incorporated by reference 10 herein in its entirety. FIELD OF THE INVENTION The present invention relates to treating tumors in pediatric patients by administering a combination of an EGFR antagonist and a chemotherapeutic agent. 15 BACKGROUND OF THE INVENTION In the United States, 11,900 children and adolescents under the age of 20 were diagnosed with cancer in 2001 and about 2,200 died of the disease. Solid tumors account for approximately 30% of cases of childhood cancers. Invasive brain and nervous system 20 cancers accounts for 17% of all pediatric cancers, second only to acute lymphocytic leukemia. About half of the diagnosed cases of brain tumors are malignant. Other common childhood cancers include Ewing's sarcoma, leukemia, neuroblastoma, osteosarcoma, rhabdomyosarcoma, soft tissue sarcoma, and Wilms' tumors. One of the major obstacles in treating brain tumors in children is that the brain is 25 still undergoing rapid development and is vulnerable to toxicities from treatments such as radiation or chemotherapy. Obstacles also arise in treating other types of childhood cancers. Therefore, new therapies for treating tumors in pediatric patients are needed. The epidermal growth factor receptor (EGFR) family is expressed or over expressed in various cancers and is generally involved in tumorigenesis. The EGFR family 30 includes the EGF receptor (EGFR, also known as erbB-1/HERI), HER2 (also known as c neu/erB-2), erbB-3/HER3, and erbB-4/HER4. For example, EGFR and HER2 are thought to play a critical role in processes that regulate tumor cell growth and survival. In particular, EGFR has been implicated in several pathways that affect survival and protection from apoptosis, dedifferentiation, and metastasis (including cell migration and 35 invasion). Among those cancers that express EGFR are some of the most prevalent including head and neck, colorectal, pancreatic, ovarian, renal cell, non-small cell lung, 1 WO 2008/014386 PCT/US2007/074448 and gliomas. Prognosis for many of these cancers is poor if not diagnosed at an early stage, and therapy for advanced disease is limited. There are various EGFR inhibitors: currently in clinical trials for the treatment of sonie of these cancers. One such example is ERBITUX® (cetuximab) (manufactured by 5 ImClone Systems Inc.), which is a chimeric (human/mouse) monoclonal antibody that blocks ligand binding to EGFR, prevents receptor activation, and inhibits growth. of cells in culture. Another example is ABX-EGF, which is a fully human monoclonal antibody specific to EGFR that reportedly blocks binding of transforming growth factor alpha (TFG-a) and epidermal growth factor (EGF), two ligands that are known to bind to EGFR. 10 HERCEPTIN® (trastuzumab) is a humanized antibody approved for the treatment of HER2.positive metastatic breast cancer, which is designed to target and block the function of HER2 protein overexpression. In addition, clinical trials are currently being conducted on various small molecule EGFR inhibitors. An example of a tyrosine kinase inhibitor is 1RESSATM, which is a 15 small molecule EGFR tyrosine kinase inhibitor that reportedly inhibits EGFR tyrosine kinase activity, is cytostatic towards a range of human cancer cells that express functional EGFR, and can inhibit tumor cell proliferation via up-regulation of p27. SUMMARY OF THE INVENTION 20 In certain embodiments, the present invention provides a method of inhibiting the growth of a tumor in a pediatric patient by treating the pediatric patient with an effective amount of an EGFR antagonist and a chemotherapeutic agent. In a preferred embodiment, the EGFR antagonist is an EGFR antibody that specifically binds to the extracellular domain of EGFR and neutralized activation thereof. In a preferred embodiment, the 25 chemotherapeutic agent is irinotecan. DETAILED DESCRIPTION OF THE INVENTION The present invention provides methods of treating tumors in pediatric patients by administering an effective amount of an EGFR antagonist and a chemotherapeutic agent. 30 Pediatric patients, according to'the present invention, are patients from birth to 18 years of age. Treatment includes: (1) preventing the disease from occurring in a patient who may be predisposed to the disease but does not yet experience or display symptoms of the disease; e.g., prevention of the outbreak of the clinical symptoms; (2) inhibiting the growth of the tumor, e.g., arresting its development; or (3) relieving the tumor, e.g., 2 WO 2008/014386 PCT/US2007/074448 causing regression of the symptoms of the tumor. Inhibiting tumor growth includes slowing or stopping growth, as well as causing tumor regression. An effective amount for the treatment of a disease means that amount which, when administered to a patient in need thereof, is sufficient to effect treatment, as defined above of the disease. 5 The tumors that are treated according to the present invention are any of the tumors that express EGFR. Such tumors include blastomas, including hepatoblastomas and neuroblastomas; carcinomas, including adenocarcinomas; gliomas, including ependymomas, astrocytomas, oligodendrogliomas and mixed gliomas; sarcomas, including rhabdomyosarcomas and adenosarcomas; and adenomas. The tumors can occur 10 in virtually all parts of the body, including, for example, the breast, heart, lung, esophagus, small intestine, colon, rectum, stomach, spleen, kidney, bladder, head and neck, larynx, ovary, prostrate, brain, pancreas,. skin, bone, bone marrow, blood, thymus, uterus, testicles, cervix, or liver. In a preferred embodiment, the tumors are tumors of the nervous system such as gliomas and neuroblastomas. The tumors to be treated include primary and 15 metastatic tumors, as well as refractory tumors. "Refractory tumors" include tumors,that fail to respond or are resistant to treatment with chemotherapeutic agents alone, antibodies alone, radiation alone or combinations thereof. Refractory tumors also encompass tumors that appear to be inhibited by treatment with such agents, but recur up to five years, sometimes up to ten years or longer after treatment is discontinued. 20 An EGFR antagonist, according to the present invention can be an extracellular antagonist or an intracellular antagonist and more than one antagonist may be employed. Extracellular antagonists include, but are not limited to, proteins or other biological molecules that bind to EGFR. In certain embodiments of the invention, an extracellular antagonist binds to the extracellular domain of EGFR and inhibits binding of EGFR to one 25 or more of its ligands and/or neutralizes ligand-induced activation of EGFR. Ligands for EGFR include EGF, TFG-a, amphiregulin, heparin-binding EGFR (HB-EGF) and betacellulin. Extracellular EGFR antagonists can also include substances that inhibit EGFR dimerization with other EGFR receptor subunits (ie. EGFR homodimers) or heterodimerization with other growth factor receptors (e.g., HER2). 30 In a preferred embodiment, the EGFR antagonist is an antibody that binds to EGFR and blocks ligand binding. One example of an EGFR antibody is cetuximab (IMC C225) (GenBank Accession No. 1NQLA), which is a chimeric (human/mouse) IgG monoclonal antibody. See e.g., U.S. Patent No. 4,943,533 (Mendelsohn et a.); U.S. Patent No. 6,217,866 (Schlessinger et al.); U.S. Application Nos. 08/973,065 (Goldstein et al.) 3 WO 2008/014386 PCT/US2007/074448 and 09/635,974 (Teufel); WO 99/60023 (Waksal et al.) and WO 00/69459, all of which are incorporated by reference herein. Cetuximab specifically binds to EGFR and blocks binding of a ligand, such as EGF. Cetuximab Fab contains the Fab fragment of Cetuximab, i.e., the heavy and light chain variable region sequences of murine antibody 5 M225 (U.S. App. Ser. No. 2004/0006212, incorporated herein by reference) with human IgGI CH1 heavy and kappa light chain constant domains. (Cetuximab includes all three IgG1 heavy chain constant domains.) The CDR regions of the heavy chain of cetuximab have the following sequences: a CDR1 region with a sequence ofN Y G V H (SEQ ID NO: 1), a CDR2 region with a sequence ofV I W S G G N T D Y N T P F T S (SEQ ID 10 NO: 2), and a CDR3 region with a sequence ofA L T Y YD YE F A Y (SEQ ID NO: 3). The CDR regions of the light chain of cetuximab have the following sequences: a CDR1 region with a sequence ofR A S Q S I G T N I H (SEQ ID NO: 4), a CDR2 region with a sequence ofY A S E S I S (SEQ ID NO: 5), and d CDR3 region with a sequence ofQ Q N N W P TT (SEQ ID NO: 6). 15. Another example of an EGFR antibody is ABX-EGF, which is a fully human IgG2 monoclonal antibody specific for EGFR. ABX-EGF binds EGFR with high specificity, blocking binding of EGFR to both of its ligands, EGF and TGF-a. See e.g., Figlin et al., Abstract 1102 presented at the 37th Annual Meeting of ASCO, San Francisco, CA, 12-15 May 2001, which is incorporated by reference herein. The sequence and characterization 20 of ABX-EGF, which was formerly known as clone E7.6.3, is disclosed in U.S. Patent No. 6,235,883 (Abgenix, Inc.) at col. 28, line 62 through col. 29, line 36 and in Fig. 29-34, which is incorporated by reference herein. (See also Yang et al., CriticalRev. Oncol./Hematol, 38(1): 17-23, 2001, which is incorporated by reference herein.) Another examples of an EGFR antibody is HERCEPTIN® (trastuzumab), which is 25 a recombinant DNA-derived humanized monoclonal antibody that selectively binds with high affinity in a cell-based assay (Kd of 5 nM) to the extracellular domain of the human EGFR2 protein, HER2. The antibody is an IgGi kappa that contains human framework regions with the complementarity-determining regions of a murine antibody (4D5) that binds to HER2. See, e.g., International Patent Publication No. WO 01/89566 (Mass), 30 which is incorporated by reference herein. Other EGFR antibodies include EMD 72000 (Merck KGaA), which is a humanized version of the murine anti-EGFR monoclonal antibody EMD 55900; h-R3 (TheraCIM), which is a humanized anti-EGFR monoclonal antibody; Y10, which is a murine monoclonal antibody and was raised against a murine homologue of the human 4 WO 2008/014386 PCT/US2007/074448 EGFRvIII mutation; and MDX-447 (Medarex). See U.S. Patent Nos. 5,558,864 (Bendig et al.), 5,884,093 (Kettleborough et al.), and 5,891,996 (Mateo de Acosta del Rio et al.), all of which are incorporated by reference herein. Intracellular EGFR antagonists can be biological molecules, but are usually small 5 molecules, such as synthetic kinase inhibitors that act directly on the cytoplasmic domain of EGFR to inhibit EGFR-mediated signal transduction. One example of a small molecule EGFR antagonist is IRESSA m (ZD1939), which is a quinozaline derivative that functions as an ATP-mimetic to inhibit EGFR. SeeU.S. Patent No. 5,616,582 (Zeneca Limited); WO 96/33980 (Zeneca Limited) at p. 4; see also, Rowinsky et al., Abstract 5 presented at 10 the 37th Annual Meeting of ASCO, San Francisco, CA, 12-15 May 2001; Anido et al., Abstract 1712 presented at the 37th Annual Meeting of ASCO, San Francisco, CA, 12-15 May 2001. Another example of a small molecule EGFR antagonist is TARCEVA® (OSI 774), which is a 4-(substitutedphenylamino)quinozaline derivative [6,7-Bis(2-methoxy ethoxy)-quinazolin-4-yl]- (3-ethynyl-phenyl)amine hydrochloride] EGFR inhibitor. See 15 WO 96/30347 (Pfizer Inc.) at, for example, page.2, line 12 through page 4, line 34 and page 19, lines 14-17. See also Moyer etal., CancerRest; 57: 4838-48 (1997); Pollack et al., . Pharmacol., 291: 739-48 (1999). TARCEVA® may function by inhibiting phosphorylation of EGFR and its downstream PI3/Akt and MAP (mitogen activated protein) kinase signal transduction pathways resulting in p27-mediated cell-cycle arrest. 20 See Hidalgo et al., Abstract 281 presented at the 37th Annual Meeting of ASCO, San Francisco, CA, 12-15 May 2001. Other small molecules are also reported to inhibit EGFR, many of which are thought to be specific to the tyrosine kinase domain of an EGFR. Some examples of such small molecule EGFR antagonists are described in WO 91/116051, WO 96/30347, WO 25 96/33980, WO 97/27199 (Zeneca Limited). WO 97/30034 (Zeneca Limited), WO 97/42187 (Zeneca Limited), WO 97/49688 (Pfizer Inc.), WO 98/33798 (Warner Lambert Company), WO 00/18761 (American Cyanamid Company), and WO 00/31048 (Warner Lambert Company). Examples of specific small molecule EGFR antagonists include Cl 1033 (Pfizer), which is a quinozaline (N-[4-(3-chloro-4-fluoro-phenylamino)-7-(3 30 morpholin-4-yl-propoxy)-quinazolin-6-yl]-acrylamide) inhibitor of tyrosine kinases, particularly EGFR and is described in WO 00/31048 at page 8, lines 22-6; PKI 66 (Novartis), which is a pyrrolopyrimidine inhibitor of EGFR and is described in WO 97/27199 at pages 10-12; GW2016 (GlaxoSmithKline), which is an inhibitor of EGFR and HER2; EKB569 (Wyeth), which is reported to inhibit the growth of tumor cells that 5 WO 2008/014386 PCT/US2007/074448 overexpress EGFR or HER2 in vitro and in vivo; AG-1478 (Tryphostin), which is a quinazoline small molecule that inhibits signaling from both EGFR and erbB-2; AG-1478 (Sugen), which is a bisubstrate inhibitor that also inhibits protein kinase CK2;, PD 153035 (Parke-Davis) which is reported to inhibit EGFR kinase activity and tumor growth, induce 5 apoptosis in cells in culture, and enhance the cytotoxicity of cytotoxic chemotherapeutic agents; SPM-924 (Schwarz Pharma), which is a tyrosine kinase inhibitor targeted for treatment of prostrate cancer; CP-546,989 (OSI Pharmaceuticals), which is reportedly an inhibitor of angiogenesis for treatment of solid tumors; ADL-681, which is a EGFR kinase inhibitor targeted for treatment of cancer; PD 158780, which is a pyridopyrimidine that's 10 reported to inhibit the tumor growth rate ofA4431 xenografts in mice; CP-358,774, which is a quinzoline that is reported to inhibit autophosphorylation in HN5 xenografts in mice; ZD 1839, which is a quinzoline that is reported to have antitumor activity in mouse xenograft models including vulvar, NSCLC, prostrate, ovarian, and colorectal cancers; CGP 59326A, which is a pyrrolopyrimidine that is reported to inhibit growth of EGFR 15 positive xenografts in mice; PD 165557 (Pfizer); CGP54211 and CGP53353 (Novartis), which are dianilnophthalimides. Naturally derived EGFR tyrosine kinase inhibitors include genisteini, herbimycin A, quercetin, and erbstatin. Further small molecules reported to inhibit EGFR and that are therefore within the scope of the present invention are tricyclic compounds such as the compounds described 20 in U.S. Patent No. 5,679,683; quinazoline derivatives such as the derivatives described in U.S. Patent No. 5,616,582; and indole compounds such as the compounds described in U.S. Patent No. 5,196,446. It will be appreciated that useful small molecule to be used in the invention are inhibitors of EGFR, but need not be completely specific for EGFR. 25 In a preferred embodiment, the EGFR antagonist is an anti-EGFR antibody that exhibits one or more of following properties: 1) The antibody binds to the external domain of EGFR and inhibits ligand binding. Inhibition can be determined, for example, by a direct binding assay using purified or membrane bound receptor. In this embodiment, the antibodies of the present invention or 30 fragments thereof, preferably bind EGFR at least as strongly as the natural ligands of EGFR (EGF, TFG-a). 2) The antibodies neutralize EGFR. Binding of a ligand to an external, extracellular domain of EGFR stimulates tyrosine kinase activity and receptor phosphorylation and/or the phosphorylation of other proteins involved in the various 6 WO 2008/014386 PCT/US2007/074448 signaling pathways. Neutralization of EGFR includes inhibition, diminution, inactivation and/or disruption of one or more of the activities normally associated with signal transduction. Neutralization can be determined in vivo, ex vivo, or in vitro using, for example, tissues, cultured cell, or purified cellular components. 5 One measure of EGFR neutralization is inhibition of the tyrosine kinase activity of the receptor. Tyrosine kinase inhibition can be determined using well-known methods; for example, by measuring the autophosphorylation level of recombinant kinase receptor, and/or phosphorylation of natural or synthetic substrates. Thus, phosphorylation assays are useful in determining neutralizing antibodies in the context of the present invention. 10 Phosphorylation can be detected, for example, using an antibody specific for phosphotyrosine in an ELISA assay or on a western blot. Some assays for tyrosine kinase activity are described in Panek et al., J. Pharmacol. Exp. Thera. 283: 1433-44 (1997) and Batley et al., Life Sc1 62:143-50 (1998). Antibodies of the invention cause a decrease in tyrosine phosphorylation of EGFR of at least about 75%, preferably at least about 85%, 15 and more preferably at least about 90% in cells that 'espond to ligand. Another measure of EGFR neutralization is inhibition of phosphorylation of downstream substrates of EGFR. Accordingly, the level of phosphorylation of MEK and ERK can be measured. The decrease in phosphorylation is at least about 40%, and can be at least about 60%, or at least about 80%. 20 In addition, methods for detection of protein expression can be utilized to determine EGFR neutralization, wherein the proteins being measured are regulated by EGFR tyrosine kinase activity. These methods include immunohistochemistry (IHC) for detection of protein expression, fluorescence in situ hybridization (FISH) for detection of gene amplification, competitive radioligand binding assays, solid matrix blotting 25 techniques, such as Northern and Southern blots, reverse transcriptase polymerase chain reaction (RT-PCR) and ELISA. See, e.g., Grandis et al., Cancer, 78:1284-92 (1996); Shimizu et al., Japan I. CancerRes., 85:567-71 (1994); Sauter et al., Am. J. Path., 148:1047-53 (1996); Collins, Glia 15:289-96 (1995); Radinsky et al., Clin. Cancer Res. 1:19-31 (1995); Petrides et al., CancerRes. 50:3934-39 (1990); Hoffmann et al., 30 AnticancerRes. 17:4419-26 (1997); Wikstrand et al., CancerRes. 55:3140-48 (1995). Ex vivo assays can also be utilized to determine EGFR neutralization. For example, receptor tyrosine kinase inhibition can be observed by mitogenic assays using cell lines stimulated with receptor ligand in the presence and absence of inhibitor. An example of such a mitogenic assay is a 3T3 cells mitogenic assay (3T3 (clone A31-714) 7 WO 2008/014386 PCT/US2007/074448 cells from American Type Culture Collection (Manassas, VA)). Another method involves testing for inhibition of growth of EGFR-expressing tumor cells or cells transfected to express EGFR. Inhibition can also be observed using tumor models, for example, human tumor cells injected into a mouse. 5 The antibodies of the present invention are not limited by any particular mechanism of EGFR neutralization. The anti-EGFR antibodies of the present invention can bind externally to EGFR cell surface receptor, block binding of ligand and subsequent signal transduction mediated via the receptor-associated tyrosine kinase, and prevent phosphorylation of the EGFR and other downstream proteins in the signal transduction 10 cascade. 3) The antibodies down modulate EGFR. The amount of EGFR present on the surface of a cell depends on receptor protein production, internalization, and degradation. The amount of EGFR present on the surface of a cell can be measured indirectly, by detecting internalization of the receptor or a molecule bound to the receptor. For example, 15 receptor internalization can be measured by contacting cells that express EGFR with a labeled antibody. Membrane-bound antibody is then stripped, collected and counted. Internalized antibody is determined by lysing the cells and detecting label in the lysates. Another way is to directly measure the amount of the receptor present on the cell following treatment with an anti-EGFR antibody or other substance, for example, by 20 fluorescence-activated cell-sorting analysis of cells stained for surface expression of EGFR. Stained cells are incubated at 37 0 C and fluorescence intensity measured over time. As a control, part of the stained population can be incubated at 4 0 C (conditions under which receptor internalization is halted). Another measure of down-modulation is reduction of the total receptor protein 25 present in a cell, and reflects degradation of internal receptors. Accordingly, treatment of cells (particularly cancer cells) with antibodies of the invention results in a reduction in total cellular EGFR. In a preferred embodiment, the reduction is at least about 70%, more preferably at least about 80%, and even more preferably at least about 90%. For treatment of human subjects, antibodies according to the invention are 30 preferably human. Alternatively, the antibodies can be from non-human primates or other mammals, or be humanized or chimeric antibodies. Antibody fragments according to the invention can be produced by cleaving a whole antibody, or by expressing DNA that encodes the fragment. Fragments of antibodies may be prepared by methods described by Lamoyi et al., J. Immunol. Methods, 8 WO 2008/014386 PCT/US2007/074448 56: 235-243 (1983) and by Parham, J. Immunol 131: 2895-2902 (1983). Such fragments may contain one or both Fab fragments or the F(ab') 2 fragment. Such fragments may also contain single-chain fragment variable region antibodies, i.e. seFv, dibodies, or other antibody fragments. Methods of producing such functional equivalents are disclosed in 5 PCT Application WO 93/21319, European Patent Application No. EP 239400; PCT Application WO 89/09622; European Patent Application EP 338745; and European Patent Application EP 332424. Preferred host cells for transformation of vectors and expression of the antibodies of the present invention are mammalian cells, e.g., COS-7 cells, Chinese hamster ovary 10 (CHO) cells, and cell lines of lymphoid origin such as lymphoma, myeloma (e.g. NSO), or hybridoma cells. Other eukaryotic hosts, such as yeasts, can be alternatively used. The transformed host cells are cultured by methods known in the art in a liquid medium containing assimilable sources of carbon (carbohydrates such as glucose or lactose), nitrogen (amino acids, peptides, proteins or their degradation products such as 15 peptones, ammonium salts or the like), and inorganic salts (sulfates, phosphates and/or carbonates of sodium, potassium, magnesium and calcium). The medium furthermore contains, for example, growth-promoting substances, such as trace elements, for example iron, zinc, manganese and the like. High affinity anti-EGFR antibodies according to the present invention can be 20 isolated from a phage display library constructed from human heavy chain and light chain variable region genes. For example, a variable domain of the invention can be obtained from a peripheral blood lymphocyte that contains a rearranged variable region gene. Alternatively, variable domain portions, such as CDR and FW regions, can be obtained from different sources and recombined. Further, portions of the variable domains (e.g., 25 FW regions) can be synthetic consensus sequences. Antibodies and antibody fragments of the present invention can be obtained, for example, from naturally occurring antibodies, or Fab or scFv phage display libraries. It is understood that, to make a single domain antibody from an antibody comprising a V, and a VL domain, certain amino acid substitutions outside the CDRs can be desired to enhance 30 binding, expression or solubility. For example, it can be desirable to modify amino acid residues that would otherwise be buried in the VH-VL interface. Further, antibodies and antibody fragments of the invention can be obtained by standard hybridoma technology (Harlow & Lane, ed., Antibodies: A Laboratory Manual, Cold Spring Harbor, 211-213 (1998), which is incorporated by reference herein) using 9 WO 2008/014386 PCT/US2007/074448 transgenic mice (e.g., KM mice from Medarex, San Jose, Calif.) that produce human immunoglobulin gamma heavy and kappa light chains. In a preferred embodiment, a substantial portion of the human antibody producing genome is inserted into the genome of the mouse, and is rendered deficient in the production of endogenous murine antibodies. 5 Such mice may be immunized subcutaneously (s.c.) with PDGFRa (usually in complete Freund's adjuvant) with boosts as needed. Immunization methods are well known in the art. Antibodies that can be used according to the invention include complete immunoglobulins, antigen binding fragments of immunoglobulins, as well as antigen 10 binding proteins that comprise antigen binding domains of immunoglobulins. Antigen binding fragments of immunoglobulins include, for example, Fab, Fab', and F(ab') 2 . Other antibody formats have been developed which retain binding specificity, but have other characteristics that may be desirable, including for example, bispecificity, multivalence (more than two binding sites), compact size (e.g., binding domains alone). 15 Single chain antibodies lack some or all of the constant domains of the whole antibodies from which they are derived. Therefore, they can overcome some of the problems associated with the use of whole antibodies. For example, single-chain antibodies tend to be free of certain undesired interactions between heavy-chain constant regions and other biological molecules. Additionally, single-chain antibodies are 20 considerably smaller than whole antibodies and can have greater permeability than whole antibodies, allowing single-chain antibodies to localize and bind to target antigen-binding sites more efficiently. Furthermore, the relatively small size of single-chain antibodies makes them less likely to provoke an unwanted immune response in a recipient than whole antibodies. 25 Multiple single chain antibodies, each single chain having one VH and one VL domain covalently linked by a first peptide linker, can be covalently linked by at least one or more peptide linker to form a multivalent single chain antibodies, which can be monospecific or multispecific. Each chain of a multivalent single chain antibody includes a variable light chain fragment and a variable heavy chain fragment, and is linked by a 30 peptide linker to at least one other chain. The peptide linker is composed of at least fifteen amino acid residues. The maximum number of amino acid residues is about one hundred. Two single chain antibodies can be combined to form a diabody, also known as a bivalent dimer. Diabodies have two chains and two binding sites, and can be monospecific or bispecific. Each chain of the diabody includes a VH domain connected to 10 WO 2008/014386 PCT/US2007/074448 a VL domain. The domains are connected with linkers that are short enough to prevent pairing between domains on the same chain, thus driving the pairing between complementary domains on different chains to recreate the two antigen-binding sites. Three single chain antibodies can be combined to form triabodies, also known as 5 trivalent trimers. Triabodies are constructed with the amino acid terminus of a VL or VH domain directly fused to the carboxyl terminus of a VL or VH domain, i.e., without any linker sequence. The triabody has three Fv heads with the polypeptides arranged in a cyclic, head-to-tail fashion. A possible conformation of the triabody is planar with the three binding sites located in a plane at an angle of 120 degrees from one another. 10 Triabodies can be monospecific, bispecific or trispecific. Thus, antibodies of the invention and fragments thereof include, but are not limited to, naturally occurring antibodies, bivalent fragments such as (Fab') 2 , monovalent fragments such as Fab, single chain antibodies, single chain Fv (scFv), single domain antibodies, multivalent single chain antibodies, diabodies, triabodies, and the like that bind 15 specifically with antigens. According to the present invention, an EGFR antagonist is administered in combination with one or more anti-neoplastic agents. As employed herein, the term, "anti-neoplastic" agent excludes an EGFR antagonist, unless otherwise specified. Any suitable anti-neoplastic agent can be used, such as a chemotherapeutic agent, radiation, or 20 combinations thereof. The anti-neoplastic agent Can be an alkylating agent or an anti metabolite. Examples of alkylating agents include, but are not limited to, cisplastin, cyclophosphamide, melphalan, and dacarbazine. Examples of anti-metabolites include, but are not limited to doxorubicin, dunorubicin, paclitaxel, and gemcitabine. In a preferred embodiment, the anti-neoplastic agent is a chemotherapeutic agent. 25 Preferred chemotherapeutic agents include amifostine (ethyol), cisplatin, dacarbazine (DTIC), dactinomycin, mechlorethamine (nitrogen mustard), streptozocin, cyclophosphamide, carmustine (BCNU), lomustine (CCNU), doxorubicin (adriamycin), doxorubicin lipo (doxil), gemcitabine (gemzar), daunorubicin, daunorubicin lipo (daunoxome),.procarbazine, mitomycin, cytarabine, etoposide, methotrexate, 5 30 fluorouracil, vinblastine, vincristine, bleomycin, paclitaxel (taxol), docetaxel (taxotere), aldesleukin, asparaginase, busulfan, carboplatin, cladribine, camptothecin, CPT-11,10 hydroxy-7-ethyl-camptothecin (SN38), dacarbazine, floxuridine, fludarabine, hydroxyurea, ifosfamide, idarubicin, mesna, interferon alpha, interferon beta, irinotecan, mitoxantrone, topotecan, leuprolide, megestrol, melphalan, mercaptopurine, plicamycin, 11 WO 2008/014386 PCT/US2007/074448 mitotane, pegaspargase, pentostatin, pipobroman, plicamycin, streptozocin, tamoxifen, teniposide, testolactone, thioguanine, thiotepa, uracil mustard, vinorelbine, chlorambucil and combinations thereof. In a more preferred embodiment, the chemotherapeutic agent is irinotecan. 5 An anti-neoplastic agent, including a chemotherapeutic agent and/or an anti metabolite, is administered in a dose and on a schedule according to the package directions and/or the clinical response of the patient. Such doses will be apparent to one of ordinary skill and do not require undue experimentation. For example, Ironotecan is preferably administered over the recommended dose range of from about 50 to about 350 mg/m2 10 although the dose maybe higher or lower, depending on the clinical response of the patient. Other agents have dosage ranges of from about 1 to about 1,000 mg/m 2 per administration and/or treatment regimen. Radiation treatment is administered based on the appropriate clinical treatment protocols and/or clinical experience of one of skill in the art. In a combination therapy, the EGFR antagonist is administered before, during, or 15 after commencing therapy with another agent, as well as any combination thereof, i.e., before and during, before and after, during and after, or before, during and after commencing the anti-neoplastic agent therapy. For example, an EGFR antibody can be administered between 1 and 30 days, preferably 3 and 20 days, more preferably between 5 and 12 days before commencing radiation therapy. In a preferred embodiment of the 20 invention, chemotherapy is administered concurrently with or, more preferably, subsequent to antibody therapy. In the present invention, any suitable method or route can be used to administer the antagonists of the invention, and optionally, to co-administer anti-neoplastic agents and/or antagonists of other receptors. The anti-neoplastic agent regimens utilized according to 25 the invention, include any regimen believed to be optimally suitable for the treatment of the patient's tumor. Different tumors can require use of specific anti-tumor antibodies and specific anti-neoplastic agents, which will be determined on a patient to patient basis. Routes of administration include, for example, oral, intravenous, intraperitoneal, subcutaneous, or intramuscular administration. The dose of antagonist administered 30 depends on numerous factors, including, for example, the type of antagonists, the type and severity of the tumor being treated and the route of administration of the antagonists. It should be emphasized, however, that the present invention is not limited to any particular method or route of administration. 12 WO 2008/014386 PCT/US2007/074448 One of skill in the art would understand that dosages arid frequency of treatment depend on the tolerance of the individual patient and on the pharmacological and pharmacokinetic porperties of blocking or inhibitory agent used. Ideally, one wishes to achieve saturable pharmacokinetics for the agent used. A loading dose for an anti-EGFR 5 antibody can range, for example, from about 10 to about 1000 mg/m 2 , preferably from about 200 to about 400 mg/m 2. This can be followed by several additional daily or weeldy dosages ranging, for example, from about 200 to about 400 mg/m 2 . The patient is monitored for side effects and the treatment is stopped when such side effects are severe. One of skill in the art would also know how to monitor the progress of the 10 treatment in order to determine an effective dose. For example, one such .way is to monitor MRI, CT, or other brain scans. EXAMPLES Example 1: The following example discloses a method of treating refractory solid tumors 15 by administering a combination of irinotecan and an EGFR antibody. From August 2005 to March 2006, 20 pediatric patients with refractory solid tumors and life expectancy of at least 8 weeks were administered irinotecan and different dose levels of cetuximab. The patients were divided into two groups (ages 1-12 = Arm A, and 13-18 = Arm B) with the following tumor types: brainstem gliomal astrocytoma (10), 20 hepatoblastoma, osteosarcoma (1), ependymoma (1), neuroblastoma (1), rhabdomyosarcoma (1) and other (5). Irinotecan was administered at a dose of 20 mg/m2 /day as a 60 minute infusion for 5 days x 2 weeks, every 21 days. The dose schedules and number of subjects with dose-limiting toxicities (DLTs) are provided in Table 1. 25 Table 1 Arm Dose Cetuximab Irinotecan No. of No. of subjects with level (mg/m 2 /day (mg/m2/day subjects DLT (dose-limiting days 1, 8, 15) days 1-5 and toxicity) 8-12) A 1 75 20 6 1 ( Gr 3 diarrhea) A 2 150 20 6 2 ( Gr 4 neutropenia, Gr 3 diarrhea) B 1 75 20 81 (Gr 3 diarrhea) Due to the DLTs experienced by two out of the six patients in Arm A, dose level 2, which were deemed to be related to irinotecan, irinotecan was de-escalated to 16 mg/nm 2 13 WO 2008/014386 PCT/US2007/074448 One subject in Annrm B experienced a Grade 3 infusion reaction and treatment was discontinued. Other observed toxicities included grade 1 rash. One subject with an EGFR-negative high grade glioma (group A dose level 1) achieved a partial response and is currently in cycle 13. One subject with Neuroblastoma 5 (group A dose level 1) has experienced a minor response and is currently receiving cycle 9. Seven subjects had a best response of stable disease for an average of 3.4 months (ranging from 2- 7+ mo). In conclusion, the combination of cetuximab and irinotecan shows promising anti cancer activity in pediatric solid tumors. 10 The foregoing description and example have been set forth merely to illustrate the invention and are not intended as being limiting. Each of the disclosed aspects and embodiments of the present invention may be considered individually or in combination with other aspects, embodiments, and variations of the invention. Further, while certain features of embodiments of the present invention may be shown in only certain figures, 15 such features can be incorporated into other embodiments shown in other figures while remaining within the scope of the present invention. In addition, unless otherwise specified, none of the steps of the methods of the present invention are confined to any particular order of performance. Modifications of the disclosed embodiments incorporating the spirit and substance of the invention may occur to persons skilled in the 20 art and such modifications are within the scope of the present invention. Furthermore, all references cited herein are incorporated by reference in their entirety. 14
Claims (14)
1. A method of inhibiting the growth of a tumor in a pediatric patient comprising treating the pediatric patient with an effective amount of an epidermal growth factor 5 receptor (EGFR) antagonist and a chemotherapeutic agent.
2. The method of claim 1, wherein the tumor is a refractory tumor.
3. The method of claim 1, wherein the tumor is a glioma. 10
4. The method of claim 1, wherein the tumor is a neuroblastoma.
5. The method of claim 1, wherein the antagonist is an antibody or fragment thereof. 15
6. The method of claim 5, wherein the antibody or fragment thereof specifically binds to the extracellular domain of EGFR.
7. The method of claim 5, wherein the antibody or fragment thereof inhibits binding of an EGFR ligand to an EGFR and neutralizes activation thereof. 20
8. The method of claim 5, wherein the antibody is cetuximab.
9. The method of claim 5, wherein the antibody or fragment thereof is monoclonal. 25
10. The method of claim 5, wherein the antibody or fragment thereof is chimeric.
11. The method of claim 5, wherein the antibody or fragment thereof is humanized.
12. The method of claim 5, wherein the antibody or fragment thereof is human. 30
13. The method of claim 1, wherein the chemotherapeutic agent is irinotecan.
14. The method of claim 1, further comprising administering radiation to the pediatric patient. 15
Applications Claiming Priority (3)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| US83348706P | 2006-07-27 | 2006-07-27 | |
| US60/833,487 | 2006-07-27 | ||
| PCT/US2007/074448 WO2008014386A2 (en) | 2006-07-27 | 2007-07-26 | Treatment of tumors in pediatric patients with epidermal growth factor receptor antagonists |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| AU2007279261A1 true AU2007279261A1 (en) | 2008-01-31 |
Family
ID=38982334
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| AU2007279261A Abandoned AU2007279261A1 (en) | 2006-07-27 | 2007-07-26 | Treatment of tumors in pediatric patients with epidermal growth factor receptor antagonists |
Country Status (15)
| Country | Link |
|---|---|
| EP (1) | EP2043688A4 (en) |
| JP (1) | JP2009544736A (en) |
| KR (1) | KR20090033841A (en) |
| CN (1) | CN101484186A (en) |
| AU (1) | AU2007279261A1 (en) |
| BR (1) | BRPI0712368A2 (en) |
| CA (1) | CA2654911A1 (en) |
| CR (1) | CR10486A (en) |
| EA (1) | EA200870603A1 (en) |
| EC (1) | ECSP089011A (en) |
| MX (1) | MX2008016187A (en) |
| NO (1) | NO20085182L (en) |
| TN (1) | TNSN08512A1 (en) |
| WO (1) | WO2008014386A2 (en) |
| ZA (1) | ZA200810600B (en) |
Families Citing this family (1)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| EA202192413A1 (en) * | 2019-03-27 | 2021-12-30 | Нэшнл Рисеч Каунсил Оф Канада | ANTIBODIES AGAINST EGFRvIII AND THEIR ANTIGEN-BINDING FRAGMENTS |
-
2007
- 2007-07-26 CA CA002654911A patent/CA2654911A1/en not_active Abandoned
- 2007-07-26 AU AU2007279261A patent/AU2007279261A1/en not_active Abandoned
- 2007-07-26 BR BRPI0712368-0A patent/BRPI0712368A2/en not_active Application Discontinuation
- 2007-07-26 JP JP2009522001A patent/JP2009544736A/en not_active Withdrawn
- 2007-07-26 WO PCT/US2007/074448 patent/WO2008014386A2/en not_active Ceased
- 2007-07-26 CN CNA2007800249991A patent/CN101484186A/en active Pending
- 2007-07-26 MX MX2008016187A patent/MX2008016187A/en not_active Application Discontinuation
- 2007-07-26 EP EP07813400A patent/EP2043688A4/en not_active Withdrawn
- 2007-07-26 KR KR1020087031606A patent/KR20090033841A/en not_active Withdrawn
- 2007-07-26 EA EA200870603A patent/EA200870603A1/en unknown
-
2008
- 2008-12-09 CR CR10486A patent/CR10486A/en not_active Application Discontinuation
- 2008-12-11 NO NO20085182A patent/NO20085182L/en not_active Application Discontinuation
- 2008-12-11 TN TNP2008000512A patent/TNSN08512A1/en unknown
- 2008-12-15 ZA ZA200810600A patent/ZA200810600B/en unknown
- 2008-12-24 EC EC2008009011A patent/ECSP089011A/en unknown
Also Published As
| Publication number | Publication date |
|---|---|
| CR10486A (en) | 2009-02-23 |
| ECSP089011A (en) | 2009-01-30 |
| JP2009544736A (en) | 2009-12-17 |
| MX2008016187A (en) | 2009-01-20 |
| CA2654911A1 (en) | 2008-01-30 |
| KR20090033841A (en) | 2009-04-06 |
| BRPI0712368A2 (en) | 2012-06-05 |
| ZA200810600B (en) | 2009-11-25 |
| CN101484186A (en) | 2009-07-15 |
| EA200870603A1 (en) | 2009-06-30 |
| WO2008014386A2 (en) | 2008-01-31 |
| TNSN08512A1 (en) | 2010-04-14 |
| EP2043688A4 (en) | 2009-11-11 |
| EP2043688A2 (en) | 2009-04-08 |
| WO2008014386A3 (en) | 2008-07-03 |
| NO20085182L (en) | 2009-04-24 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| CN101612399B (en) | Anti-PDGFRα antibody for use in the treatment of secondary bone tumors | |
| JP5179702B2 (en) | Pharmaceutical composition for receptor Erb-B1 | |
| JP2022071071A (en) | Antibodies that bind to ErbB-2 and ErbB-3 | |
| CN109071666A (en) | Human poliovirus receptor (PVR) specific antibody | |
| AU2006332212B8 (en) | Combination therapy using anti-EGFR and anti-HER2 antibodies | |
| US20220112283A1 (en) | Antibodies specific to human nectin-2 | |
| US20120201817A1 (en) | Methods of inhibiting receptor tyrosine kinases with an extracellular antagonist and an intracellular antagonist | |
| KR20240150805A (en) | Antibody drug conjugates comprising STING agonists, combinations and methods of use | |
| AU2007279261A1 (en) | Treatment of tumors in pediatric patients with epidermal growth factor receptor antagonists | |
| WO2008095015A2 (en) | Method of treating recurrent tumors | |
| KR20220153615A (en) | Medicines for the treatment and/or prevention of cancer | |
| JP7788118B2 (en) | Antibody specific to human nectin-2 | |
| HK1135917B (en) | Antibody against pdgfr-alpha for use in the treatment of tumours | |
| HK1135916A (en) | Pdgfr-alpha antagonists for treatment of metastatic bone cancer |
Legal Events
| Date | Code | Title | Description |
|---|---|---|---|
| MK1 | Application lapsed section 142(2)(a) - no request for examination in relevant period |