34 results on '"Christine Fazenbaker"'
Search Results
2. Antitumor activity of AZD0754, a dnTGFβRII-armored, STEAP2-targeted CAR-T cell therapy, in prostate cancer
- Author
-
Peter Zanvit, Dewald van Dyk, Christine Fazenbaker, Kelly McGlinchey, Weichuan Luo, Jessica M. Pezold, John Meekin, Chien-ying Chang, Rosa A. Carrasco, Shannon Breen, Crystal Sao-Fong Cheung, Ariel Endlich-Frazier, Benjamin Clark, Nina J. Chu, Alessio Vantellini, Philip L. Martin, Clare E. Hoover, Kenesha Riley, Steve M. Sweet, David Chain, Yeoun Jin Kim, Eric Tu, Nathalie Harder, Sandrina Phipps, Melissa Damschroder, Ryan N. Gilbreth, Mark Cobbold, Gordon Moody, and Emily E. Bosco
- Subjects
Oncology ,Medicine - Abstract
Prostate cancer is generally considered an immunologically “cold” tumor type that is insensitive to immunotherapy. Targeting surface antigens on tumors through cellular therapy can induce a potent antitumor immune response to “heat up” the tumor microenvironment. However, many antigens expressed on prostate tumor cells are also found on normal tissues, potentially causing on-target, off-tumor toxicities and a suboptimal therapeutic index. Our studies revealed that six-transmembrane epithelial antigen of prostate-2 (STEAP2) was a prevalent prostate cancer antigen that displayed high, homogeneous cell surface expression across all stages of disease with limited distal normal tissue expression, making it ideal for therapeutic targeting. A multifaceted lead generation approach enabled development of an armored STEAP2 chimeric antigen receptor T cell (CAR-T) therapeutic candidate, AZD0754. This CAR-T product was armored with a dominant-negative TGF-β type II receptor, bolstering its activity in the TGF-β–rich immunosuppressive environment of prostate cancer. AZD0754 demonstrated potent and specific cytotoxicity against antigen-expressing cells in vitro despite TGF-β–rich conditions. Further, AZD0754 enforced robust, dose-dependent in vivo efficacy in STEAP2-expressing cancer cell line–derived and patient-derived xenograft mouse models, and exhibited encouraging preclinical safety. Together, these data underscore the therapeutic tractability of STEAP2 in prostate cancer as well as build confidence in the specificity, potency, and tolerability of this potentially first-in-class CAR-T therapy.
- Published
- 2023
- Full Text
- View/download PDF
3. Supplemental Figure S1, Supplemental Figure S2, Supplemental Table S1, Supplemental Figure S3, Supplemental Figure S4, Supplemental Figure S5, Supplemental Figure S6 from Improved Therapeutic Window in BRCA-mutant Tumors with Antibody-linked Pyrrolobenzodiazepine Dimers with and without PARP Inhibition
- Author
-
David A. Tice, Ronald Herbst, Andrew J. Pierce, Jay Harper, Rajiv Raja, Nicholas Holoweckyj, Brandon W. Higgs, James Conway, Maureen Kennedy, Christine Fazenbaker, Jing Zhang, Shannon Breen, Ravinder Tammali, Cui Chen, and Haihong Zhong
- Abstract
Supplemental Figure S1: Knockdown of BRCA1 or BRCA2 sensitizes Hela cells to PBD payload and PBD-based ADC in vitro. Supplemental Figure S2: Genetic deletion of BRCA1 sensitizes cells to PBD-based ADC. Supplemental Table S1: BRCA mutation(s) in patient-derived xenograft models. Supplemental Figure S3: Mean tumor volume graphs of 23 BRCA-deficient PDX tumors response to PBD-ADC treatment compared to untreated group. Supplemental Figure S4: Mean tumor volume graphs of BRCA wild-type PDX tumors response to PBD-ADC treatment compared to untreated group. Supplemental Figure S5: Representative IHC images of 5T4 staining in PDX models. Supplemental Figure S6: No in vivo efficacy was observed in tumor model DMS-114 that does not express 5T4.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Data from MEDI-573, Alone or in Combination with Mammalian Target of Rapamycin Inhibitors, Targets the Insulin-like Growth Factor Pathway in Sarcomas
- Author
-
Robert E. Hollingsworth, Yihong Yao, Christopher Morehouse, Jiaqi Huang, Cui Chen, Shannon Breen, Christine Fazenbaker, and Haihong Zhong
- Abstract
Supplementary Data. Supplemental Table S1: Summary of MEDI-573 IC50 on the proliferation of sarcoma cell lines with or without exogenously added IGF-1 or IGF-2. Supplemental Figure S1: Effect of MEDI-573 in combination with AZD2014 on SJSA-1 tumor growth in vivo.
- Published
- 2023
- Full Text
- View/download PDF
5. Data from MEDI-573, Alone or in Combination with Mammalian Target of Rapamycin Inhibitors, Targets the Insulin-like Growth Factor Pathway in Sarcomas
- Author
-
Robert E. Hollingsworth, Yihong Yao, Christopher Morehouse, Jiaqi Huang, Cui Chen, Shannon Breen, Christine Fazenbaker, and Haihong Zhong
- Abstract
MEDI-573 is a human antibody that neutralizes insulin-like growth factor (IGF) I and IGFII. IGFs are overexpressed in multiple types of cancer; their overexpression is a potential mechanism for resistance to IGFI receptor (IGFIR)-targeting therapy. Effects of IGF on cell proliferation, differentiation, and survival are mediated through its binding to and activation of IGFIR or insulin receptor A (IR-A). In this study, we measured the mRNA levels of IGFI, IGFII, and IGFIR in human pediatric sarcoma xenografts, and protein levels in sarcoma cell lines. MEDI-573 potently inhibited in vitro proliferation of sarcoma cell lines, with Ewing sarcoma cell lines being the most sensitive. In addition, MEDI-573 inhibited IGFI- and IGFII-induced sarcoma cell proliferation in vitro. The effect of MEDI-573 on IGF signaling was also examined. Treatment with MEDI-573 markedly reduced levels of pIGFIR, pIR-A, and pAKT and significantly blocked IGFI- and IGFII-induced activation of the IGFIR and AKT pathways. MEDI-573 inhibited the growth of sarcoma xenografts in vivo and inhibition correlated with neutralization of IGFI and IGFII. Combination of MEDI-573 with either rapamycin or AZD2014, another mTOR inhibitor (mTORi), significantly enhanced the antitumor activity of MEDI-573, and this response correlated with modulation of AKT and mTOR signaling. In summary, sarcoma cells respond to autocrine or paracrine growth stimulation by IGFI and IGFII, and inhibition of IGFI and IGFII by MEDI-573 results in significant slowing of tumor growth rate in sarcoma models, particularly in Ewing sarcoma. These data provide evidence for the potential benefits of MEDI-573 and mTORi combinations in patients with Ewing sarcoma. Mol Cancer Ther; 13(11); 2662–73. ©2014 AACR.
- Published
- 2023
- Full Text
- View/download PDF
6. Data from Improved Therapeutic Window in BRCA-mutant Tumors with Antibody-linked Pyrrolobenzodiazepine Dimers with and without PARP Inhibition
- Author
-
David A. Tice, Ronald Herbst, Andrew J. Pierce, Jay Harper, Rajiv Raja, Nicholas Holoweckyj, Brandon W. Higgs, James Conway, Maureen Kennedy, Christine Fazenbaker, Jing Zhang, Shannon Breen, Ravinder Tammali, Cui Chen, and Haihong Zhong
- Abstract
Pyrrolobenzodiazepine dimers (PBD) form cross-links within the minor groove of DNA causing double-strand breaks (DSB). DNA repair genes such as BRCA1 and BRCA2 play important roles in homologous recombination repair of DSB. We hypothesized that PBD-based antibody–drug conjugates (ADC) will have enhanced killing of cells in which homologous recombination processes are defective by inactivation of BRCA1 or BRCA2 genes. To support this hypothesis, we found 5T4–PBD, a PBD-dimer conjugated to anti-5T4 antibody, elicited more potent antitumor activity in tumor xenografts that carry defects in DNA repair due to BRCA mutations compared with BRCA wild-type xenografts. To delineate the role of BRCA1/2 mutations in determining sensitivity to PBD, we used siRNA knockdown and isogenic BRCA1/2 knockout models to demonstrate that BRCA deficiency markedly increased cell sensitivity to PBD-based ADCs. To understand the translational potential of treating patients with BRCA deficiency using PBD-based ADCs, we conducted a “mouse clinical trial” on 23 patient-derived xenograft (PDX) models bearing mutations in BRCA1 or BRCA2. Of these PDX models, 61% to 74% had tumor stasis or regression when treated with a single dose of 0.3 mg/kg or three fractionated doses of 0.1 mg/kg of a PBD-based ADC. Furthermore, a suboptimal dose of PBD-based ADC in combination with olaparib resulted in significantly improved antitumor effects, was not associated with myelotoxicity, and was well tolerated. In conclusion, PBD-based ADC alone or in combination with a PARP inhibitor may have improved therapeutic window in patients with cancer carrying BRCA mutations.
- Published
- 2023
- Full Text
- View/download PDF
7. Abstract LB085: Antitumor activity of AZD0754, a dnTGFbRII armored STEAP2 targeted CAR-T therapy, in preclinical models of prostate cancer
- Author
-
Dewald van Dyk, Peter Zanvit, Christine Fazenbaker, Kelly McGlinchey, Weichuan Luo, Jessica Pezold, John Meekin, Chien-ying Chang, Benjamin Clark, Philip L. Martin, Clare Hoover, Eric Tu, Ryan Gilbreth, Mark Cobbold, Gordon Moody, and Emily E. Bosco
- Subjects
Cancer Research ,Oncology - Abstract
Prostate cancer is traditionally considered an immunologically “cold” tumor type rendering patients insensitive to immunotherapy. Targeting surface antigens on tumors through cellular therapy can induce a potent anti tumor immune response to “heat up” the tumor microenvironment. However, many antigens expressed on prostate tumors are also found on normal tissues, potentially causing on-target, off-tumor toxicities and a sub-optimal therapeutic index. Our target discovery and validation efforts identified STEAP2 as a superior prostate antigen for therapeutic targeting. Importantly, STEAP2 is a highly prevalent prostate cancer antigen displaying high, homogeneous cell surface expression across all stages of disease. A novel lead generation approach facilitated the development of a potent and specific armored STEAP2 CAR-T therapeutic candidate, AZD0754. This second generation CAR-T product is armored with a dominant-negative TGFβRII, thereby bolstering activity in the TGFβ-rich immunosuppressive environment of prostate cancer. Armored STEAP2 CAR-T cells demonstrate favorable in vitro properties, robust dose dependent in vivo efficacy in STEAP2 expressing cell line- and patient derived- mouse xenograft models and encouraging preclinical safety. Taken together, this data builds confidence in the specificity and potency of this potential first in class STEAP2 targeted CAR-T therapy and supports future clinical development. Citation Format: Dewald van Dyk, Peter Zanvit, Christine Fazenbaker, Kelly McGlinchey, Weichuan Luo, Jessica Pezold, John Meekin, Chien-ying Chang, Benjamin Clark, Philip L. Martin, Clare Hoover, Eric Tu, Ryan Gilbreth, Mark Cobbold, Gordon Moody, Emily E. Bosco. Antitumor activity of AZD0754, a dnTGFbRII armored STEAP2 targeted CAR-T therapy, in preclinical models of prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 2 (Clinical Trials and Late-Breaking Research); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(8_Suppl):Abstract nr LB085.
- Published
- 2023
- Full Text
- View/download PDF
8. Abstract A37: Evaluation of eDHFR/iTag PET reporter gene immunogenicity and application in GPC3 CAR T cells
- Author
-
Mark A Sellmyer, Iris K Lee, Kyle Kuszpit, Jyoti Roy, Alex Alfaro, Virginie Ory, Lily Cheng, Daniel Sutton, Emily Bosco, Christine Fazenbaker, Shabazz Novarra, Ryan Gilbreth, Nick Tschernia, Deborah Berry, Xiaoru Chen, Yuling Wu, and Ryan Wong
- Subjects
Cancer Research ,Immunology - Abstract
Genetically engineered medicines such as chimeric antigen receptor (CAR) T cells have great potential to be the next pillar of medical therapy beyond chemo- and traditional biologic therapies. To develop genetic medicines, new methods to understand their pharmacokinetics (PK) in humans are crucial. It is not feasible to perform traditional PK analysis for “living drugs”, because the genes themselves (in the form of DNA or RNA), are not typically responsible for the therapeutic effect. Rather, the protein products of the genes or the cells harboring the engineered genes are the actuators, and thus cannot be measured using standard HPLC or ligand binding immunoassays for PK analysis. We used a positron emission tomography (PET) reporter gene or “imaging tag” based on the intracellular bacterial enzyme dihydrofolate reductase (eDHFR) that can be paired with radiolabeled versions of trimethoprim (TMP). In this work, we evaluate the potential for immunogenicity using primary human cells and assays geared to assess low affinity and rare T cell clones that may react to eDHFR. We used overlapping pools of 15-mer eDHFR peptides and found that across 9 patients, there was little reactivity compared to EBV and CMV peptide controls. Further, the relative strength of reactivity to the eDHFR peptides was less than that of the viral peptides. Next, we showed that eDHFR iTag harboring CAR T cells were functionally comparable to unlabeled CAR T cells in vitro, and demonstrated strong, selective [18F]-TMP uptake in the eDHFR-expressing CAR T cells. Finally, using a glypican 3 (GPC3) CAR T rodent model, we performed a feasibility study to non-invasively track proliferation in antigen-harboring xenograft tumors over time with ex vivo correlation to anti-CD3 immunohistochemistry. These data demonstrate the potential for non-invasive monitoring of CAR T cells using PET imaging and translational applicability of DHFR/TMP radiotracers. Citation Format: Mark A Sellmyer, Iris K Lee, Kyle Kuszpit, Jyoti Roy, Alex Alfaro, Virginie Ory, Lily Cheng, Daniel Sutton, Emily Bosco, Christine Fazenbaker, Shabazz Novarra, Ryan Gilbreth, Nick Tschernia, Deborah Berry, Xiaoru Chen, Yuling Wu, Ryan Wong. Evaluation of eDHFR/iTag PET reporter gene immunogenicity and application in GPC3 CAR T cells [abstract]. In: Proceedings of the AACR Special Conference: Tumor Immunology and Immunotherapy; 2022 Oct 21-24; Boston, MA. Philadelphia (PA): AACR; Cancer Immunol Res 2022;10(12 Suppl):Abstract nr A37.
- Published
- 2022
- Full Text
- View/download PDF
9. A Reactive Antibody Platform for One-Step Production of Antibody–Drug Conjugates through a Diels–Alder Reaction with Maleimide
- Author
-
Fengying Huang, Balakumar Vijayakrishnan, Chacko Chakiath, Shenlan Mao, Daniel Lemen, Christine Fazenbaker, Marcello Marelli, Javier Read de Alaniz, Neki V. Patel, Lauren Adams, Philip Howard, Haihong Zhong, Wenshu Xu, R. James Christie, André H. St. Amant, Jay Harper, Changshou Gao, Herren Wu, and Jia Lin
- Subjects
Models, Molecular ,Immunoconjugates ,Cell Survival ,Protein Conformation ,Biomedical Engineering ,Pharmaceutical Science ,Bioengineering ,CHO Cells ,02 engineering and technology ,01 natural sciences ,Adduct ,Maleimides ,chemistry.chemical_compound ,Cricetulus ,Animals ,Humans ,Spiro Compounds ,Maleimide ,Diels–Alder reaction ,Pharmacology ,chemistry.chemical_classification ,Bioconjugation ,Cycloaddition Reaction ,010405 organic chemistry ,Organic Chemistry ,021001 nanoscience & nanotechnology ,Combinatorial chemistry ,Cycloaddition ,3. Good health ,0104 chemical sciences ,Amino acid ,chemistry ,Covalent bond ,PC-3 Cells ,0210 nano-technology ,Biotechnology ,Conjugate - Abstract
The normal electron-demand Diels-Alder (DA) cycloaddition is a classic transformation routinely used in synthesis; however, applications in biological systems are limited. Here, we report a spiro[2.4]hepta-4,6-diene-containing noncanonical amino acid (SCpHK) capable of efficient incorporation into antibodies and subsequent coupling with maleimide via a DA reaction. SCpHK was stable throughout protein expression in mammalian cells and enabled covalent attachment of maleimide drug-linkers yielding DA antibody-drug conjugates (DA-ADCs) with nearly quantitative conversion in a one-step process. The uncatalyzed DA reaction between SCpHK and maleimide in aqueous buffer was rapid (1.8-5.4 M-1 s-1), and the antibody-drug adduct was stable in rat serum for at least 1 week at 37 °C. Anti-EphA2 DA-ADCs containing AZ1508 or SG3249 maleimide drug-linkers were potent inhibitors of tumor growth in PC3 tumor models in vivo. The DA bioconjugation strategy described here represents a simple method to produce site-specific and stable ADCs with maleimide drug-linkers.
- Published
- 2019
- Full Text
- View/download PDF
10. Design and characterization of homogenous antibody-drug conjugates with a drug-to-antibody ratio of one prepared using an engineered antibody and a dual-maleimide pyrrolobenzodiazepine dimer
- Author
-
Luke Masterson, Jason White, Mary Jane Hinrichs, Ryan Fleming, Kim Rosenthal, Rakesh Dixit, Herren Wu, Changshou Gao, Christine Fazenbaker, Vanessa Muniz-Medina, Ben T Ruddle, Philip Howard, Haihong Zhong, Nazzareno Dimasi, Molly Reed, and Patrick Strout
- Subjects
Drug ,Immunoconjugates ,Receptor, ErbB-2 ,media_common.quotation_subject ,Dimer ,Immunology ,Mice, Nude ,Pyrrolobenzodiazepine ,Antineoplastic Agents ,Benzodiazepines ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Stomach Neoplasms ,Report ,Animals ,Humans ,Immunology and Allergy ,Pyrroles ,Maleimide ,030304 developmental biology ,media_common ,0303 health sciences ,biology ,Pyrrolobenzodiazepine Dimer ,Trastuzumab ,Xenograft Model Antitumor Assays ,Combinatorial chemistry ,Rats ,body regions ,chemistry ,030220 oncology & carcinogenesis ,MCF-7 Cells ,biology.protein ,Female ,Antibody ,Drug to antibody ratio ,Conjugate - Abstract
Most strategies used to prepare homogeneous site-specific antibody-drug conjugates (ADCs) result in ADCs with a drug-to-antibody ratio (DAR) of two. Here, we report a disulfide re-bridging strategy to prepare homogeneous ADCs with DAR of one using a dual-maleimide pyrrolobenzodiazepine (PBD) dimer (SG3710) and an engineered antibody (Flexmab), which has only one intrachain disulfide bridge at the hinge. We demonstrate that SG3710 efficiently re-bridge a Flexmab targeting human epidermal growth factor receptor 2 (HER2), and the resulting ADC was highly resistant to payload loss in serum and exhibited potent anti-tumor activity in a HER2-positive gastric carcinoma xenograft model. Moreover, this ADC was tolerated in rats at twice the dose compared to a site-specific ADC with DAR of two prepared using a single-maleimide PBD dimer (SG3249). Flexmab technologies, in combination with SG3710, provide a platform for generating site-specific homogenous PBD-based ADCs with DAR of one, which have improved biophysical properties and tolerability compared to conventional site-specific PBD-based ADCs with DAR of two.
- Published
- 2019
- Full Text
- View/download PDF
11. Improved Therapeutic Window in BRCA-mutant Tumors with Antibody-linked Pyrrolobenzodiazepine Dimers with and without PARP Inhibition
- Author
-
Jay Harper, Ravinder Tammali, David A. Tice, Brandon W. Higgs, Nicholas Holoweckyj, Christine Fazenbaker, Jing Zhang, James Conway, Cui Chen, Shannon Breen, Haihong Zhong, Maureen Kennedy, Andrew J. Pierce, Ronald Herbst, and Rajiv Raja
- Subjects
0301 basic medicine ,Cancer Research ,Mutation ,endocrine system diseases ,DNA repair ,Poly ADP ribose polymerase ,Pyrrolobenzodiazepine ,medicine.disease_cause ,behavioral disciplines and activities ,Olaparib ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,chemistry ,030220 oncology & carcinogenesis ,PARP inhibitor ,medicine ,Cancer research ,skin and connective tissue diseases ,Homologous recombination ,DNA - Abstract
Pyrrolobenzodiazepine dimers (PBD) form cross-links within the minor groove of DNA causing double-strand breaks (DSB). DNA repair genes such as BRCA1 and BRCA2 play important roles in homologous recombination repair of DSB. We hypothesized that PBD-based antibody–drug conjugates (ADC) will have enhanced killing of cells in which homologous recombination processes are defective by inactivation of BRCA1 or BRCA2 genes. To support this hypothesis, we found 5T4–PBD, a PBD-dimer conjugated to anti-5T4 antibody, elicited more potent antitumor activity in tumor xenografts that carry defects in DNA repair due to BRCA mutations compared with BRCA wild-type xenografts. To delineate the role of BRCA1/2 mutations in determining sensitivity to PBD, we used siRNA knockdown and isogenic BRCA1/2 knockout models to demonstrate that BRCA deficiency markedly increased cell sensitivity to PBD-based ADCs. To understand the translational potential of treating patients with BRCA deficiency using PBD-based ADCs, we conducted a “mouse clinical trial” on 23 patient-derived xenograft (PDX) models bearing mutations in BRCA1 or BRCA2. Of these PDX models, 61% to 74% had tumor stasis or regression when treated with a single dose of 0.3 mg/kg or three fractionated doses of 0.1 mg/kg of a PBD-based ADC. Furthermore, a suboptimal dose of PBD-based ADC in combination with olaparib resulted in significantly improved antitumor effects, was not associated with myelotoxicity, and was well tolerated. In conclusion, PBD-based ADC alone or in combination with a PARP inhibitor may have improved therapeutic window in patients with cancer carrying BRCA mutations.
- Published
- 2019
- Full Text
- View/download PDF
12. Abstract 1270: The novel PARP1-selective inhibitor, AZD5305, is efficacious as monotherapy and in combination with standard of care chemotherapy in the in vivo preclinical models
- Author
-
Kimberly M Cook, Joanne Wilson, James W. Yates Jwt, Aaron Smith, Christine Fazenbaker, Anna Staniszewska, Elisabetta Leo, Emily Bosco, and Andrew Pike
- Subjects
Oncology ,Cancer Research ,Chemotherapy ,medicine.medical_specialty ,business.industry ,medicine.medical_treatment ,Cancer ,medicine.disease ,Carboplatin ,Olaparib ,chemistry.chemical_compound ,Breast cancer ,Paclitaxel ,chemistry ,Tolerability ,In vivo ,Internal medicine ,Medicine ,business - Abstract
Poly (ADP-ribose) polymerase inhibitors (PARPi) have shown efficacy in homologous recombination deficient (HRD) tumours, such as those with BRCA mutations (BRCAm). In this setting PARPi treatments lead to accumulation of DNA damage and cancer cell death. PARPi currently in clinical use inhibit both PARP1 and PARP2, as well as other members of the PARP family. Here, we report for the first time in vivo profiling of AZD5305, a potent and highly selective PARP1 inhibitor and trapper, currently in Ph1 clinical trials. Dose response efficacy of AZD5305 was evaluated in the BRCA1m triple-negative breast cancer (TNBC) xenograft model MDA-MB-436. AZD5305 dosed at 0.1mg/kg QD or higher for 35 days delivered about 90% regression, compared with 83% regression caused by treatment with 100mg/kg QD olaparib. Anti-tumour effects of AZD5305 continued after cessation of treatment and complete responses were achieved which were sustained for the whole duration of the study, over 100 days after treatment withdrawal, in contrast to the olaparib-treated group where regrowth of tumours was observed from day 63 after treatment withdrawal. Investigation of the PK/PD/efficacy relationship in MDA-MB-436 showed that maximum efficacy of AZD5305 was achieved when unbound plasma concentrations were maintained above the IC95 estimated from an in vitro DLD-1 BRCA2-/- cell growth assay. Similar results were obtained in a BRCA1m patient-derived explant (PDX) model, HBCx-17. Anti-tumour efficacy of AZD5305 was also tested in the DLD-1 BRCA2-/- and wild-type (WT) isogenic xenograft models. In the DLD-1 BRCA2-/- model, AZD5305 dosed at 10mg/kg QD and 1mg/kg QD delivered 78% and 63% tumour regression, respectively. AZD5305 at 0.1mg/kg QD resulted in responses similar to those observed in the olaparib 100mg/kg QD group (40-54% tumour growth inhibition, TGI). As expected, AZD5305 and olaparib showed no anti-tumour efficacy in the DLD-1 WT tumour model. Due to improved PARP1 selectivity, AZD5305 has the potential to show improved efficacy and tolerability in combination with standard of care chemotherapy when compared to non-selective PARPi. Hence, we investigated the anti-tumour effects of AZD5305 in combination with carboplatin or paclitaxel in a BRCA1m TNBC xenograft, SUM149PT, and BRCA WT TNBC PDX model, HBCx-9. In both models, combination of AZD5305 with carboplatin was well tolerated and demonstrated clear benefit compared to each monotherapy treatment. The effects of adjusted dosing and scheduling of the combination on the anti-tumour efficacy will be presented. Citation Format: Anna D. Staniszewska, James W. Yates JWT, Andy Pike, Christine Fazenbaker, Kimberly Cook, Emily Bosco, Aaron Smith, Joanne Wilson, Elisabetta Leo. The novel PARP1-selective inhibitor, AZD5305, is efficacious as monotherapy and in combination with standard of care chemotherapy in the in vivo preclinical models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1270.
- Published
- 2021
- Full Text
- View/download PDF
13. Targeted Fcγ Receptor (FcγR)-mediated Clearance by a Biparatopic Bispecific Antibody
- Author
-
Christine Fazenbaker, G. Jonah A. Rainey, Craig N. Jenne, Tracy Chen, Srinath Kasturirangan, Alyse D. Portnoff, Xinwei Wang, Changshou Gao, Helen Zhong, Jared S. Bee, Herren Wu, Linda Xu, and Zhutian Zeng
- Subjects
0301 basic medicine ,microscopic imaging ,Kupffer Cells ,medicine.drug_class ,Immunology ,Fc receptor ,Antigen-Antibody Complex ,macrophage ,Monoclonal antibody ,Biochemistry ,Epitope ,Mice ,03 medical and health sciences ,Immune system ,Antigen ,antibody ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,Macrophage ,Receptor ,Molecular Biology ,IL-6 ,antibody engineering ,biology ,Interleukin-6 ,protein complex ,Receptors, IgG ,Antibodies, Monoclonal ,Cell Biology ,Cell biology ,HEK293 Cells ,030104 developmental biology ,Liver ,Immunoglobulin G ,biology.protein ,Antibody ,electron microscopy (EM) ,pharmacokinetics ,Protein Binding - Abstract
Soluble ligands have commonly been targeted by antibody therapeutics for cancers and other diseases. Although monoclonal antibodies targeting such ligands can block their interactions with their cognate receptors, they can also significantly increase the half-life of their ligands by FcRn-mediated antibody recycling, thereby evading ligand renal clearance and requiring increasingly high antibody doses to neutralize the increasing pool of target. To overcome this issue, we generated a bispecific/biparatopic antibody (BiSAb) that targets two different epitopes on IL-6 to block IL-6-mediated signaling. The BiSAb formed large immune complexes with IL-6 that can bind Fcγ receptors on phagocytic cells and are rapidly internalized. In addition, rapid clearance of the BiSAb·IL-6 complex was observed in mice while the parental antibodies prolonged the serum half-life of IL-6. Intravital imaging of the liver in mice confirmed that the rapid clearance of these large immune complexes was associated with Fcγ receptor-dependent binding to Kupffer cells in the liver. The approach described here provides a general strategy for therapeutic antibodies with the ability to not only neutralize but also actively drive clearance of their soluble antigens.
- Published
- 2017
- Full Text
- View/download PDF
14. Tuning the Diels-Alder Reaction for Bioconjugation to Maleimide Drug-Linkers
- Author
-
Haihong Zhong, Stelios Florinas, André H. St. Amant, Herren Wu, Javier Read de Alaniz, Daniel Lemen, Christine Fazenbaker, R. James Christie, Shenlan Mao, and Changshou Gao
- Subjects
Drug ,Immunoconjugates ,media_common.quotation_subject ,Biomedical Engineering ,Pharmaceutical Science ,Bioengineering ,Alkenes ,010402 general chemistry ,01 natural sciences ,Antibodies ,Adduct ,Maleimides ,chemistry.chemical_compound ,Drug Stability ,In vivo ,Drug conjugation ,Maleimide ,Diels–Alder reaction ,media_common ,Pharmacology ,Bioconjugation ,Cycloaddition Reaction ,010405 organic chemistry ,Organic Chemistry ,Combinatorial chemistry ,3. Good health ,0104 chemical sciences ,Cross-Linking Reagents ,chemistry ,Pharmaceutical Preparations ,Biotechnology ,Conjugate - Abstract
The thiol-maleimide linkage is widely used for antibody-drug conjugate (ADC) production; however, conjugation of maleimide-drugs could be improved by simplified procedures and reliable conjugate stability. Here, we report the evaluation of electron-rich and cyclic dienes that can be appended to antibodies and reacted with maleimide-containing drugs through the Diels-Alder (DA) reaction. Drug conjugation is fast and quantitative due to reaction acceleration in water, and the linkage is more stable in serum than in the corresponding thiol-maleimide adduct with the same drug. ADCs produced using the DA reaction (DAADCs) are effective in vitro and in vivo, demonstrating the utility of this reaction in producing effective biotherapeutics. Given the large number of commercially available maleimide compounds, this conjugation approach could be readily applied to the production of a wide range of antibody (or protein) conjugates.
- Published
- 2018
15. Improved Therapeutic Window in
- Author
-
Haihong, Zhong, Cui, Chen, Ravinder, Tammali, Shannon, Breen, Jing, Zhang, Christine, Fazenbaker, Maureen, Kennedy, James, Conway, Brandon W, Higgs, Nicholas, Holoweckyj, Rajiv, Raja, Jay, Harper, Andrew J, Pierce, Ronald, Herbst, and David A, Tice
- Subjects
BRCA2 Protein ,Immunoconjugates ,Membrane Glycoproteins ,BRCA1 Protein ,Cell Survival ,Drug Synergism ,Neoplasms, Experimental ,Poly(ADP-ribose) Polymerase Inhibitors ,Xenograft Model Antitumor Assays ,Piperazines ,Benzodiazepines ,Mice ,Antineoplastic Agents, Immunological ,Cell Line, Tumor ,Mutation ,Exome Sequencing ,Animals ,Humans ,Phthalazines ,Administration, Intravenous ,Pyrroles ,Cell Proliferation ,HeLa Cells - Abstract
Pyrrolobenzodiazepine dimers (PBD) form cross-links within the minor groove of DNA causing double-strand breaks (DSB). DNA repair genes such as
- Published
- 2018
16. Efficient Preparation of Site-Specific Antibody-Drug Conjugates Using Cysteine Insertion
- Author
-
Binyam Bezabeh, Ryan Fleming, Herren Wu, Haihong Zhong, Nazzareno Dimasi, Changshou Gao, Krista Kinneer, Christine Fazenbaker, and Ronald James Christie
- Subjects
0301 basic medicine ,Drug ,Immunoconjugates ,media_common.quotation_subject ,Pharmaceutical Science ,Mice, Nude ,Antineoplastic Agents ,Antibodies, Monoclonal, Humanized ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Cell Line, Tumor ,Drug Discovery ,Cytotoxic T cell ,Animals ,Humans ,Cysteine ,media_common ,chemistry.chemical_classification ,biology ,Mutagenesis ,Antibodies, Monoclonal ,Mammary Neoplasms, Experimental ,Trastuzumab ,Xenograft Model Antitumor Assays ,Amino acid ,body regions ,030104 developmental biology ,Monomer ,chemistry ,Biochemistry ,biology.protein ,Molecular Medicine ,Female ,Antibody ,Conjugate - Abstract
Antibody-drug conjugates (ADCs) are a class of biopharmaceuticals that combine the specificity of antibodies with the high-potency of cytotoxic drugs. Engineering cysteine residues in the antibodies using mutagenesis is a common method to prepare site-specific ADCs. With this approach, solvent accessible amino acids in the antibody have been selected for substitution with cysteine for conjugating maleimide-bearing cytotoxic drugs, resulting in homogeneous and stable site-specific ADCs. Here we describe a cysteine engineering approach based on the insertion of cysteines before and after selected sites in the antibody, which can be used for site-specific preparation of ADCs. Cysteine-inserted antibodies have expression level and monomeric content similar to the native antibodies. Conjugation to a pyrrolobenzodiazepine dimer (SG3249) resulted in comparable efficiency of site-specific conjugation between cysteine-inserted and cysteine-substituted antibodies. Cysteine-inserted ADCs were shown to have biophysical properties, FcRn, and antigen binding affinity similar to the cysteine-substituted ADCs. These ADCs were comparable for serum stability to the ADCs prepared using cysteine-mutagenesis and had selective and potent cytotoxicity against human prostate cancer cells. Two of the cysteine-inserted variants abolish binding of the resulting ADCs to FcγRs in vitro, thereby potentially preventing non-target mediated uptake of the ADCs by cells of the innate immune system that express FcγRs, which may result in mitigating off-target toxicities. A selected cysteine-inserted ADC demonstrated potent dose-dependent anti-tumor activity in a xenograph tumor mouse model of human breast adenocarcinoma expressing the oncofetal antigen 5T4.
- Published
- 2017
17. MEDI-573, Alone or in Combination with Mammalian Target of Rapamycin Inhibitors, Targets the Insulin-like Growth Factor Pathway in Sarcomas
- Author
-
Robert E. Hollingsworth, Jiaqi Huang, Christine Fazenbaker, Cui Chen, Shannon Breen, Christopher Morehouse, Yihong Yao, and Haihong Zhong
- Subjects
Cancer Research ,Morpholines ,medicine.medical_treatment ,Mice, Nude ,Antibodies, Monoclonal, Humanized ,Ligands ,Mice ,Random Allocation ,Paracrine signalling ,Insulin-like growth factor ,Somatomedins ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Phosphorylation ,Autocrine signalling ,Protein kinase B ,Cell Proliferation ,Sirolimus ,biology ,Cell growth ,TOR Serine-Threonine Kinases ,Growth factor ,Sarcoma ,medicine.disease ,Antibodies, Neutralizing ,Insulin receptor ,Pyrimidines ,Oncology ,Benzamides ,Immunology ,biology.protein ,Cancer research ,Female ,Broadly Neutralizing Antibodies ,Signal Transduction - Abstract
MEDI-573 is a human antibody that neutralizes insulin-like growth factor (IGF) I and IGFII. IGFs are overexpressed in multiple types of cancer; their overexpression is a potential mechanism for resistance to IGFI receptor (IGFIR)-targeting therapy. Effects of IGF on cell proliferation, differentiation, and survival are mediated through its binding to and activation of IGFIR or insulin receptor A (IR-A). In this study, we measured the mRNA levels of IGFI, IGFII, and IGFIR in human pediatric sarcoma xenografts, and protein levels in sarcoma cell lines. MEDI-573 potently inhibited in vitro proliferation of sarcoma cell lines, with Ewing sarcoma cell lines being the most sensitive. In addition, MEDI-573 inhibited IGFI- and IGFII-induced sarcoma cell proliferation in vitro. The effect of MEDI-573 on IGF signaling was also examined. Treatment with MEDI-573 markedly reduced levels of pIGFIR, pIR-A, and pAKT and significantly blocked IGFI- and IGFII-induced activation of the IGFIR and AKT pathways. MEDI-573 inhibited the growth of sarcoma xenografts in vivo and inhibition correlated with neutralization of IGFI and IGFII. Combination of MEDI-573 with either rapamycin or AZD2014, another mTOR inhibitor (mTORi), significantly enhanced the antitumor activity of MEDI-573, and this response correlated with modulation of AKT and mTOR signaling. In summary, sarcoma cells respond to autocrine or paracrine growth stimulation by IGFI and IGFII, and inhibition of IGFI and IGFII by MEDI-573 results in significant slowing of tumor growth rate in sarcoma models, particularly in Ewing sarcoma. These data provide evidence for the potential benefits of MEDI-573 and mTORi combinations in patients with Ewing sarcoma. Mol Cancer Ther; 13(11); 2662–73. ©2014 AACR.
- Published
- 2014
- Full Text
- View/download PDF
18. Insertion of scFv into the hinge domain of full-length IgG1 monoclonal antibody results in tetravalent bispecific molecule with robust properties
- Author
-
C. Kendall Stover, Ryan Fleming, Ching Ching Leow, Changshou Gao, Christine Fazenbaker, Karen Coffman, Susan Wilson, Haihong Zhong, Nazzareno Dimasi, Binyam Bezabeh, Nerea Gibson, Xiang-Qing Yu, and Herren Wu
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,medicine.drug_class ,Immunology ,Hinge ,Antineoplastic Agents ,Computational biology ,Monoclonal antibody ,Protein Engineering ,Domain (software engineering) ,Molecular engineering ,Angiopoietin-2 ,03 medical and health sciences ,Mice ,0302 clinical medicine ,In vivo ,Report ,Antibodies, Bispecific ,medicine ,Immunology and Allergy ,Molecule ,Animals ,Humans ,biology ,Chemistry ,Antibodies, Monoclonal ,Molecular biology ,Xenograft Model Antitumor Assays ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunoglobulin G ,biology.protein ,Antibody ,Colorectal Neoplasms ,Function (biology) ,Single-Chain Antibodies - Abstract
By simultaneous binding two disease mediators, bispecific antibodies offer the opportunity to broaden the utility of antibody-based therapies. Herein, we describe the design and characterization of Bs4Ab, an innovative and generic bispecific tetravalent antibody platform. The Bs4Ab format comprises a full-length IgG1 monoclonal antibody with a scFv inserted into the hinge domain. The Bs4Ab design demonstrates robust manufacturability as evidenced by MEDI3902, which is currently in clinical development. To further demonstrate the applicability of the Bs4Ab technology, we describe the molecular engineering, biochemical, biophysical, and in vivo characterization of a bispecific tetravalent Bs4Ab that, by simultaneously binding vascular endothelial growth factor and angiopoietin-2, inhibits their function. We also demonstrate that the Bs4Ab platform allows Fc-engineering similar to that achieved with IgG1 antibodies, such as mutations to extend half-life or modulate effector functions.
- Published
- 2016
19. EphA2 Immunoconjugate as Molecularly Targeted Chemotherapy for Ovarian Carcinoma
- Author
-
John Gooya, Mian M.K. Shahzad, Anil K. Sood, David A. Tice, Chunhua Lu, Seung Wook Kim, Hee Dong Han, Rosemarie Schmandt, Jeong Won Lee, Shenlan Mao, Robert R. Langley, Christine Fazenbaker, Robert L. Coleman, Alpa M. Nick, Dowdy Jackson, Hye Sun Kim, Lingegowda S. Mangala, and Charles N. Landen
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Immunoconjugates ,Cell Survival ,medicine.drug_class ,medicine.medical_treatment ,Blotting, Western ,Fluorescent Antibody Technique ,Antineoplastic Agents ,Apoptosis ,Monoclonal antibody ,Targeted therapy ,Mice ,Cell Line, Tumor ,Proliferating Cell Nuclear Antigen ,Ovarian carcinoma ,In Situ Nick-End Labeling ,medicine ,Animals ,Humans ,Ovarian Neoplasms ,Chemotherapy ,biology ,Receptor, EphA2 ,Antibodies, Monoclonal ,Cancer ,Articles ,medicine.disease ,Immunohistochemistry ,Xenograft Model Antitumor Assays ,Immunoconjugate ,Platelet Endothelial Cell Adhesion Molecule-1 ,Oncology ,biology.protein ,Cancer research ,Female ,Antibody ,Ovarian cancer ,Oligopeptides - Abstract
EphA2 is overexpressed in many types of human cancer but is absent or expressed at low levels in normal epithelial tissues. We investigated whether a novel immunoconjugate containing an anti-EphA2 monoclonal antibody (1C1) linked to a chemotherapeutic agent (monomethyl auristatin phenylalanine [MMAF]) through a noncleavable linker maleimidocaproyl (mc) had antitumor activity against ovarian cancer cell lines and tumor models.Specificity of 1C1-mcMMAF was examined in EphA2-positive HeyA8 and EphA2-negative SKMel28 ovarian cancer cells by antibody binding and internalization assays. Controls were phosphate-buffered saline (PBS), 1C1, or control IgG-mcMMAF. Viability and apoptosis were investigated in ovarian cancer cell lines and tumor models (10 mice per group). Antitumor activities were tested in the HeyA8-luc and SKOV3ip1 orthotopic mouse models of ovarian cancer. Endothelial cells were identified by use of immunohistochemistry and anti-CD31 antibodies. All statistical tests were two-sided.The 1C1-mcMMAF immunoconjugate specifically bound to EphA2-positive HeyA8 cells but not to EphA2-negative cells and was internalized by HeyA8 cells. Treatment with 1C1-mcMMAF decreased the viability of HeyA8-luc cells in an EphA2-specific manner. In orthotopic mouse models, treatment with 1C1-mcMMAF inhibited tumor growth by 85%-98% compared with that in control mice (eg, for weight of HeyA8 tumors, 1C1-mcMMAF = 0.05 g and control = 1.03 g; difference = 0.98 g, 95% confidence interval [CI] = 0.40 to 1.58 g; P = .001). Even in bulkier disease models with HeyA8-luc cells, 1C1-mcMMAF treatment, compared with control treatment, caused regression of established tumors and increased survival of the mice (eg, 1C1-mcMMAF vs control, mean = 60.6 days vs 29.4 days; difference = 31.2 days, 95% CI = 27.6 to 31.2 days; P = .001). The antitumor effects of 1C1-mcMMAF therapy, in SKOV3ip1 tumors, for example, were statistically significantly related to decreased proliferation (eg, 1C1-mcMMAF vs control, mean = 44.1% vs 55.8% proliferating cells; difference = 11.7%, 95% CI = 2.45% to 20.9%; P = .01) and increased apoptosis of tumor cells (eg, 1C1-mcMMAF vs control, mean = 8.6% vs 0.9% apoptotic cells; difference = 7.7%, 95% CI = 3.8% to 11.7%; P.001) and of mouse endothelial cells (eg, 1C1-mcMMAF vs control, mean 2.8% vs 0.4% apoptotic endothelial cells; difference = 2.4%, 95% CI = 1.4% to 4.6%; P = .034).The 1C1-mcMMAF immunoconjugate had antitumor activity in preclinical models of ovarian carcinoma.
- Published
- 2009
- Full Text
- View/download PDF
20. A Human Antibody–Drug Conjugate Targeting EphA2 Inhibits Tumor GrowthIn vivo
- Author
-
John Gooya, Sudha Swamynathan, Changshou Gao, Michael S. Kinch, Linda Xu, Margarita Camara, Damon L. Meyer, Steven Coats, Peter A. Kiener, Peter D. Senter, Ryan Fleming, Herren Wu, David A. Tice, Christine Fazenbaker, Krista Kinneer, Shenlan Mao, and Dowdy Jackson
- Subjects
Cancer Research ,Antibody-drug conjugate ,Immunoconjugates ,medicine.drug_class ,media_common.quotation_subject ,Biology ,Monoclonal antibody ,Mice ,chemistry.chemical_compound ,In vivo ,medicine ,Animals ,Humans ,Receptor ,Internalization ,media_common ,Receptor, EphA2 ,Antibodies, Monoclonal ,Tyrosine phosphorylation ,Neoplasms, Experimental ,Molecular biology ,Rats, Inbred F344 ,Rats ,Oncology ,chemistry ,Female ,Growth inhibition ,Oligopeptides ,Conjugate - Abstract
The EphA2 receptor tyrosine kinase is selectively expressed on the surface of many different human tumors. We have previously shown that tumor cells can be targeted by EphA2 monoclonal antibodies and that these antibodies function, in part, by inducing EphA2 internalization and degradation. In this report, we describe the isolation and characterization of a fully human monoclonal antibody (1C1) that selectively binds both the human and rodent EphA2 receptor. After cell binding, the antibody induces rapid tyrosine phosphorylation, internalization, and degradation of the EphA2 receptor. Because monoclonal antibodies that selectively bind tumor cells and internalize provide a vehicle for targeted delivery of cytotoxics, 1C1 was conjugated to the microtubule inhibitor monomethylauristatin phenylalanine using a stable maleimidocaproyl linker. The anti-EphA2 antibody-drug conjugate [1C1–maleimidocaproyl-MMAF (mcMMAF)] stimulated the activation of caspase-3/caspase-7 and the death of EphA2-expressing cells with IC50 values as low as 3 ng/mL. Similarly, the conjugate induced degradation of the EphA2 receptor and inhibited tumor growth in vivo. Administration of 1C1-mcMMAF at doses as low as 1 mg/kg once weekly resulted in significant growth inhibition of EphA2-expressing tumors without any observable adverse effects in mouse xenograft and rat syngeneic tumor models. Our data support the use of an antibody-drug conjugate approach to selectively target and inhibit the growth of EphA2-expressing tumors. [Cancer Res 2008;68(22):9367–74]
- Published
- 2008
- Full Text
- View/download PDF
21. Overproduction of IGF-2 drives a subset of colorectal cancer cells, which specifically respond to an anti-IGF therapeutic antibody and combination therapies
- Author
-
Helen Zhong, Robert E. Hollingsworth, Christine Fazenbaker, Cui Chen, Shannon Breen, P Ren, Ronald Herbst, X Yao, Yihong Yao, and Jiaqi Huang
- Subjects
0301 basic medicine ,Cancer Research ,Colorectal cancer ,Mice, Nude ,Apoptosis ,Biology ,Molecular oncology ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Growth factor receptor ,Trastuzumab ,Insulin-Like Growth Factor II ,Cell Line, Tumor ,Genetics ,medicine ,Animals ,Humans ,neoplasms ,Molecular Biology ,Cetuximab ,Gene Amplification ,Antibodies, Monoclonal ,Cell cycle ,medicine.disease ,Xenograft Model Antitumor Assays ,digestive system diseases ,030104 developmental biology ,030220 oncology & carcinogenesis ,Immunology ,DNA methylation ,Cancer research ,Selumetinib ,Female ,Colorectal Neoplasms ,medicine.drug ,Signal Transduction - Abstract
Colorectal cancer (CRC) is a heterogeneous disease with a broad spectrum of genetic and epigenetic changes. A comprehensive molecular characterization of CRC by The Cancer Genome Atlas Network detected the overexpression of the insulin-like growth factor 2 (IGF2) gene, encoding a ligand for the insulin-like growth factor 1 receptor (IGF-1R), in a subset of CRC tumors. In this study, we investigated the oncogenic potential of IGF-2 in IGF2-overexpressing CRC models and the efficacy of MEDI-573, an IGF-1/2-neutralizing antibody. We found that a subset of CRC cell lines express high IGF-2 levels owing to an increased DNA copy number and hypermethylation in the H19 promoter of the IGF2 gene. MEDI-573 efficiently neutralized IGF-2 and induced apoptosis, which resulted in significant tumor growth inhibition in CRC mouse models that express high levels of IGF-2. These effects were specific to CRCs overexpressing IGF-2, as MEDI-573 did not affect the growth CRC cell lines with normal levels. Moreover, blockade of IGF-2 by MEDI-573 modulated other signaling pathways, suggesting combination therapies with inhibitors of these pathways. Indeed, in vivo efficacy was significantly enhanced when MEDI-573 was used in combination with trastuzumab, AZD2014 (dual mTORC1/2i), AZD5363 (AKTi) and selumetinib (AZD6244/ARRY-142886, MEK1/2i) or cetuximab. These results demonstrate that overexpressed IGF-2 is the major tumorigenic driver in a subset of CRCs and encourage testing of MEDI-573, alone and in combinations, in IGF2-overexpressing CRC patients.
- Published
- 2015
22. Amifostine (ETHYOL) protects rats from mucositis resulting from fractionated or hyperfractionated radiation exposure
- Author
-
Christine Fazenbaker, Christine M. Bachy, Gizachew Kifle, Michael P. McCarthy, and David R. Cassatt
- Subjects
Cancer Research ,Injections, Subcutaneous ,medicine.medical_treatment ,Drug Evaluation, Preclinical ,Radiation-Protective Agents ,Pharmacology ,Rats, Sprague-Dawley ,Hyperfractionated Radiation ,Amifostine ,Pharmacokinetics ,Mucositis ,Animals ,Medicine ,Tissue Distribution ,Radiology, Nuclear Medicine and imaging ,Stomatitis ,Active metabolite ,Radiation ,business.industry ,Mouth Mucosa ,Dose fractionation ,Mammary Neoplasms, Experimental ,medicine.disease ,Mercaptoethylamines ,Rats, Inbred F344 ,Rats ,Radiation therapy ,Radiation Injuries, Experimental ,Oncology ,Injections, Intravenous ,Female ,Dose Fractionation, Radiation ,business ,Nuclear medicine ,medicine.drug - Abstract
Purpose The cytoprotective drug amifostine (Ethyol) protects rats from oral mucositis resulting from a single dose of γ-irradiation. We expanded earlier studies to determine whether multiple doses of amifostine protect against fractionated or hyperfractionated radiation and whether the active metabolite of amifostine (WR-1065) accumulates in tissues upon repeated administration. Methods and materials Rats received amifostine daily for 5 days in conjunction with a 1-week fractionated radiation schedule and were evaluated for oral mucositis. Rats also received amifostine before the am or pm exposure or b.i.d. in conjunction with hyperfractionated radiation. To determine the pharmacokinetics of WR-1065 after repeated dosing, amifostine was given 5 days a week for 1 or 3 weeks, and rat tissue and plasma were collected at intervals during and after treatment and analyzed for WR-1065. Results Amifostine protected rats from mucositis resulting from fractionated or hyperfractionated radiation. When the number of days of amifostine administration was reduced, protection was diminished. A dose of 100 mg/kg given in the morning or 2 doses at 50 mg/kg provided the best protection against hyperfractionated radiation. WR-1065 did not accumulate in tissues or tumor upon repeated administration. Conclusions Amifostine prevented radiation-induced mucositis in a rat model; protection was dose and schedule dependent.
- Published
- 2005
- Full Text
- View/download PDF
23. Tissue Levels of WR-1065, the Active Metabolite of Amifostine (Ethyol®), Are Equivalent following Intravenous or Subcutaneous Administration in Cynomolgus Monkeys
- Author
-
David R. Cassatt, Michael P. McCarthy, Gizachew Kifle, Christine M. Bachy, and Christine Fazenbaker
- Subjects
Drug ,Cancer Research ,business.industry ,media_common.quotation_subject ,Metabolite ,medicine.medical_treatment ,General Medicine ,Amifostine ,Pharmacology ,Bioavailability ,Radiation therapy ,chemistry.chemical_compound ,Route of administration ,Oncology ,chemistry ,Pharmacokinetics ,Medicine ,business ,Active metabolite ,media_common ,medicine.drug - Abstract
Amifostine (Ethyol®) is a cytoprotective drug approved for the reduction of xerostomia in head and neck cancer when administered to patients receiving postoperative radiation therapy. Although amifostine is approved for intravenous infusion, the off-label subcutaneous route of administration has become more prevalent. Although human patient data indicate higher plasma bioavailability of the active metabolite (WR-1065) following intravenous compared to subcutaneous administration, there are no corresponding data showing human tissue levels of WR-1065 following either route of administration due to the difficulty in obtaining human specimens. In our study we compared plasma and tissue pharmacokinetics of WR-1065 in primates following both routes of administration. Monkeys received amifostine at a dose of 260 mg/m2 either intravenously or subcutaneously. Plasma samples were analyzed for total WR-1065 by reverse-phase high-pressure liquid chromatography (HPLC) and fluorescence detection up to 4 h after amifostine administration. Tissues were analyzed for free WR-1065 by reverse-phase HPLC and electrochemical detection 30 and 60 min after administration. Following intravenous administration, plasma WR-1065 levels peaked rapidly and showed a bi-exponential decline, while following subcutaneous administration WR-1065 levels rose slowly and declined exponentially. The relative plasma bioavailability of WR-1065 given subcutaneously was lower at 30 and 60 min. Interestingly, after 30 min, tissues showed equal or slightly greater concentrations of WR-1065 following subcutaneous administration. Levels following 60 min were comparable following both routes. The plasma bioavailability studies performed in primates confirm human plasma data. Expanding the study to evaluate primate tissue levels of WR-1065 revealed that despite lower plasma bioavailability following subcutaneous administration, tissue levels of the active metabolite were surprisingly greater than or equal to those measured in animals that received the drug intravenously. These studies strengthen the argument for subcutaneous administration of amifostine in radiation oncology.
- Published
- 2004
- Full Text
- View/download PDF
24. Effects of dose and schedule on the efficacy of Ethyol: preclinical studies1 1The authors are employees and shareholders of MedImmune Inc
- Author
-
Gizachew Kifle, Christine M. Bachy, Christine Fazenbaker, and David R. Cassatt
- Subjects
business.industry ,Rat model ,Hematology ,Plasma levels ,Amifostine ,Pharmacology ,medicine.disease ,Radiation exposure ,Oncology ,Pharmacokinetics ,Mucositis ,medicine ,Dosing ,business ,Active metabolite ,medicine.drug - Abstract
The chemo- and radioprotectant drug amifostine (Ethyol; MedImmune, Inc, Gaithersburg, MD) is approved for intravenous (IV) administration; however, the subcutaneous (SC) route is being explored as a practical alternative. We have previously reported equivalence between IV and SC administration using a rat model of radioprotection and active metabolite (WR-1065) tissue pharmacokinetics. To examine the more clinically relevant fractionated and hyperfractionated radiation schedules and the effects of variations in the time of amifostine administration, we expanded these studies to include radioprotection and pharmacokinetic studies of WR-1065 using multiple dosing. To measure radioprotection using a fractionated radioprotection model, rats were given amifostine over a 1-week period at various doses (25 mg/kg, 50 mg/kg, 100 mg/kg; or 162.5 mg/m(2), 325 mg/m(2), 650 mg/m(2), respectively) IV or SC daily 30 minutes before exposure to 7.5 Gy/dose. Rats were fully protected from mucositis at the highest amifostine dose, with protection diminishing as the amifostine was decreased. Equivalent protection was observed whether the drug was given IV or SC. When the number of days of amifostine administration was reduced, protection was diminished. Amifostine also protected against radiation delivered using a 1-week hyperfractionated schedule (4.5 Gy/exposure twice daily), with optimal protection occurring when the drug was administered bid 30 minutes before each exposure (50 mg/kg) or every day before the morning exposure (100 mg/kg). The need for daily dosing to achieve optimal radioprotection was consistent with the tissue pharmacokinetics of the active metabolite. We found that WR-1065 did not accumulate in tissues or in SC-implanted tumors when amifostine was administered daily for 3 weeks. In addition, tissue and tumor levels of WR-1065 declined to baseline 24 hours after each amifostine dose. In a monkey pharmacokinetic model, plasma levels of WR-1065 (characterized by a pronounced spike of WR-1065 immediately after IV administration that was absent when the drug was given SC) were similar to those of humans; however, levels of WR-1065 in the tissues were higher 30 minutes following SC administration and were equivalent 60 minutes following IV or SC administration. These results suggest that maximum tissue levels and protection occur when amifostine is given 30 to 60 minutes before radiation exposure, that treatment breaks reduce the radioprotection by amifostine, and that protection from hyperfractionated radiation is dependent on amifostine dose and schedule.
- Published
- 2003
- Full Text
- View/download PDF
25. Abstract 76: Synthetic lethal targeting of BRCA mutant tumors with antibody linked pyrrolobenzodiazepine dimers
- Author
-
Ravinder Tammali, Noel R. Monks, Ronald Herbst, Haihong Zhong, Cui Chen, Jay Harper, Christine Fazenbaker, Dave Tice, and Kennedy Maureen
- Subjects
0301 basic medicine ,Cancer Research ,endocrine system diseases ,biology ,Chemistry ,DNA repair ,Mutant ,Wild type ,Cancer ,Pyrrolobenzodiazepine ,Synthetic lethality ,medicine.disease ,biology.organism_classification ,Molecular biology ,HeLa ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,skin and connective tissue diseases ,Homologous recombination - Abstract
Pyrrolbenzodiazepine dimers (PBDs) are amongst the most potent DNA alkylating agents, with activity against a broad spectrum of tumors. PBDs form cross-links within the minor groove of DNA causing double strand breaks (DSB). DNA repair genes such as BRCA1 and BRCA2 play important roles in homologous recombination repair (HRR) of DSB. Cells defective in BRCA1 or BRCA2 are known to be sensitive to DNA interstrand crosslinks. Accordingly, it is possible that PBD-based ADCs will have enhanced killing of cells (synthetic lethality) in which HR processes are defective by inactivation of BRCA1 or BRCA2 genes in breast, ovarian and other cancers. To determine anti-tumor activity of PBD dimers, we have used MEDI0641, PBD-dimer conjugated to anti-5T4 antibody, against BRCA wild type and mutant xenograft tumor models. MEDI0641 was >3-fold more potent in BRCA1 or BRCA2 mutant models than in wild-type xenografts. Similar observations were seen in 25 patient-derived xenograft (PDX) models (19 breast and 6 ovarian) bearing mutations in BRCA1 or BRCA2 (blinded to 5T4 expression) treated with MEDI0641. Out of a total of 25 PDX models, 17 models had tumor regression with a single administration of MEDI0641 at 0.3 mg/kg (response rate = 68%), and 14 models showed response to 0.1 mg/kg of MEDI0641 (response rate = 56%). In BRCA wild-type PDX models, a higher dose of 1 mg/kg was required to achieve full anti-tumor efficacy. Retrospective analysis of 5T4 expression in PDX tumors demonstrated no correlation between efficacy and target expression in BRCA mutant PDX models. To further delineate the role of BRCA1/2 mutations in determining sensitivity to PBD, we used siRNA knock-down of both BRCA1 and BRCA2 in the DNA repair wild type HeLa cells. Knockdown of BRCA genes sensitized Hela cells to PBD payload and MEDI0641 in vitro. Anti-tumor activity of MEDI0641 was further examined in isogenic BRCA2 knockout xenograft models. Genetic deletion of BRCA2 markedly increased anti-tumor activity of MEDI0641. In conclusion, PBD based ADCs may have improved therapeutic window in cancer patients with somatic BRCA mutations. Citation Format: Haihong Zhong, Ravinder Tammali, Cui Chen, Christine Fazenbaker, Kennedy Maureen, Noel Monks, Jay Harper, Ronald Herbst, Dave Tice. Synthetic lethal targeting of BRCA mutant tumors with antibody linked pyrrolobenzodiazepine dimers [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 76. doi:10.1158/1538-7445.AM2017-76
- Published
- 2017
- Full Text
- View/download PDF
26. MEDI3617, a human anti-Angiopoietin 2 monoclonal antibody, inhibits angiogenesis and tumor growth in human tumor xenograft models
- Author
-
Meggan Czapiga, Ching Ching Leow, Serguei Soukharev, Norman Peterson, Karen Coffman, Ivan Inigo, Robert M. Woods, Theresa LaVallee, Shannon Breen, Sally-Ann Ricketts, Neill Gingles, Christine Fazenbaker, Yong Chang, Steve Coats, and Bahija Jallal
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Time Factors ,Paclitaxel ,Bevacizumab ,CD30 ,Angiogenesis ,Mice, Nude ,Neovascularization, Physiologic ,Angiogenesis Inhibitors ,Retinal Neovascularization ,Biology ,Antibodies, Monoclonal, Humanized ,Corrosion Casting ,Transfection ,Fluorescence ,Angiopoietin-2 ,Neovascularization ,Mice ,Cell Line, Tumor ,Neoplasms ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Animals ,Humans ,Phosphorylation ,Dose-Response Relationship, Drug ,Neovascularization, Pathologic ,Oncogene ,Tumor hypoxia ,Antibodies, Monoclonal ,X-Ray Microtomography ,Receptor, TIE-2 ,Xenograft Model Antitumor Assays ,Tumor Burden ,HEK293 Cells ,HIF1A ,Oncology ,Blood vessel maturation ,Cancer research ,Female ,medicine.symptom ,medicine.drug - Abstract
Angiopoietin 2 (Ang2) is an important regulator of angiogenesis, blood vessel maturation and integrity of the vascular endothelium. The correlation between the dynamic expression of Ang2 in tumors with regions of high angiogenic activity and a poor prognosis in many tumor types makes Ang2 an ideal drug target. We have generated MEDI3617, a human anti-Ang2 monoclonal antibody that neutralizes Ang2 by preventing its binding to the Tie2 receptor in vitro, and inhibits angiogenesis and tumor growth in vivo. Treatment of mice with MEDI3617 resulted in inhibition of angiogenesis in several mouse models including: FGF2-induced angiogenesis in a basement extract plug model, tumor and retinal angiogenesis. In xenograft tumor models, treatment with MEDI3617 resulted in a reduction in tumor angiogenesis and an increase in tumor hypoxia. The administration of MEDI3617 as a single agent to mice bearing human tumor xenografts resulted in tumor growth inhibition against a broad spectrum of tumor types. Combining MEDI3617 with chemotherapy or bevacizumab resulted in a delay in tumor growth and no body weight loss was observed in the combination groups. These results, combined with pharmacodynamic studies, demonstrate that treatment of tumor-bearing mice with MEDI3617 significantly inhibited tumor growth as a single agent by blocking tumor angiogenesis. Together, these data show that MEDI3617 is a robust antiangiogenic agent and support the clinical evaluation and biomarker development of MEDI3617 in cancer patients.
- Published
- 2012
- Full Text
- View/download PDF
27. Antitumor efficacy of IPI-504, a selective heat shock protein 90 inhibitor against human epidermal growth factor receptor 2-positive human xenograft models as a single agent and in combination with trastuzumab or lapatinib
- Author
-
David E. Weng, Ching Ching Leow, Karen Coffman, Bahija Jallal, Christine Fazenbaker, Yong Chang, Steve Coats, Dowdy Jackson, John Gooya, and Jon Chesebrough
- Subjects
Cancer Research ,Receptor, ErbB-2 ,Lactams, Macrocyclic ,Pharmacology ,Lapatinib ,Antibodies, Monoclonal, Humanized ,In vivo ,Trastuzumab ,hemic and lymphatic diseases ,Heat shock protein ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,Benzoquinones ,Humans ,HSP90 Heat-Shock Proteins ,skin and connective tissue diseases ,Receptor ,neoplasms ,Protein kinase B ,Protein Kinase Inhibitors ,biology ,Chemistry ,Antibodies, Monoclonal ,Drug Synergism ,Hsp90 ,Xenograft Model Antitumor Assays ,Oncology ,Mechanism of action ,biology.protein ,Quinazolines ,medicine.symptom ,Drug Screening Assays, Antitumor ,medicine.drug - Abstract
IPI-504 is a novel, highly soluble small-molecule inhibitor of heat shock protein 90 (Hsp90), a protein chaperone essential for regulating homeostasis of oncoproteins and cell signaling proteins. Human epidermal growth factor receptor 2 (HER2; ErbB2) oncoprotein, expressed in a subset of metastatic breast cancers, is a Hsp90 client protein. In this study, we investigated the antitumor activity and the mechanism of action of IPI-504 in HER2+, trastuzumab-sensitive and trastuzumab-refractory cell lines in vitro and in vivo. IPI-504 exhibited potent antiproliferative activities (range of IC50, 10-40 nmol/L) against several tumor cell lines examined, whereby mechanism of action was mediated through HER2 and Akt degradation. Both intravenous and oral administration of IPI-504 assessed in multiple schedules showed potent tumor growth inhibition in vivo with corresponding degradation of HER2. The tolerability and efficacy of IPI-504 combined with either trastuzumab or lapatinib were also investigated in HER2+ tumor xenograft models. Combination of IPI-504 with trastuzumab significantly enhanced tumor growth delay and induced greater responses when compared with either agent alone. Although, as expected, trastuzumab alone did not exhibit any significant antitumor activity in the trastuzumab-resistant JIMT-1 model, IPI-504 administered in combination with trastuzumab yielded greater antitumor efficacy than either agent alone. Finally, combination of IPI-504 and lapatinib was well tolerated up to 50 mg/kg IPI-504 and 100 mg/kg lapatinib and resulted in significant delay in tumor growth, including partial and complete tumor responses. These lines of evidence support the development of IPI-504 in HER2-positive breast cancers as a single agent and in combination with either trastuzumab or lapatinib.[Mol Cancer Ther 2009;8(8):2131–41]
- Published
- 2009
28. Subcutaneous administration of amifostine (ethyol) is equivalent to intravenous administration in a rat mucositis model
- Author
-
Christine M. Bachy, David R. Cassatt, Christine Fazenbaker, and Gizachew Kifle
- Subjects
Cancer Research ,medicine.medical_treatment ,Injections, Subcutaneous ,Rat model ,Radiation-Protective Agents ,Pharmacology ,Xerostomia ,Rats, Sprague-Dawley ,Amifostine ,Pharmacokinetics ,Mucositis ,Medicine ,Animals ,Radiology, Nuclear Medicine and imaging ,In patient ,Radiation Injuries ,Active metabolite ,Stomatitis ,Radiation ,business.industry ,Mouth Mucosa ,medicine.disease ,Mercaptoethylamines ,Rats ,Radiation therapy ,Oncology ,Anesthesia ,Injections, Intravenous ,Models, Animal ,Female ,business ,medicine.drug - Abstract
Purpose Amifostine (Ethyol) is currently approved for intravenous (IV) administration to prevent xerostomia in patients receiving radiotherapy for head-and-neck cancer. Recently, subcutaneous (SC) administration has been explored as an alternative route. To determine whether SC administration was equivalent to IV administration, we used models to follow pharmacokinetics and oral mucosal protection in rats. Methods Amifostine was administered to rats at doses of 200, 100, or 50 mg/kg (1300, 650, or 325 mg/m 2 ) IV or SC at various times before radiation at 15.3 Gy (protection studies) or harvest of blood and tissues for analysis by HPLC (pharmacokinetic studies). Results Amifostine administered IV or SC 1 h before radiation protected rats from mucositis, but the protective effect was more prolonged when amifostine was administered SC. Tissue levels of the active metabolite (WR-1065) were equivalent after SC administration. The correlation between tissue levels of WR-1065 and protection was strong, but that between blood levels of WR-1065 and protection was only weak. Conclusions These data demonstrate that, in a rat model, SC administration of amifostine was at least as effective as that by IV.
- Published
- 2003
29. Preclinical studies on the radioprotective efficacy and pharmacokinetics of subcutaneously administered amifostine
- Author
-
Christine Fazenbaker, Christine M. Bachy, David R. Cassatt, and Gizachew Kifle
- Subjects
Drug ,media_common.quotation_subject ,Injections, Subcutaneous ,Drug Evaluation, Preclinical ,Biological Availability ,Radiation-Protective Agents ,Pharmacology ,Amifostine ,Pharmacokinetics ,medicine ,Mucositis ,Animals ,Dosing ,Radiation Injuries ,Stomatitis ,Active metabolite ,media_common ,business.industry ,Hematology ,medicine.disease ,Mercaptoethylamines ,Rats ,Macaca fascicularis ,Oncology ,Cytoprotection ,Injections, Intravenous ,Animal studies ,business ,medicine.drug - Abstract
The radioprotective effects and pharmacokinetics of subcutaneously (SC) administered amifostine have been investigated in animal studies. Studies in rats using a single dose of amifostine showed that SC administration gave protection from radiation-induced mucositis that is at least equivalent to that achieved by intravenous administration of the drug. These studies also indicate that tissue levels of the active metabolite WR-1065 correlated better with the radioprotective effects of amifostine than do plasma WR-1065 levels. Multiple-dose studies in rats show radioprotective effects equal to or greater than those obtained with intravenous dosing in the setting of fractionated irradiation. In addition, there is no evidence of drug accumulation in either normal or tumor tissue, with tumor WR-1065 levels peaking just above the limits of quantitation during treatment. Preliminary data from studies of SC amifostine in monkeys indicate a plasma pharmacokinetic profile similar to that reported earlier in humans. Tissue WR-1065 levels were higher at 30 minutes after SC dosing than they were after intravenous dosing and were comparable for the two routes at 60 minutes. Semin Oncol 29 (suppl 19):2-8. Copyright 2002, Elsevier Science (USA). All rights reserved.
- Published
- 2003
30. Preclinical modeling of improved amifostine (Ethyol) use in radiation therapy
- Author
-
Mark S. Hanson, David R. Cassatt, Christine M. Bachy, and Christine Fazenbaker
- Subjects
Cancer Research ,medicine.medical_specialty ,Erythema ,medicine.medical_treatment ,Injections, Subcutaneous ,Radiation-Protective Agents ,Radiation Dosage ,Gastroenterology ,Rats, Sprague-Dawley ,Amifostine ,Pharmacokinetics ,Internal medicine ,medicine ,Mucositis ,Animals ,Radiology, Nuclear Medicine and imaging ,Radiation Injuries ,Active metabolite ,Stomatitis ,business.industry ,Head and neck cancer ,Mouth Mucosa ,medicine.disease ,Parotid gland ,Rats ,Radiation therapy ,medicine.anatomical_structure ,Oncology ,Anesthesia ,Injections, Intravenous ,Female ,medicine.symptom ,business ,Head ,medicine.drug - Abstract
Amifostine (Ethyol) has been evaluated clinically as a radioprotective agent for the prevention of xerostomia and mucositis for patients receiving radiotherapy (RT). Currently, amifostine is approved for the prevention of xerostomia in head and neck cancer patients receiving RT when administered intravenously (IV) before RT. For the clinician, there would be several advantages to administering the drug subcutaneously and to being able to show its protective effects on mucositis. The authors have developed a rat RT model to examine the protective effects of amifostine after IV and subcutaneous (SC) administration in a mucositis model. Rats (5 per group) were given 200 mg/kg (human dose equivalent of approximately 1,300 mg/m(2)) of amifostine either IV or SC, and their head and neck regions were exposed to 15.3 Gy of gamma radiation 0.5, 2, 4, and 8 hours after amifostine administration. For 10 days after treatment, the oral cavities of the rats were examined for signs of mucositis. Mucosal erythema and mucosal edema were scored according to 0 through 5 and 0 through 2 scales, respectively, with the scores added to indicate overall mucositis. The average mucositis score for the untreated animals was 3.5. Rats were protected from mucositis up to 4 hours when given amifostine either IV or SC. Rats that received amifostine SC, but not IV, were protected from mucositis 8 hours after administration. Preliminary pharmacokinetic data have revealed slightly higher active metabolite (WR-1065) levels in the parotid gland and small intestine in the rats given amifostine SC compared with IV and equivalent levels in the plasma and kidney. The data showed that SC administration of amifostine gave radioprotection comparable to IV administration up to 4 hours before RT and may be more effective than IV administration at longer pretreatment intervals.
- Published
- 2002
31. Abstract 4493: Medi-573 alone or in combination with mammalian target of rapamycin inhibitors, targets the insulin-like growth factor pathway in sarcomas
- Author
-
Jiaqi Huang, Cui Chen, Shannon Breen, Haihong Zhong, Morehouse Chris, Robert E. Hollingsworth, Yihong Yao, and Christine Fazenbaker
- Subjects
Cancer Research ,medicine.medical_specialty ,biology ,business.industry ,Cell growth ,Growth factor ,medicine.medical_treatment ,medicine.disease ,Paracrine signalling ,Insulin-like growth factor ,Insulin receptor ,Endocrinology ,Oncology ,Internal medicine ,biology.protein ,Cancer research ,Medicine ,Sarcoma ,business ,Autocrine signalling ,Protein kinase B - Abstract
MEDI-573 is a human antibody that neutralizes the insulin-like growth factors, IGF-1 and IGF-2. IGFs are over-expressed in multiple types of cancer; their over-expression is a potential mechanism for resistance to IGF-1 receptor (IGF-1R)-targeting therapy. Effects of IGFs on cell proliferation, differentiation, and survival are mediated through their binding to and activation of IGF-1R or insulin receptor A (IR-A). In this study, we studied the anti-tumor activity and mechanism of action of MEDI-573 in models of sarcoma. MEDI-573 potently inhibited in vitro proliferation of several sarcoma cell lines, with Ewing's sarcoma cell lines being the most sensitive. This inhibition also occurred after growth stimulation with added IGF-1- and IGF-2. The effect of MEDI-573 on IGF signaling was also examined. Treatment with MEDI-573 markedly reduced levels of pIGF-1R, pIR, and pAKT, and significantly blocked IGF-1- and IGF-2-induced activation of the IGF-1R and AKT pathways. MEDI-573 inhibited the growth of sarcoma xenografts in vivo, and inhibition correlated with neutralization of IGF-1 and IGF-2. Combination of MEDI-573 with either rapamycin or another mTOR inhibitor, AZD2014, significantly enhanced the anti-tumor activity of MEDI-573, and this response correlated with modulation of AKT and mTOR signaling. In summary, sarcoma cells respond to autocrine or paracrine growth stimulation by secreted IGF-1 and IGF-2, and inhibition of IGF-1 and IGF-2 by MEDI-573 results in potent anti-tumor activity in several sarcoma models. Our data provide evidence for evaluation of MEDI-573 and mTORi combinations in clinical studies of sarcoma patients. Note: This abstract was not presented at the meeting. Citation Format: Haihong Zhong, Christine Fazenbaker, Shannon Breen, Cui Chen, Jiaqi Huang, Morehouse Chris, Yihong Yao, Robert Hollingsworth. Medi-573 alone or in combination with mammalian target of rapamycin inhibitors, targets the insulin-like growth factor pathway in sarcomas. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4493. doi:10.1158/1538-7445.AM2014-4493
- Published
- 2014
- Full Text
- View/download PDF
32. Conformational nature of the Borrelia burgdorferi decorin binding protein A epitopes that elicit protective antibodies
- Author
-
Christine M. Bachy, Christine Fazenbaker, William C. Roberts, Nancy Ulbrandt, Nita K. Patel, Mark S. Hanson, and David R. Cassatt
- Subjects
Immunogen ,Protein Conformation ,Immunology ,Molecular Sequence Data ,Cross Reactions ,Microbiology ,Epitope ,Mice ,Antigen ,Bacterial Proteins ,Borrelia burgdorferi Group ,Animals ,Amino Acid Sequence ,Borrelia burgdorferi ,Adhesins, Bacterial ,Antigens, Bacterial ,Lyme Disease ,Mice, Inbred C3H ,Vaccines, Synthetic ,biology ,Binding protein ,Vaccination ,Lyme Disease Vaccines ,Ligand (biochemistry) ,biology.organism_classification ,Antibodies, Bacterial ,Cell biology ,Disease Models, Animal ,Infectious Diseases ,Microbial Immunity and Vaccines ,biology.protein ,Epitopes, B-Lymphocyte ,Parasitology ,Female ,Antibody ,Protein A ,Carrier Proteins ,Bacterial Outer Membrane Proteins - Abstract
Decorin binding protein A (DbpA) has been shown by several laboratories to be a protective antigen for the prevention of experimental Borrelia burgdorferi infection in the mouse model of Lyme borreliosis. However, different recombinant forms of the antigen having either lipidated amino termini, approximating the natural secretion and posttranslational processing, or nonprocessed cytosolic forms have elicited disparate levels of protection in the mouse model. We have now used the unique functional properties of this molecule to investigate the structural requirements needed to elicit a protective immune response. Genetic and physicochemical alterations to DbpA showed that the ability to bind to the ligand decorin is indicative of a potent immunogen but is not conclusive. By mutating the two carboxy-terminal nonconserved cysteines of DbpA from B. burgdorferi strain N40, we have determined that the stability afforded by the putative disulfide bond is essential for the generation of protective antibodies. This mutated protein was more sensitive to thermal denaturation and proteolysis, suggesting that it is in a less ordered state. Immunization with DbpA that was thermally denatured and functionally inactivated stimulated an immune response that was not protective and lacked bactericidal antibodies. Antibodies against conformationally altered forms of DbpA also failed to kill heterologous B. garinii and B. afzelii strains. Additionally, nonsecreted recombinant forms of DbpA N40 were found to be inferior to secreted lipoprotein DbpA N40 in terms of functional activity and antigenic potency. These data suggest that elicitation of a bactericidal and protective immune response to DbpA requires a properly folded conformation for the production of functional antibodies.
- Published
- 2001
33. Tissue levels of the active metabolite WR-1065 in monkeys following a subcutaneous injection of amifostine (Ethyol®) are equivalent to or higher than levels measured following an intravenous infusion
- Author
-
Christine Fazenbaker, Christine M. Bachy, and David R. Cassatt
- Subjects
Cancer Research ,Subcutaneous injection ,Radiation ,Oncology ,business.industry ,Anesthesia ,Medicine ,Radiology, Nuclear Medicine and imaging ,Amifostine ,business ,Active metabolite ,medicine.drug - Published
- 2002
- Full Text
- View/download PDF
34. Abstract 1364: Combination of MEDI3617, a fully human anti-angiopoietin 2 monoclonal antibody, with inhibitors of the VEGF pathway enhances antitumor activity in vivo
- Author
-
Ivan Inigo, Ching Ching Leow, Steve Coats, Christine Fazenbaker, Yong Chang, Pamela Trail, Karen Coffman, Bahija Jallal, and Theresa LaVallee
- Subjects
Sorafenib ,Cancer Research ,Bevacizumab ,Angiogenesis ,business.industry ,medicine.drug_class ,Cancer ,Pharmacology ,medicine.disease ,Monoclonal antibody ,Oncology ,In vivo ,Pancreatic cancer ,Blood vessel maturation ,medicine ,business ,medicine.drug - Abstract
Angiopoietin 2 (Ang2) is an important regulator of angiogenesis, blood vessel maturation, and maintenance of the integrity of the vascular endothelium. Ang2-Tie2 interactions for vascular remodeling have been shown to play an important role in tumor vessel plasticity. MEDI3617 is a fully human anti-Ang2 monoclonal antibody that neutralizes the activity of Ang2, thereby inhibiting Ang2 from binding and signaling through the Tie2 receptor. Previously, we have demonstrated that treatment with MEDI3617 resulted in tumor growth inhibition in a wide spectrum of subcutaneous human tumor xenograft models. Given that angiogenesis is a process that requires concerted interplay of mutiple pathways, predominantly the VEGF pathway and the Ang/Tie pathway, we hypothesized that maximal inhibition of tumoral angiogenesis and consequently tumor growth could be achieved by blocking both pathways. We investigated the activity and tolerability of combining MEDI3617 with inhibitors of the VEGF pathway such as sorafenib and bevacizumab. In PLCPRF/5, a hepatocellular carcinoma xenograft model, combination of MEDI3617 and sorafenib resulted in a significant increase tumor growth inhibition. Similarly, in HPAC, a pancreatic cancer xenograft model, combination of MEDI3617 and bevacizumab resulted in greater tumor growth inhibition than either single agent alone. Mice exhibited no adverse effects or wt. loss following combination treatments. Taken together, these data support the clinical evaluation of the combination treatment of MEDI3617 and a VEGF inhibitor in cancer patients. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 1364.
- Published
- 2010
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.