WO2012173867A1 - Dosage and administration of anti-erbb3 antibodies in combination with tyrosine kinase inhibitors - Google Patents
Dosage and administration of anti-erbb3 antibodies in combination with tyrosine kinase inhibitors Download PDFInfo
- Publication number
- WO2012173867A1 WO2012173867A1 PCT/US2012/041339 US2012041339W WO2012173867A1 WO 2012173867 A1 WO2012173867 A1 WO 2012173867A1 US 2012041339 W US2012041339 W US 2012041339W WO 2012173867 A1 WO2012173867 A1 WO 2012173867A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- dose
- antibody
- seq
- erlotinib
- gly
- Prior art date
Links
Classifications
-
- 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
- A61K31/495—Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
- A61K31/505—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
- A61K31/517—Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
-
- 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
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K16/00—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
- C07K16/18—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
- C07K16/32—Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
Definitions
- the ErbB3 receptor is 148 kilodalton (kD) transmembrane receptor tyrosine kinase belonging to the ErbB/EGFR family and is known to be kinase dead.
- the ErbB family of transmembrane receptor tyrosine kinases impacts the physiology of cells and organs by eliciting ligand-dependent activation of multiple signal transduction pathways.
- ErbB3-containing heterodimers such as ErbB2/ErbB3 in tumor cells have been shown to be the most mitogenic and oncogenic receptor complex within the ErbB family.
- HRG heregulin
- ErbB3/ErbB2 dimerization results in transphosphorylation of ErbB3 on tyrosine residues contained within the cytoplasmic tail of the protein. Phosphorylation of these sites creates SH2 docking sites for SH2-containing proteins, including PI3-kinase.
- ErbB3-containing heterodimeric complexes are potent activators of AKT as ErbB3 possesses six tyrosine phosphorylation sites with YXXM motifs that, when phosphorylated, serve as excellent binding sites for phosphoinositol-3-kinase (PI3K) the action of which results in subsequent downstream activation of the AKT pathway. These six PI3K sites serve as a strong amplifier of ErbB3 signaling. Activation of this pathway further elicits several important biological processes involved in tumorogenesis, such as cell growth and survival.
- Heregulin a cognate ligand of ErbB2/ErbB3 heterodimers, has been shown to be involved in several different types of cancer, including breast, ovarian, endometrial colon, gastric, lung, thyroid, glioma, medulloblastoma, melanoma as well as head and neck squamous cell carcinoma.
- HRG regulates growth, invasion and angiogenesis through either over expression or the activation of an autocrine or paracrine loop.
- Disruption of the heregulin autocrine loop by blocking HRG binding or disruption of the ErbB2/ErbB3 dimer may provide an important therapeutic measure to control growth in certain cancers.
- compositions and methods for treating non-small-cell lung cancer (NSCLC) in a human patient comprising administering to the patient a combination of an anti-ErbB3 antibody and a small molecule tyrosine kinase inhibitor (TKI), wherein the combination is administered according to a particular clinical dosage regimen (i.e., at a particular dose amount and according to a specific dosing schedule).
- NSCLC non-small-cell lung cancer
- an exemplary anti-ErbB3 antibody is Antibody A (including antigen binding fragments and variants thereof)-
- the anti-ErbB3 antibody comprises variable heavy (VH) and/or variable light (VL) regions encoded by the nucleic acid sequences set forth in SEQ ID NOs: l and 3, respectively.
- the antibody comprises VH and/or VL regions comprising the amino acid sequences set forth in SEQ ID NOs 2 and 4, respectively.
- the antibody comprises CDRHl , CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and/or CDRL1, CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRL1) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3).
- an antibody is used that competes for binding with and/or binds to the same epitope on human ErbB3 as the above-mentioned antibodies.
- the epitope comprises residues 92-104 of human ErbB3 (SEQ ID NO: 11).
- the antibody competes with Antibody A for binding to human ErbB3 and has at least 90% variable region amino acid sequence identity with the above-mentioned anti-ErbB3 antibodies.
- Small molecule TKIs useful in the methods disclosed herein include erlotinib, gefitinib, vandetanib, lapatinib, afatinib and neratinib.
- compositions and methods for treating non-small-cell lung cancer (NSCLC) in a human patient comprising
- composition in combination with erlotinib, the composition
- an anti-ErbB3 antibody comprising CDRH1 , CDRH2, and CDRH3
- CDRL1 amino acid sequences set forth in SEQ ID NO: 5 (CDRH1) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and CDRLl , CDRL2, and
- CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8
- CDRLl SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3) for treatment of the patient in combination with erlotinib according to one or more of the following Dose Schedules 1 , 2, 3, 3a, 4a, 4b, or 4c of Table 1.
- the anti-ErbB3 antibody is Antibody A.
- the dose schedule is Dose Schedule 1 , wherein the antibody is administered (or is for administration) intravenously at a dose of 6 mg/kg weekly with erlotinib administered orally at a dose of 100 mg per day.
- the dose schedule is Dose Schedule 3, wherein the antibody is administered (or is for administration) intravenously at a dose of 12 mg/kg weekly with erlotinib administered orally at a dose of 150 mg per day.
- the dose schedule is Dose Schedule 3a, wherein the antibody is administered (or is for administration) intravenously at a dose of 12 mg/kg weekly with erlotinib administered orally at a dose of 100 mg per day.
- the dose schedule is Dose Schedule 4a, wherein the antibody is administered (or is for administration) intravenously at a dose of 20 mg/kg weekly with erlotinib administered orally at a dose of 100 or 150 mg per day.
- the dose schedule is Dose Schedule 4b, wherein the antibody is administered (or is for administration) intravenously at a dose of 20 mg/kg every other week with erlotinib administered orally at a dose of 100 or 150 mg per day.
- the dose schedule is Dose Schedule 4c, wherein the antibody is administered (or is for administration) intravenously at a dose of 20 mg/kg every third week with erlotinib administered orally at a dose of 100 or 150 mg per day.
- methods of treating non-small-cell lung cancer (NSCLC) in a human patient comprising: administering combination therapy to the patient comprising an anti-ErbB3 antibody comprising CDRHl , CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and CDRLl , CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRLl) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3), in combination with gefitinib, according to one or more of the following Dose Schedules 5, 6, 7, 7a, 8a, 8b, or 8c of Table 2.
- the anti-ErbB3 antibody is Antibody A.
- methods of treating non-small-cell lung cancer (NSCLC) in a human patient comprising: administering combination therapy to the patient comprising an anti-ErbB3 antibody comprising CDRHl, CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and CDRLl , CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRLl ) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3), in combination with vandetanib, according to one or more of the following Dose Schedules 9, 10, 1 1, 1 1a, 12a, 12b, or 12c of Table 3.
- the anti-ErbB3 antibody is Antibody A.
- methods of treating non-small-cell lung cancer (NSCLC) in a human patient comprising: administering combination therapy to the patient comprising an anti-ErbB3 antibody comprising CDRHl, CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and CDRLl , CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRLl ) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3), in combination with neratinib, according to one or more of the following Dose Schedules 13, 14, 15, 15a, 16a, 16b, or 16c of Table 4.
- the anti-ErbB3 antibody is Antibody A.
- methods of treating non-small-cell lung cancer (NSCLC) in a human patient comprising: administering combination therapy to the patient comprising an anti-ErbB3 antibody comprising CDRHl , CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and CDRLl , CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRLl ) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3), in combination with lapatinib, according to one or more of the following Dose Schedules 17, 18, 19, 19a, 20a, 20b, or 20c of Table 5.
- the anti-ErbB3 antibody is Antibody A.
- the patient i.e., a human subject
- the patient has locally advanced or metastatic NSCLC.
- the patient has a NSCLC tumor in which the EGFR tyrosine kinase domain is wild-type, and the subject has previously undergone at least one chemotherapy-containing regimen.
- the patient has a NSCLC tumor that has demonstrated acquired resistance to EGFR TKIs.
- the patient has a NSCLC tumor in which the EGFR tyrosine kinase domain comprises an activating mutation, and the subject has not received any prior EGFR TKI therapy.
- the patient has NSCLC that is characterized as having a mutation in the tyrosine kinase domain of EGFR that sensitizes the NSCLC to TKI treatment.
- the patient has NSCLC that is characterized as having a mutation in the tyrosine kinase domain of EGFR (SEQ ID NO: 15) that is an exon 19 deletion of fewer than 10 contiguous amino acids comprising the deletion of amino acids R748 and E749 of the tyrosine kinase domain of EGFR (SEQ ID NO: 15).
- Patients can be tested or selected for one or more of the above described clinical attributes prior to, during or after treatment.
- methods of selecting therapy for a patient having NSCLC comprising: (a) determining whether the patient's cancer comprises an activating mutation of the EGFR tyrosine kinase domain; and (b) if so, administering to the patient an effective amount of (1 ) an anti-ErbB3 antibody comprising CDRHl , CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and CDRL1 , CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRL1) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3) and (2) erlotinib.
- an anti-ErbB3 antibody comprising CDRHl , CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDR
- the activating mutation of the EGFR tyrosine kinase domain is a deletion of residues E746-A750 of the EGFR tyrosine kinase domain (SEQ ID NO: 15).
- the anti-ErbB3 antibody is Antibody A.
- compositions for use in the treatment of a human patient diagnosed with NSCLC (non-small-cell lung cancer) in combination with treatment with a TKI e.g., erlotinib, gefitinib, vandetanib or neratinib
- the compositions comprising an anti-ErbB3 antibody comprising CDRHl, CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3)
- CDRL1 , CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRL1) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3)
- the anti-ErbB3 antibody is Antibody A.
- compositions for use in the treatment of a human patient diagnosed with NSCLC in combination with treatment with a TKI comprising an anti- ErbB3 antibody comprising CDRHl , CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and CDRL1 , CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRL1) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3), formulated for intravenous administration at a dose of 12 mg/kg.
- the anti-ErbB3 antibody is Antibody A.
- compositions for use in the treatment (e.g., effective treatment) of a human patient diagnosed with NSCLC in combination with treatment with a TKI e.g., erlotinib, gefitinib, vandetanib or neratinib
- said compositions comprising an anti-ErbB3 antibody comprising CDRHl, CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRHl) SEQ ID NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and CDRLl, CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRLl) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3), formulated for intravenous administration at a dose of 20 mg/kg.
- a TKI e.g., erlotinib, gefitinib, vandetanib or neratin
- the antibody is for administration intravenously at a dose of 6 mg/kg weekly and the TKI is erlotinib, which is for administration orally daily at a dose of 100 mg per day, or the TKI is gefitinib, which is for administration orally daily at a dose of 125 mg per day, or with vandetanib administration orally daily at a dose of 150 mg per day, or with neratinib administration orally daily at a dose of 120 mg per day.
- the TKI is erlotinib, which is for administration orally daily at a dose of 100 mg per day
- the TKI is gefitinib, which is for administration orally daily at a dose of 125 mg per day, or with vandetanib administration orally daily at a dose of 150 mg per day, or with neratinib administration orally daily at a dose of 120 mg per day.
- the antibody is for administration intravenously at a dose of 6 mg/kg weekly in conjunction with erlotinib administration orally daily at a dose of 150 mg per day, or with gefitinib administration orally daily at a dose of 250 mg per day, or with vandetanib administration orally daily at a dose of 300 mg per day, or with neratinib administration orally daily at a dose of 240 mg per day.
- the antibody is for administration intravenously at a dose of 12 mg/kg weekly in conjunction with erlotinib administration orally daily at a dose of 150 mg per day, or with gefitinib administration orally daily at a dose of 250 mg per day, or with vandetanib administration orally daily at a dose of 300 mg per day, or with neratinib administration orally daily at a dose of 240 mg per day.
- Figure 1 shows a schematic diagram of the phase 2 clinical trial.
- Figure 2 provides data from phase 1 clinical trial employing the combination of Antibody A and erlotinib.
- Cohort 1 includes patients treated with 6 mg/kg of Antibody A and erlotinib 100 mg/day.
- Cohort 2 includes patients treated with 6 mg/kg of Antibody A and erlotinib 150 mg/day.
- Cohort 3 includes patients treated with 12 mg/kg of Antibody A and erlotinib 150 mg day.
- Cohort 3A includes patients treated with 12 mg/kg of Antibody A and erlotinib 100 mg/day.
- Cohort 4A includes patients treated with 20 mg/kg QW of Antibody A and erlotinib 100 mg/day.
- Cohort 4B includes patients treated with 20 mg/kg QOW of Antibody A and erlotinib 100 mg/day.
- Cohort 4C includes patients treated with 20 mg/kg Q3W of Antibody A and erlotinib 100 mg/day. The heading of the right-hand column reads "Reason for Study Discontinuation.”
- panel A shows the Phase 1 3+3 design which evaluated patients for toxicity, with escalating doses of Antibody A and erlotinib until maximum tolerated dose or maximum target dose was identified.
- Panel B shows history information on the 33 patients in the study.
- Cohort 1 6 mg/kg Antibody A, 100 mg erlotinib.
- Cohort 2 6 mg/kg Antibody A, 150 mg erlotinib.
- Cohort 3 12 mg/kg Antibody A, 150 mg erlotinib.
- Cohort 3 A 12 mg/kg Antibody A, 100 mg erlotinib.
- Cohort 4a 20 mg/kg Antibody A administered weekly, 100 mg erlotinib.
- Cohort 4b 20 mg/kg Antibody A administered every other week, 100 mg erlotinib.
- Cohort 4c 20 mg/kg Antibody A administered every 3 weeks, 100 mg erlotinib.
- FIG. 5 shows Progression Free Survival (PFS) for all patients in the Phase 1 study as well as for patients in Groups A and Groups C.
- Figure 6 provides a waterfall plot of overall survival data from the Phase I.
- Figure 7 provides a cell viability plot and demonstrates that treating cancer cells having an activating mutation of the EGFR tyrosine kinase domain with a combination of Antibody A and erlotinib has a synergistic effect on cell viability.
- Figure 8 shows the synergistic effects of Antibody A in combination with erlotinib in ACHN and HCC827 cells.
- the term "subject” or "patient” is a human cancer patient.
- effective treatment refers to treatment producing a beneficial effect, e.g., amelioration of at least one symptom of a disease or disorder.
- a beneficial effect can take the form of an improvement over baseline, i.e., an improvement over a measurement or observation made prior to initiation of therapy according to the method.
- a beneficial effect can also take the form of arresting, slowing, retarding, or stabilizing of a deleterious progression of a marker of NSCLC.
- Effective treatment may refer to alleviation of at least one symptom of non-small cell lung cancer. Such effective treatment may, e.g., reduce patient pain, reduce the size and/or number of lesions, may reduce or prevent metastasis of a tumor, and/or may slow tumor growth.
- an effective amount refers to an amount of an agent clinically proven to provide the desired biological, therapeutic, and/or prophylactic result. That result can be reduction, amelioration, palliation, lessening, delaying, and/or alleviation of one or more of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system.
- an effective amount comprises an amount sufficient to cause a tumor to shrink and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation.
- an effective amount is an amount sufficient to delay tumor development.
- an effective amount is an amount sufficient to prevent or delay tumor recurrence.
- An effective amount can be administered in one or more administrations.
- the effective amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and may stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and may stop) tumor metastasis; (v) inhibit tumor growth; (vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer.
- an "effective amount" for therapeutic uses is the amount of Antibody A and the amount of erlotinib, gefitinib, neratinib, lapatinib or vandetanib required to provide a clinically significant decrease in NSCLC or slowing of progression of the NSCLC.
- antibody includes antibodies and antibody variants comprising at least one antibody-derived antigen binding site (e.g., VH/VL region or Fv) that specifically binds to ErbB3.
- Antibodies include known forms of antibodies.
- the antibody can be a human antibody, a humanized antibody, a bispecific antibody, or a chimeric antibody.
- the antibody also can be a Fab, Fab'2, ScFv, SMTP, AFFIBODY®, nanobody, or a domain antibody.
- the antibody also can be of any of the following isotypes: IgGl, IgG2, IgG3, IgG4, IgM, IgAl , IgA2, IgAsec, IgD, and IgE.
- antibody variant includes naturally occurring antibodies which have been altered (e.g., by mutation, deletion, substitution, conjugation to a non- antibody moiety) to include at least one variant amino acid which changes a property of the antibody. For example, numerous such alterations are known in the art which affect, e.g., half-life, effector function, and/or immune responses to the antibody in a subject.
- antibody variant also includes artificial polypeptide constructs which comprise at least one antibody-derived antigen binding site.
- TKI tyrosine kinase inhibitor
- TKI tyrosine kinase inhibitor
- Examples of TKIs include erlotinib, gefitinib, imatinib, lapatinib, neratinib, nilotinib, sunitinib, and vandetanib.
- Useful anti-ErbB3 antibodies can be made using methods well known in the art.
- art recognized anti-ErbB3 antibodies can be used.
- Ab#3, Ab #14, Ab #17, Ab # 19, described in U.S. 7,846,440 can be used.
- Antibodies that compete with any of these antibodies for binding to ErbB3 also can be used.
- anti-ErbB3 antibodies which can be used include those disclosed in US 7,285,649; US20200310557; US20100255010, as well as antibodies IB4C3 and 2D1D12 (U3 Pharma Ag), both of which are described in e.g., US2004/0197332; anti-ErbB3 antibody referred to as AMG888 (U3- 1287 - U3 Pharma Ag and Amgen); and monoclonal antibody 8B8, described in US 5,968,51 1.
- anti-ErbB3 antibodies are disclosed in the art in the context of a bispecific antibody (see e.g., B2B3-1 or B2B3-2 in WO/2009/126920 and those described in US 7,846,440 and US 2010/0266584, the entire contents of which are incorporated by reference herein.
- One example of such an antibody is Antibody A having heavy and light chains comprising the amino acid sequences set forth in SEQ ED NOs 12 and 13, respectively.
- Antibody A is referred to as "Ab #6" in US 7,846,440.
- the anti-ErbB3 antibody comprises variable heavy (VH) and/or variable light (VL) regions encoded by the nucleic acid sequences set forth in SEQ ID NOs: l and 3, respectively.
- the antibody comprises VH and/or VL regions comprising the amino acid sequences set forth in SEQ ID NOs 2 and 4, respectively.
- the antibody comprises CDRH1, CDRH2, and CDRH3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 5 (CDRH1) SEQ ED NO: 6 (CDRH2) and SEQ ID NO: 7 (CDRH3), and/or CDRL1, CDRL2, and CDRL3 sequences comprising the amino acid sequences set forth in SEQ ID NO: 8 (CDRL1 ) SEQ ID NO: 9 (CDRL2) and SEQ ID NO: 10 (CDRL3).
- the antibody competes for binding with and/or binds to the same epitope on human ErbB3 as the above-mentioned antibodies.
- the epitope comprises residues 92-104 of human ErbB3 (SEQ ID NO: 11).
- the antibody binds to human ErbB3 and has at least 90% variable region sequence identity with the above-mentioned antibodies.
- the antibody is a fully human monoclonal antibody, such as an IgG2, that binds to ErbB3 and prevents the HRG and EGF-like ligand-induced phosphorylation of ErbB3.
- Anti-ErbB3 antibodies such as Antibody A
- Antibody A can be generated, e.g., in prokaryotic or eukaryotic cells, using methods well known in the art.
- the antibody is produced in a cell line capable of glycosylating proteins such as CHO cells.
- TKIs that target intracellular ErbB signaling pathways include, for example, erlotinib, gefitinib, neratinib, lapatinib or vandetanib.
- the TKI specifically targets the epidermal growth factor receptor (EGFR) tyrosine kinase, which is highly expressed and occasionally mutated in various forms of cancer.
- EGFR epidermal growth factor receptor
- Such TKIs include neratinib, afatinib, dacomitinb, vandetanib, lapatinib, erlotinib and gefitinib.
- Some TKIs binds in a reversible fashion, to the adenosine triphosphate (ATP) binding site of the receptor.
- ATP adenosine triphosphate
- two EGFR family member molecules need to associate together to form a homodimer (other than an ErbB3 ErbB3 homodimer) or a heterodimer.
- Phosphorylation may expose binding sites on the cytoplasmic domain of the receptor for binding cell proteins that, when bound to the phosphorylated receptor, initiate a signaling cascade transmitting mitogenic stimuli to the nucleus.
- the TKI acts by inhibiting phosphorylation so that signal transduction to the nucleus is reduced or stopped.
- compositions suitable for administration to a subject can be in the form of tablets, capsules, pills, lozenges, powders or granules, or solutions or dispersions in a liquid.
- compositions typically comprise a pharmaceutically acceptable earner.
- pharmaceutically acceptable means approved by a government regulatory agency or listed in the U.S. Pharmacopeia or another generally recognized pharmacopeia for use in animals, particularly in humans.
- carrier refers to a diluent, adjuvant, excipient, or vehicle with which the compound is administered.
- Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
- Liquid compositions for parenteral administration e.g., comprising an antibody
- routes of administration by injection or infusion include
- intravenous, intraperitoneal, intramuscular, intrathecal and subcutaneous are intravenous, intraperitoneal, intramuscular, intrathecal and subcutaneous.
- composition in the form of a tablet can be prepared employing pharmaceutical excipients known in the art for that purpose and conventionally used for the preparation of solid pharmaceutical compositions.
- Erlotinib (TARCEVA®) tablets for oral administration are commercially available in three dosage strengths containing erlotinib hydrochloride (27.3 mg, 109.3 mg and 163.9 mg) equivalent to 25 mg, 100 mg and 150 mg erlotinib and the following inactive ingredients: lactose monohydrate, hypromellose, hydroxypropyl cellulose, magnesium stearate, microcrystalline cellulose, sodium starch glycolate, sodium lauryl sulfate and titanium dioxide.
- the tablets also contain trace amounts of color additives for product identification, including (for 25 mg only) FD&C Yellow #6.
- Erlotinib has the formula:
- Gefitinib (IRESSA®) tablets for oral administration are commercially available in one dosage strength (250 mg) containing a tablet core comprising gefitinib, lactose monohydrate, microcrystalline cellulose, croscarmellose sodium, povidone, sodium lauryl sulfate and magnesium stearate, which core is coated with a coating comprising hypromellose, polyethylene glycol 300, titanium dioxide, red ferric oxide and yellow ferric oxide.
- Gefitinib has the formula:
- Vandetanib (CAPRELSA®) tablets for oral administration are commercially available in 100 and 300 mg dosage strength tablets containing a tablet core comprising vandetanib (calcium hydrogen phosphate dihydrate, microcrystalline cellulose, crospovidone, povidone and magnesium stearate; and a film-coat comprising
- Vandetanib has the formula:
- Lapatinib (TYKERB®) tablets for oral administration are commercially available in one dosage strength (250 mg) containing a tablet core comprising lapatinib ditosylate monohydrate, 405 mg, equivalent to 398 mg of lapatinib ditosylate or 250 mg lapatinib free base, magnesium stearate, microcrystalline cellulose, povidone, sodium starch glycolate, and an orange film-coat comprising FD&C yellow No.6/sunset yellow FCF aluminum lake, hypromellose, macrogol/PEG 400, polysorbate 80, titanium dioxide.
- a tablet core comprising lapatinib ditosylate monohydrate, 405 mg, equivalent to 398 mg of lapatinib ditosylate or 250 mg lapatinib free base, magnesium stearate, microcrystalline cellulose, povidone, sodium starch glycolate, and an orange film-coat comprising FD&C yellow No.6/sunset yellow FCF aluminum lake, hypromellose, macrogol
- Lapatinib has the formula:
- Neratinib has the formula:
- Antibody A for intravenous infusion is, supplied as a clear liquid solution in sterile, single-use vials containing 10.1 ml of Antibody A at a concentration of 25 mg/ml in 20mM histidine, 150mM sodium chloride, pH 6.5, which should be stored at 2-8°C
- the subject has histologically or cytologically confirmed NSCLC that is locally advanced or metastatic.
- the mutation status of the EGFR tyrosine kinase domain of the patient's NSCLC tumor is determined, e.g., using art recognized techniques.
- the tumor can be biopsied and the mutation status of the EGFR tyrosine kinase domain determined using PCR with known primers or hybridization with known probes.
- the mutation status of the EGFR tyrosine kinase domain is wild-type, i.e., not-mutated.
- the patient's cancer has recurred or progressed following at least one chemotherapy-containing regimen in the metastatic setting that is considered standard of care for NSCLC.
- the patient is tested or selected, prior to treatment, for one or more of the above characteristics.
- the subject can be tested for histologically or cytologically confirmed locally advanced or metastatic NSCLC; a mutation status of the EGFR tyrosine kinase domain of the tumor that is wild-type; and/or cancer that has recurred or progressed following at least one chemotherapy-containing regimen in the metastatic setting that is considered standard of care for NSCLC.
- the patient is tested or selected as having a tumor characterized as EGFR wild-type after receiving one or more prior regimen(s) of standard chemotherapy.
- the patient is tested or selected as having a tumor with a known activating mutation of the EGFR tyrosine kinase domain.
- the patient is tested or selected as having not received any prior EGFR TKI therapy in the metastatic setting for NSCLC.
- the patient is tested or selected as having a tumor with a known activating mutation of the EGFR tyrosine kinase domain, and having not received any prior EGFR TKI therapy in the metastatic setting for NSCLC.
- the patient is tested or selected as having a NSCLC that has demonstrated acquired resistance to EGFR TKIs such as erlotinib or gefitinib.
- Drug sensitivity can be measured using techniques well known in the art. For example, drug sensitivity can be determined by measuring the response of cells derived from the tumor to the drug. In another embodiment, drug sensitivity/resistance can be
- an EGFR mutation or another mutation known to be associated with drug sensitivity/resistance e.g., G719X, exon 19 deletion, L858R, L861Q, T790M, L747S, D761Y, amplification of the MET receptor and activation of IGFR signaling, or a K-RAS mutation.
- the patient has previously received EGFR TKI therapy and had a response to treatment, but has subsequently progressed and become resistant to this therapy.
- the patient has NSCLC that is characterized as having a mutation in the tyrosine kinase domain of EGFR that sensitizes the NSCLC to TKI treatment.
- the patient has NSCLC that is characterized as having a mutation in the tyrosine kinase domain of EGFR (SEQ ID NO: 15) that is an exon 19 deletion of fewer than 10 contiguous amino acids comprising the deletion of amino acids R748 and E749 of the tyrosine kinase domain of EGFR (SEQ ID NO: 15).
- the patient meets one or more of the following clinical criteria: 1) no history of smoking; 2) female gender; 3) adenocarcinoma subtype of NSCLC or its bronchoalveolar variant, 4) Asian ethnicity; and/or 5) one EGFR mutation selected from the group consisting of: tyrosine kinase domain mutations and truncating mutations involving exons 2 to 7.
- anti-ErbB3 antibodies are administered adjunctively with a TKI, e.g., erlotinib or gefitinib, to effect improvement in subjects having NSCLC.
- the anti-ErbB3 antibody is Antibody A and the TKI is erlotinib.
- the NSCLC is characterized as having an activating mutation in the tyrosine kinase domain of EGFR (SEQ ID NO: 15).
- the mutation sensitizes the NSCLC to TKI treatment.
- the EGFR mutation results in a deletion of amino acids in exon 19 of the EGFR gene.
- the mutation is an E746-A750 deletion of the tyrosine kinase domain of EGFR (i.e., residues 743-750 of SEQ ID NO: 15).
- the mutation is an L747-T751 deletion, an L747-P753 deletion, an L747-E749 deletion, an E746-A750 deletion, an E746-T751 deletion or an exon 19 deletion of fewer than 10 contiguous amino acids comprising the deletion of amino acids R748 and E749 of the tyrosine kinase domain of EGFR (SEQ ID NO: 15).
- the NSCLC has a point mutation in exon 18, such as a G719A mutation, a G719C mutation or a G719S mutation.
- the NSCLC has a point mutation in exon 21, such as an L858R mutation, an L858M mutation, or an L861Q mutation.
- the EGFR gene does not comprise a mutation conferring resistance to TKI treatment such as an L747S mutation, a D761Y mutation, a T790M mutation, a D770-N771insNPG insertion mutation, a D770- N771insSVD insertion mutation, an A767-V769dusp ASV insertion mutation, or another such mutation in exon 20 of the EGFR gene.
- a mutation conferring resistance to TKI treatment such as an L747S mutation, a D761Y mutation, a T790M mutation, a D770-N771insNPG insertion mutation, a D770- N771insSVD insertion mutation, an A767-V769dusp ASV insertion mutation, or another such mutation in exon 20 of the EGFR gene.
- the full length EGFR sequence is shown in SEQ ID NO: 14.
- adjunctive or combined administration includes simultaneous administration of the compounds in the same or different dosage form, or separate administration of the compounds (e.g., sequential administration).
- the antibody can be simultaneously administered with the TKI, wherein both the antibody and TKI are formulated together.
- the antibody can be administered in combination with the TKI, wherein both the antibody and TKI are formulated for separate administration and are administered concurrently or sequentially.
- the antibody can be administered first followed by the administration of the TKI, or vice versa.
- the TKI is formulated for oral administration.
- the TKI is erlotinib, and is administered at a dose selected from 150 mg/day, 100 mg/day, 80 mg/day and/or 50 mg/day.
- the TKI is administered at its maximum tolerated dose.
- the dose of TKI may be varied over time. For example, the TKI may be initially administered at a relatively high dose and may be lowered over time. In another embodiment, the TKI may be initially administered at a relatively low dose and may be increased over time.
- ahti-ErbB3 antibody is formulated for intravenous administration.
- the anti-ErbB3 antibody is administered at a dose selected from: of 40 mg/kg, 20 mg/kg, 12 mg/kg, 10 mg/kg, 6 mg/kg, and/or 3.2 mg/kg.
- the dose of antibody may be varied over time. For example, the antibody may be initially administered at a high dose and may be lowered over time. In another embodiment, the antibody may be initially administered at a low dose and may be increased over time. For example, the antibody may be initially administered at a high dose and may be lowered over time. In another embodiment, the antibody may be initially administered at a low dose and may be increased over time. In one embodiment, a dose of 3.2, 6, 10, 12, 15, 20, or 40 mg/kg of Antibody Aantibody is administered.
- Suitable treatment protocols include, for example, those wherein (A) the TKI is administered to a patient (i.e., human subject) daily, and (B) the anti-ErbB3 antibody is administered to the patient once per week, every other week, or every three weeks.
- erlotinib is administered in combination with an amount of Antibody A at an interval measured in days. Suitable daily dosages of erlotinib include, for example, 100, 125 or 150 mg/day.
- the method comprises coadministering to the patient a dose of Antibody A, followed at least one seven day interval by at least one further administration of a dose of Antibody A.
- four doses of Antibody A are administered four times in a 4-week cycle, i.e., one dose is administered per week.
- two doses of Antibody A are administered in each 4 week cycle, i.e., one dose is administered every other week.
- one dose of Antibody A is administered every three weeks. In one
- the administration cycle is repeated, as necessary.
- the amount of Antibody A administered is constant for each dose. In another embodiment, the amount of antibody administered varies with each dose. For example, the maintenance (or follow-on) dose of the antibody can be higher or the same as the loading dose which is first administered. In another embodiment, the maintenance dose of the antibody can be lower or the same as the loading dose. Exemplary doses include 3.2, 6, 10, 15, 20, and 40mg/kg.
- the subject is treated with a combination of Antibody A and erlotinib according to the dosages set forth in the Table 6 below.
- Antibody A is administered at 6 mg/kg once per week, once per two weeks or once per 3 weeks in combination with daily administration of 100, 125, or 150 mg of erlotinib. In other embodiments, Antibody A is administered at 12 mg/kg once per week, once per two weeks or once per 3 weeks, in combination with daily administration of 100, 125, or 150 mg of erlotinib. In still other embodiments, Antibody A is administered at 20 mg/kg once per week, once per two weeks or once per 3 weeks in combination with daily administration of 100, 125, or 150 mg of erlotinib.
- the subject experiences tumor shrinking and/or decrease in growth rate, i.e., suppression of tumor growth.
- unwanted cell proliferation is reduced or inhibited.
- one or more of the following can occur: the number of cancer cells can be reduced; tumor size can be reduced; cancer cell infiltration into peripheral organs can be inhibited, retarded, slowed, or stopped; tumor metastasis can be slowed or inhibited; tumor growth can be inhibited; recurrence of tumor can be prevented or delayed; one or more of the symptoms associated with cancer can be relieved to some extent.
- such improvement is measured by a reduction in the quantity and/or size of measurable tumor lesions.
- Measurable lesions are defined as those that can be accurately measured in at least one dimension (longest diameter is to be recorded) as >10 mm by CT scan (CT scan slice thickness no greater than 5 mm), 10 mm caliper measurement by clinical exam or >20 mm by chest X-ray.
- CT scan CT scan slice thickness no greater than 5 mm
- 10 mm caliper measurement by clinical exam >20 mm by chest X-ray.
- the size of non-target lesions e.g.,
- pathological lymph nodes can also be measured for improvement.
- lesions can be measured on chest x-rays or CT or MRI films.
- cytology or histology can be used to evaluate responsiveness to a therapy.
- the cytological confirmation of the neoplastic origin of any effusion that appears or worsens during treatment when the measurable tumor has met criteria for response or stable disease can be considered to differentiate between response or stable disease (an effusion may be a side effect of the treatment) and progressive disease.
- Exemplary therapeutic responses to therapy may include:
- Partial Response At least a 30% decrease in the sum of dimensions of target lesions, taking as reference the baseline sum diameters; Stable Disease (SD): Neither sufficient shrinkage to qualify for partial response, nor sufficient increase to qualify for progressive disease, taking as reference the smallest sum diameters while on study; or
- CR Complete Response
- Non-CR/Non-PD refers to a response presenting a persistence of one or more non-target lesion(s) and/or maintenance of tumor marker level above the normal limits.
- Progressive Disease refers to a response presenting at least a 20% increase in the sum of dimensions of target lesions, taking as reference the smallest sum on study (this includes the baseline sum if that is the smallest on study). In addition to the relative increase of 20%, the sum must also demonstrate an absolute increase of 5 mm. The appearance of one or more new lesions is also considered progression.
- an effective amount of the compositions provided herein produce at least one therapeutic effect selected from the group consisting of reduction in size of a lung tumor, reduction in metastasis, complete remission, partial remission, stable disease, increase in overall response rate, or a pathologic complete response.
- the improvement of clinical benefit rate is about 20% 20%, 30%, 40%, 50%, 60%, 70%, 80% or more.
- kits that include a pharmaceutical composition containing an anti- ErbB3 antibody, such as Antibody A, and a pharmaceutically-acceptable carrier, in a therapeutically effective amount adapted for use in the preceding methods.
- the kits can optionally also include instructions, e.g., comprising administration schedules, to allow a practitioner (e.g., a physician, nurse, or patient) to administer the composition contained therein to administer the composition to a subject having cancer, e.g., NSCLC.
- the kit further comprises a TKI.
- the kit includes a syringe.
- kits include multiple packages of the single-dose pharmaceutical composition(s) containing an effective amount of the antibody (e.g., Antibody A) for a single administration in accordance with the methods provided above.
- instruments or devices necessary for administering the pharmaceutical composition(s) may be included in the kits.
- a kit may provide one or more pre-filled syringes containing an amount of Antibody A that is about 100 times the dose in mg/kg indicated for administration in the above methods.
- the kit may further comprise a TKI, e.g., erlotinib or gefitinib in a desired unit dosage form (e.g., a unit dosage form distributed by the manufacturer of the TKI) for administration.
- the activating mutation of the EGFR tyrosine kinase domain is a deletion of residues E746-A750 of the EGFR tyrosine kinase domain (SEQ ID NO: 15).
- the anti-ErbB3 antibody is Antibody A.
- the primary objective in Phase 1 is to determine the safety of the Antibody A + erlotinib combination and to determine the recommended Phase 2 doses of the Antibody A + erlotinib combination.
- Antibody A is administered as a one hour intravenous infusion either once per week, every other week or every three weeks, depending on the cohort assignment. Erlotinib is administered orally once per day.
- the Antibody A dose is escalated in this study only if the ongoing study of single-agent Antibody A has shown the planned Antibody A dose to be safe and if the DLT (dose-limiting toxicity) evaluation period is successfully cleared.
- An initial protocol for Phase 1 utilized single-patient cohorts for Cohort 1.
- the protocol was revised to version 2.1 per which the Phase 1 portion of this study employed a standard 3 + 3 design.
- the protocol was revised to the current version to incorporate additional dosing cohorts, alternate dosing schedule, and a modified 3+3 design, according to which 6 patients are enrolled in each of the cohorts 4a, 4b and 4c.
- Cohorts 4b and 4c may be enrolled in parallel.
- Doses of the Antibody A + erlotinib combination in this study will escalate until either the maximum tolerated dose (MTD) is identified or the combination is shown to be tolerable at the highest planned doses of Antibody A and erlotinib.
- MTD maximum tolerated dose
- Antibody A Four doses of Antibody A are administered in each 4-week cycle in cohorts 1 to 4a.
- cohort 4b two doses of Antibody A are administered in each 4 week cycle (dosing is every other week) and in cohort 4c, 1 dose of Antibody A is administered every 3 weeks (cycle duration is 3 weeks).
- the safety assessment period for purposes of DLT evaluation, and dose escalation decisions is 4 weeks after the first dose. If no DLT is observed in 3 patients in a cohort, the next subject may be dosed at the next consecutive dose level.
- the DLT evaluation period is 4 weeks after the first dose for each of the first three patients in cohort 4b and 4c.
- Safety will continue to be assessed in patients 4 through 6 of each cohort, however, the intention of enrolling these additional three patient per cohort is to collect additional PK data to make a determination regarding dosing schedule for Phase 2.
- Patients in cohorts 4b and 4c are randomly assigned to a cohort at the time of enrollment to minimize bias between these concurrently enrolling cohorts.
- Rate of Enrollment During Phase 1 enrollment can proceed to the next cohort after the dose limiting toxicity (DLT) evaluation period has elapsed and the safety data is reviewed.
- DLT dose limiting toxicity
- cohorts 4b and 4c may be enrolled in parallel and patients are randomly assigned between these cohorts to allow unbiased evaluation of both the dosing schedules.
- Enrollment in Phase 2 is opened as soon as the last subject in the final cohort completes the DLT evaluation period and a Phase 2 dose is determined.
- Subjects are treated until disease progression. Subjects are re-assessed for evidence of disease progression in accordance with current RECIST criteria [56
- 'Dose level 1 is a single-subject cohort in which the cohort is only expanded to 6 if the one subject experiences a DLT.
- the subject may elect to receive the higher dose of erlotinib ( 150 mg).
- Cohort 4b and 4c are enrolled in parallel and patients are randomly assigned between the two cohorts to allow unbiased evaluation of both the dosing schedules.
- the erlotinib dose may be increased, in increments of 25 mg daily per week, up to 150 mg.
- Cohorts utilizing Antibody A doses > 20 mg/kg or alternate dosing schedules may be evaluated upon successful completion of the Cohort 4 DLT evaluation period.
- Each subject will complete a four week DLT evaluation period prior to evaluating dose escalation. In the event a subject experiences a DLT and the cohort expands to 6 subjects, all 6 subjects will complete the 4 week evaluation period. In the event that 2 or more subjects experience a DLT in either of the cohorts, no further dose escalation will take place.
- Cohorts 4b and 4c are enrolled as expanded cohorts of 6 subjects each, and in parallel. Subjects are randomly assigned between these cohorts 4b and 4c to allow unbiased evaluation of the dosing schedules.
- the DLT evaluation period is 4 weeks after the first dose for each of the first three patients in cohort 4b and 4c.
- all 6 patients must complete the 4 week DLT evaluation period in order to determine the dose level to be safe.
- the dose will escalate to the next level only after the safety data have been evaluated at the current dose level for all subjects enrolled at the dose level and the criteria for MTD have not been met.
- any drug-related toxicities of Grade 3 or higher that arise after the 4 week DLT evaluation period are assessed for their potential relationship to cumulative Antibody A+erlotinib dose and considered in the decision to escalate dose.
- the Antibody A dose is escalated only if the ongoing study of single-agent Antibody A has shown the Antibody A dose to be safe in >3 subjects.
- any drug-related Grade 3-4 hematologic or non-hematologic toxicity including Grade 3-4 infusion reactions related to Antibody A are considered dose limiting.
- Nausea, vomiting and diarrhea are dose-limiting when Grade 3-4 toxicity occurs despite use of standard anti-emetic or anti-diarrheal agents.
- Grade 3 rash is considered dose limiting only if it lasts longer than 14 days despite optimal rash management.
- inability to deliver at least three of the planned doses over the first cycle of treatment due to drug-related toxicities is considered a dose- limiting effect.
- a subject who experiences a drug related dose-limiting toxicity may not receive additional doses of Antibody A and is removed from the study. Such a subject may continue on study at the next lower dose level if the consensus judgment is that continued treatment is in the subject's best interest.
- Subjects should have recovered from toxicity to baseline or Grade 1 (except alopecia) prior to re-treatment. For DLTs due to low hemoglobin, subjects' hemoglobin concentrations should have returned to their baseline grade before re-treatment. FDA is notified if there are any instances of retreatment following a DLT.
- the Maximum Tolerated Dose is defined as the highest dose level of both Antibody A and erlotinib in which a DLT is experienced by fewer than two subjects in a cohort of 3 - 6 subjects. When a DLT is observed in at least two subjects in a cohort of 3 - 6 subjects, the MTD is determined to have been exceeded and additional subjects (up to a total of six) may be treated at the next lower dose level.
- the recommended Phase 2 dose of the Antibody A and erlotinib combination is 20 mg/kg of Antibody A and 100 mg of erlotinib; single agent erlotinib dose is 150 mg daily.
- escalation may continue per inclusion criteria in subjects who at baseline have ⁇ 2 x ULN (upper limit of normal) in these enzyme levels (i.e. separate MTDs may be defined for populations with elevated liver enzyme levels due to liver metastases). In this case, if two different MTD's are defined for the two groups, Phase 2 will proceed with the higher MTD in subjects with baseline AST, SLT or Alkaline Phosphatase of ⁇ 2x ULN.
- Phase 1 approximately 25 to 37 subjects are enrolled, depending on the number of expansions and additional cohorts required. In Phase 2, approximately 229 evaluable subjects are enrolled.
- Group A includes subjects with NSCLC that have received at least one chemotherapy-containing regiment that is considered standard of care for NSCLC and a tumor with a mutation status of the EGFR tyrosine kinase domain that is wild-type.
- chemotherapy regimens that also contain biologic agents such as bevacizumab or cetuximab.
- Group B includes subjects that have not received any prior EGFR TKI therapy for their cancer in the metastatic setting for NSCLC; however, such subjects must have a known activating mutation of the EGFR tyrosine kinase domain.
- Group C includes subjects that have previously received EGFR TKI therapy and had a response to treatment but have subsequently progressed and become resistant to this therapy. Such subjects must meet the first 2 following criteria and one additional criterion (3a or 3b) as adapted from Jackman, et al. [Jackman D, Pao W, Riely GJ, et al. Clinical Definition of Acquired Resistance to Epidermal Growth Factor Receptor TKIs in Non-Small-Cell Lung Cancer. J Clin Oncol, 2009. (Epub ahead of print) doi:
- EGFR kinase inhibitor e.g. gefitinib or erlotinib
- a tumor that harbors an EGFR mutation known to be associated with drug sensitivity i.e., G719X, exon 19 deletion, L858R, L861Q
- OR b.
- the subject's tumor is characterized as EGFR wild-type or EGFR unknown status; however, the subject has documented response to a prior EGFR kinase inhibitor therapy lasting at least 12 weeks.
- EGFR wild type or mutation status
- the classification of EGFR is based upon information available to the investigative sites at the time of enrollment. However, samples obtained at baseline is subsequently evaluated to confirm the EGFR status. Any discrepancies are reconciled at the time of analysis.
- Group A The subject must have a tumor with a mutation status of the EGFR tyrosine kinase domain that is wild-type.
- the subject's cancer have recurred or progressed following at least one chemotherapy-containing regimen that is considered standard of care for NSCLC. This would include chemotherapy regimens that also contain biologic agents such as bevacizumab or cetuximab; OR,
- Group B The subject must not have received any prior EGFR TKI therapy for NSCLC and have a tumor that has a known activating mutation of the EGFR tyrosine kinase (TKI) domain; OR,
- Group C The subject's cancer must have demonstrated acquired resistance to EGFR TKI, as outlined in above. For such subjects, any number of prior therapies is permitted.
- Subjects must have histologically or cytologically confirmed locally advanced or metastatic non-small cell lung cancer
- Subjects being considered for the phase II portion of the study must have a lesion amenable to biopsy and must be willing to undergo a pre-treatment biopsy, unless both of the following conditions are met:
- Subject underwent a biopsy within 2 months prior to enrollment and has sufficient tumor tissue available;
- Subjects must be > 18 years of age. Subjects or their legal representatives must be able to understand and sign an informed consent. Subjects must have non-measurable or measurable tumor(s) for the phase 1 portion of the trial and must have measurable disease for the phase 2 portion in accordance with RECIST v 1.1. Subjects must have archived tumor samples available for analysis. Approximately 125 m of tumor sample is required (as FFPE blocks or prepared as slides). During the Phase 1 portion of the study, if the subject does not have archived tumor tissue available, they must be willing to undergo a biopsy prior to treatment initiation.
- Subjects must have an ECOG Performance Score (PS) of 0, 1 or 2 Subjects must have adequate bone marrow reserves as evidenced by:ANC > 1 ,500/ ⁇ 1 and Platelet count > 100,000/ ⁇ 1 and Hemoglobin > 9 g/dL. Subjects must have adequate hepatic function as evidenced by: Serum total bilirubin ⁇ 1.5 x ULN and AST, ALT and alkaline phosphatase ⁇ 2 x ULN ( ⁇ 5 x ULN is acceptable if liver metastases are present, and ⁇ 5 x ULN of alkaline phosphatase is acceptable if bone metastases are present).
- PS ECOG Performance Score
- Subjects must have adequate renal function as evidenced by a serum creatinine ⁇ 1.5 x ULN. Subjects must be recovered from clinically significant effects of any prior surgery, radiotherapy or other antineoplastic therapy. Up to CTCAE Grade 1 is acceptable for subjects with known peripheral neuropathy. Women of childbearing potential as well as fertile men and their partners must agree to abstain from sexual intercourse or to use an effective form of contraception during the study and for 90 days following the last dose of study drugs (an effective form of contraception is an oral contraceptive or a double barrier method).
- subjects are excluded based on one or more of: history of any second malignancy (recurrence or initial diagnosis) in the last 5 years (subjects with prior history of in-situ cancer or basal or squamous cell skin cancer are eligible); subjects with other malignancies are eligible if they have been continuously disease free for at least 5 years; subjects who are pregnant or lactating; subjects with an active infection or with an unexplained fever > 38.5°C during screening visits or on the first scheduled day of dosing (subjects with tumor fever may be enrolled); subjects with untreated and/or symptomatic CNS malignancies (primary or metastatic); subjects with CNS metastases who have undergone surgery or radiotherapy, whose disease is stable, and who have been on a stable dose or tapering of corticosteroids for at least 2 weeks prior to the first scheduled day of dosing are eligible for the trial; subjects with known hypersensitivity to any of the components of Antibody A or who have had hypersensitivity reactions to fully human monoclonal antibodies;
- Treatment may be withheld for up to 28 consecutive days to allow for recovery from toxicity, especially for patients who are benefiting from study treatment. If a patient does not recover within 28 days from a toxicity, which is unrelated to study drug, then their continuation in the study is evaluated.
- Subjects that experience toxicities that are less than dose-limiting may continue to receive daily erlotinib.
- erlotinib dosing should be modified as indicated below, per guidelines established during previous clinical trials of erlotinib [57 FDA
- Antibody A is supplied in sterile, single-use vials containing 10.1 mL of Antibody A at a concentration of 25 mg/ml in 20 mM histidine, 150 mM sodium chloride, pH 6.5.
- Antibody A appears as a colorless liquid solution and may contain a small amount of visible, white, amorphous, Antibody A particulates.
- Antibody A drug product should be stored at 2-8°C (36 to 46°F) with protection from light. Light protection is not required during infusion. Antibody A must not be frozen.
- Antibody A has been shown to be compatible with Alaris®, Paclitaxel, Lifeshield® and Kawasumi infusion sets that utilize an in-line 0.2 micron filter.
- Antibody A is administered weekly in cohorts 1 to 4a, every other week in cohort 4b and every 3 weeks in cohort 4c (+/- 2 days). Dose levels are determined by the cohort in which the subject is enrolled.
- the Antibody A dose to be given in combination with erlotinib is determined by the MTD identified in the phase I portion of the study or the target optimal dose of Antibody A as determined by pharmacokinetic findings and safety data.
- the pharmacy is provided with expiration dates for stored Antibody A. Stability is generated on a continual basis and the expiration date is continually updated via a Sponsor notification to the pharmacy or via direct printing on the MM-12 vial, as required by local regulation.
- Antibody A should be brought to room temperature prior to administration. Vials of Antibody A should not be shaken. The appropriate quantity of Antibody A is removed from the vial, diluted in 250 mLs of 0.9% normal saline and administered as an IV infusion over 90 minutes (for the first infusion) or 60 minutes (for subsequent infusions in the absence of infusion reactions) using a low protein binding 0.22 micrometer in-line filter.
- Erlotinib dosing begins the day after the first dose of Antibody A (i.e. Cycle 1 Day 2). Dose levels should be determined by the cohort in which the subject is enrolled. In the Phase II portion of the study, the erlotinib dose to be given in combination with Antibody A is determined either by the MTD identified in the phase I portion of the study or the target optimal dose of erlotinib as determined by pharmacokinetic findings and safety data. Patients randomized to receive erlotinib alone will receive erlotinib 150 mg daily.
- Erlotinib is to be taken orally (PO) at least one hour before or two hours after the ingestion of food. It should be taken at the same time each day. On days which the subject is to receive the Antibody A infusion, the subject should take the erlotinib just prior to Antibody A dose administration (e.g. within a few minutes of starting the infusion).
- Serum levels of Antibody A and erlotinib are measured at a central analytical lab using an ELISA based assay. In order to better understand the PK and safety profile of Antibody A and erlotinib combination, additional analytes may also be measured.
- Tumor samples are fixed in formalin and subsequently embedded in paraffin blocks. These samples are used to identify the subject's EGFR mutation status, evaluate potentially predictive biomarkers and complete other correlative studies. Other mutations may also be evaluated, as required. Archived paraffin blocks may be used if available. Approximately 125 ⁇ 1 of tumor sample is required for this purpose.
- tumor samples are collected from all patients through biopsies performed prior to the first dose administration and, if possible, at the time of disease progression (post treatment biopsies are optional). Tumor samples collected through these biopsies are compared with the historical samples and will also be analyzed to explore the biomarkers that could predict response to the Antibody A and erlotinib combination.
- Clinical activity was observed including 1 PR (an EGFR TKI naive EGFR mutant) and 14 SD. Average duration of disease stabilization was 21.6 wks (range 7.1 -89.3 wks). Median PFS was 7.9 weeks, and the 16 week PFS rate was 41 % in the overall population. For EGFRwt and EGFRresist patients, the median PFS was 7.6 weeks and 15 weeks, and the 16 week PFS rates were 32% and 44%, respectively. 7/20 EGFRwt patients and 5/9 EGFRresist patients achieved SD.
- OS Overall survival (OS) for all 33 patients in the phase 1 study was 9.8mos (6.5 - inf)- In EGFRwt patients, median OS was 9.8mos (6.5 - inf), while in EGFR resistant patients OS was 1 1.0 months (4.0 - inf).
- OS for erlotinib was 5.3mos (TITAN Study) and 6.3mos (BR.21 Study).
- Figure 2 provides data from phase 1 clinical trial employing the combination of Antibody A and erlotinib.
- Figure 3 shows the Phase 1 3+3 design which evaluated patients for toxicity, with escalating doses of Antibody A and erlotinib until maximum tolerated dose or maximum target dose was identified.
- Panel B provides history information on the 33 patients in the study.
- Figure 4A shows adverse events reported
- Figure 4B provides a summary of number of adverse events by dosing cohorts. These events are similar to known erlotinib toxicities with a trend towards a higher frequency, but not higher severity, when combined with Antibody A.
- Figure 5 shows Progression Free Survival (PFS) for all patients in the Phase 1 study as well as for patients in Groups A and Groups C.
- Median PFS overall was 7.9 weeks (95% CI: 7.6 - 20.1).
- Figure 6 provides a waterfall plot of overall survival data from the phase I.
- Phase 2 the trial will evaluate the combination of Antibody A and erlotinib in three different populations of approximately 229 NSCLC patients. See Figure 1. .
- Group C Subjects in which the cancer has demonstrated acquired resistance to EGFR TKI, as outlined in above. For such subjects, any number of prior therapies is permitted. All subjects in this group will receive Antibody A plus erlotinib.
- the recommended Phase 2 dose is 100 mg/day of erlotinib and
- the Antibody A dose for future dose(s) may be reduced.
- Categorical variables will be summarized by frequency distributions (number and percentages of subjects) and continuous variables will be summarized by descriptive statistics (mean, standard deviation, median, minimum, and maximum).
- Safety analyses will be performed using the Intent-to-Treat (ITT) population (all subjects who received at least one infusion of study drug). Efficacy analyses will be performed using the ITT population and the Evaluable Subject population (subjects receiving > 6 doses of Antibody A). Results from EGFR testing completed on baseline subject samples will be compared to initial enrollment classification (Group A, Group B, Group C). If any discrepancies are noted, an additional efficacy analysis will be completed to incorporate the correct classification. These results will be reported in addition to the primary efficacy analysis based on Investigator classification of subject subgroup. In addition, subanalyses will be completed to further explore key prognostic factors and the impact of the presence/absence of key biomarkers.
- Example 6 Combination treatment with Antibody A and erlotinib on cancer cells with an activating mutation of the EGFR tyrosine kinase domain
- HCC827 non-small cell lung adenocarcinoma cell line (ATCC CRL- 2868TM) have an acquired E746-A750 deletion in the tyrosine kinase domain-encoding region of the gene encoding EGFR (SEQ ID NO: 15). This mutation renders HCC827 cells responsive to erlotinib treatment.
- HCC827 cells were maintained in RP I-1640 medium (Lonza) supplemented with 10%FBS (Hyclone), 100 units/ml penicillin, and lOOmg/ml streptomycin (Gibco). Cells were seeded in 96- well black-masked tissue culture plates (1000 cells per well), grown overnight, then switched to low-serum medium (0.5% FBS) for 24 hours. To measure growth response, cells were treated with several concentrations of Antibody A, erlotinib, or an equimolar combination of Antibody A and erlotinib (erl+MM).
- Antibody A Concentrations of Antibody A were 0.0073, 3, 40, 120, 200, and 1000 nM. Cells were grown for 3 days. Then, ATP levels were measured using CellTitre-GloOCell Viability Assay (Promega) as follows: cells were lysed at room temperature with CellTitre Glo® reagent for 5 minutes on a shaker, then equilibrated for an additional 10 minutes;
- luminescent signal was measured using an Envision Plate Reader (Perkin Elmer); and raw. luminescent signal for each treatment was normalized to medium only control and plotted using Prism software (GraphPad Software, Inc.).
- Example 7 Co-inhibition of ErbB3 Ligand Activation with an anti-ErbB3 ligand- blocking antibody and EGFR Signaling with an EGFR tyrosine kinase inhibitor
- ACHN renal carcinoma cells were seeded at 1000 cells/well in 96-well culture plates, and HCC827 NSCLC cells were seeded at 2000 cells/well in 96-well culture plates. Cells were grown overnight, then switched to low serum media (0.5% FBS) for
- Antibody A an anti-ErbB3 ligand- blocking antibody
- erlotinib an EGFR tyrosine kinase inhibitor
- Antibody A + erlotinib an EGFR tyrosine kinase inhibitor
- ATP levels were measured using CellTitre Glo® (Promega Corp.) assay and then normalized to a vehicle control.
- Bliss synergy/additivity analysis was then performed. Per the Bliss analysis, the fractional response if two drugs are exactly additive was calculated by taking the product of control normalized data for each drug alone. Then the difference between the observed and calculated fractional response was divided by the calculated fractional response to get a bliss index value that is negative
- tacgaggtgt cccagaggcc cagcggcgtg agcaacaggt tcagcggcag caagagcggc aacaccgcca gcctgaccat cagcggcctg cagaccgagg acgaggccga ctactactgc tgcagctacg ccggcagcag catcttcgtg atcttcggcg gagggaccaa ggtgaccgtc eta
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Organic Chemistry (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Oncology (AREA)
- Biomedical Technology (AREA)
- Microbiology (AREA)
- Biophysics (AREA)
- Genetics & Genomics (AREA)
- Molecular Biology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Biochemistry (AREA)
- Mycology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Peptides Or Proteins (AREA)
- Enzymes And Modification Thereof (AREA)
Abstract
Description
Claims
Priority Applications (1)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
AU2012271041A AU2012271041A1 (en) | 2011-06-16 | 2012-06-07 | Dosage and administration of anti-ErbB3 antibodies in combination with tyrosine kinase inhibitors |
Applications Claiming Priority (10)
Application Number | Priority Date | Filing Date | Title |
---|---|---|---|
US201161497834P | 2011-06-16 | 2011-06-16 | |
US61/497,834 | 2011-06-16 | ||
US201161555141P | 2011-11-03 | 2011-11-03 | |
US61/555,141 | 2011-11-03 | ||
US201261596097P | 2012-02-07 | 2012-02-07 | |
US61/596,097 | 2012-02-07 | ||
US201261602365P | 2012-02-23 | 2012-02-23 | |
US61/602,365 | 2012-02-23 | ||
US201261616912P | 2012-03-28 | 2012-03-28 | |
US61/616,912 | 2012-03-28 |
Publications (1)
Publication Number | Publication Date |
---|---|
WO2012173867A1 true WO2012173867A1 (en) | 2012-12-20 |
Family
ID=47357420
Family Applications (1)
Application Number | Title | Priority Date | Filing Date |
---|---|---|---|
PCT/US2012/041339 WO2012173867A1 (en) | 2011-06-16 | 2012-06-07 | Dosage and administration of anti-erbb3 antibodies in combination with tyrosine kinase inhibitors |
Country Status (4)
Country | Link |
---|---|
AU (1) | AU2012271041A1 (en) |
TW (1) | TW201302792A (en) |
UY (1) | UY34131A (en) |
WO (1) | WO2012173867A1 (en) |
Citations (2)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20080221138A1 (en) * | 2005-11-04 | 2008-09-11 | Paul Bunn | Method of using SAHA and Erlotinib for treating cancer |
US7846440B2 (en) * | 2007-02-16 | 2010-12-07 | Merrimack Pharmaceuticals, Inc. | Antibodies against ErbB3 and uses thereof |
-
2012
- 2012-06-07 WO PCT/US2012/041339 patent/WO2012173867A1/en active Application Filing
- 2012-06-07 AU AU2012271041A patent/AU2012271041A1/en not_active Abandoned
- 2012-06-15 UY UY0001034131A patent/UY34131A/en unknown
- 2012-06-15 TW TW101121448A patent/TW201302792A/en unknown
Patent Citations (3)
Publication number | Priority date | Publication date | Assignee | Title |
---|---|---|---|---|
US20080221138A1 (en) * | 2005-11-04 | 2008-09-11 | Paul Bunn | Method of using SAHA and Erlotinib for treating cancer |
US7846440B2 (en) * | 2007-02-16 | 2010-12-07 | Merrimack Pharmaceuticals, Inc. | Antibodies against ErbB3 and uses thereof |
US20110123523A1 (en) * | 2007-02-16 | 2011-05-26 | Merrimack Pharmaceuticals, Inc. | Antibodies against erbb3 and uses thereof |
Also Published As
Publication number | Publication date |
---|---|
UY34131A (en) | 2013-01-31 |
TW201302792A (en) | 2013-01-16 |
AU2012271041A1 (en) | 2013-04-04 |
Similar Documents
Publication | Publication Date | Title |
---|---|---|
CN104271601B (en) | Methods of treating cancer using PD-1 axis binding antagonists and VEGF antagonists | |
EP3498734B1 (en) | Combination of a pd-1 antagonist and a vegfr inhibitor for treating cancer | |
US20150231238A1 (en) | Overcoming resistance to erbb pathway inhibitors | |
KR20170074962A (en) | Combination therapy for cancer | |
WO2013023043A2 (en) | Treatment of advanced solid tumors using combination of anti-erbb3 immunotherapy and selected chemotherapy | |
US20140248280A1 (en) | Anti-erbb3 antibodies in combination with paclitaxel for treatment of gynecological cancers | |
US20220241412A1 (en) | Combination therapies using cdk inhibitors | |
AU2012273361A1 (en) | Dosage and administration of anti-ErbB3 antibodies in combination with paclitaxel | |
TW202011991A (en) | Methods of treating lung cancer with a pd-1 axis binding antagonist, an antimetabolite, and a platinum agent | |
AU2012211258A1 (en) | Treatment of advanced solid stage tumors using anti-ErbB3 antibodies | |
WO2017070475A1 (en) | Methods of treating cancer by administering a mek inhibitor and a combination of anti-egfr antibodies | |
CN117715936A (en) | Methods and compositions for treating cancer | |
CN113117072A (en) | Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody | |
WO2012173867A1 (en) | Dosage and administration of anti-erbb3 antibodies in combination with tyrosine kinase inhibitors | |
CN112915202A (en) | Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody | |
CN111386117A (en) | Compositions and methods for treating cancer | |
US20220235141A1 (en) | Combination therapies using cdk inhibitors | |
CN114432438A (en) | Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody | |
CN114470191A (en) | Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody | |
CN114470190A (en) | Pharmaceutical composition of quinoline derivative and PD-1 monoclonal antibody | |
WO2017035482A1 (en) | Combination therapies for treatment of heregulin positive cancers | |
WO2017070456A1 (en) | Methods of treating cancer by administering a bispecific antibody antagonist of igf-ir and erbb3 and a combination of anti-egfr antibodies |
Legal Events
Date | Code | Title | Description |
---|---|---|---|
121 | Ep: the epo has been informed by wipo that ep was designated in this application |
Ref document number: 12801280 Country of ref document: EP Kind code of ref document: A1 |
|
ENP | Entry into the national phase |
Ref document number: 2012271041 Country of ref document: AU Date of ref document: 20120607 Kind code of ref document: A |
|
NENP | Non-entry into the national phase |
Ref country code: DE |
|
122 | Ep: pct application non-entry in european phase |
Ref document number: 12801280 Country of ref document: EP Kind code of ref document: A1 |