9 results on '"Mollie Daugherty"'
Search Results
2. Table S1 from MOR209/ES414, a Novel Bispecific Antibody Targeting PSMA for the Treatment of Metastatic Castration-Resistant Prostate Cancer
- Author
-
John W. Blankenship, Jane A. Gross, Sateesh K. Natarajan, Catherine J. McMahan, David Bienvenue, Paul A. Algate, Jennifer Wiens, Padma Ravikumar, Robert E. Miller, John Kumer, Rebecca Gottschalk, Hang Fang, Maria Dasovich, Mollie Daugherty, Ruth A. Chenault, Jeannette Bannink, Robert Bader, Toddy Sewell, and Gabriela Hernandez-Hoyos
- Abstract
Phenotype of T-cells used in Xenograft Studies
- Published
- 2023
- Full Text
- View/download PDF
3. Figure S3 from MOR209/ES414, a Novel Bispecific Antibody Targeting PSMA for the Treatment of Metastatic Castration-Resistant Prostate Cancer
- Author
-
John W. Blankenship, Jane A. Gross, Sateesh K. Natarajan, Catherine J. McMahan, David Bienvenue, Paul A. Algate, Jennifer Wiens, Padma Ravikumar, Robert E. Miller, John Kumer, Rebecca Gottschalk, Hang Fang, Maria Dasovich, Mollie Daugherty, Ruth A. Chenault, Jeannette Bannink, Robert Bader, Toddy Sewell, and Gabriela Hernandez-Hoyos
- Abstract
Serum levels of human PSA from day 56 of the MDA-PCa-2b xenograft study (as shown in Figures 5C, 5D).
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Methods from MOR209/ES414, a Novel Bispecific Antibody Targeting PSMA for the Treatment of Metastatic Castration-Resistant Prostate Cancer
- Author
-
John W. Blankenship, Jane A. Gross, Sateesh K. Natarajan, Catherine J. McMahan, David Bienvenue, Paul A. Algate, Jennifer Wiens, Padma Ravikumar, Robert E. Miller, John Kumer, Rebecca Gottschalk, Hang Fang, Maria Dasovich, Mollie Daugherty, Ruth A. Chenault, Jeannette Bannink, Robert Bader, Toddy Sewell, and Gabriela Hernandez-Hoyos
- Abstract
Supplementary Methods and Supplementary Figure Legends
- Published
- 2023
- Full Text
- View/download PDF
5. MOR209/ES414, a Novel Bispecific Antibody Targeting PSMA for the Treatment of Metastatic Castration-Resistant Prostate Cancer
- Author
-
Ruth A. Chenault, Jeannette Bannink, Hang Fang, Toddy Sewell, Catherine J. McMahan, John W. Blankenship, Robert R. Bader, David Bienvenue, Sateesh Kumar Natarajan, Paul A. Algate, Robert E. Miller, Jane A. Gross, Mollie Daugherty, Jennifer Wiens, Maria M. Dasovich, Gabriela Hernandez-Hoyos, John Kumer, Rebecca Gottschalk, and Padma Ravikumar
- Subjects
Cytotoxicity, Immunologic ,Glutamate Carboxypeptidase II ,Male ,0301 basic medicine ,Cancer Research ,CD3 Complex ,medicine.medical_treatment ,Cell ,Antineoplastic Agents ,Mice, Transgenic ,Lymphocyte Activation ,Protein Engineering ,Mice ,03 medical and health sciences ,Prostate cancer ,0302 clinical medicine ,Antigen ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Antibodies, Bispecific ,Animals ,Humans ,Medicine ,Cytotoxic T cell ,Cytotoxicity ,biology ,business.industry ,Cancer ,Immunotherapy ,medicine.disease ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,Prostatic Neoplasms, Castration-Resistant ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Drug Design ,030220 oncology & carcinogenesis ,Antigens, Surface ,Immunology ,biology.protein ,Cancer research ,Antibody ,business ,Single-Chain Antibodies - Abstract
Treatment of metastatic, castration-resistant prostate cancer (mCRPC) remains a highly unmet medical need and current therapies ultimately result in disease progression. Immunotherapy is a rapidly growing approach for treatment of cancer but has shown limited success to date in the treatment of mCRPC. We have developed a novel humanized bispecific antibody, MOR209/ES414, built on the ADAPTIR (modular protein technology) platform, to redirect T-cell cytotoxicity toward prostate cancer cells by specifically targeting T cells through CD3ϵ to prostate cancer cells expressing PSMA (prostate-specific membrane antigen). In vitro cross-linking of T cells with PSMA-expressing tumor cells by MOR209/ES414 triggered potent target-dependent tumor lysis and induction of target-dependent T-cell activation and proliferation. This activity occurred at low picomolar concentrations of MOR209/ES414 and was effective at low T-effector to tumor target cell ratios. In addition, cytotoxic activity was equivalent over a wide range of PSMA expression on target cells, suggesting that as few as 3,700 PSMA receptors per cell are sufficient for tumor lysis. In addition to high sensitivity and in vitro activity, MOR209/ES414 induced limited production of cytokines compared with other bispecific antibody formats. Pharmacokinetic analysis of MOR209/ES414 demonstrated a serum elimination half-life in NOD/SCID γ (NSG) mice of 4 days. Administration of MOR209/ES414 in murine xenograft models of human prostate cancer significantly inhibited tumor growth, prolonged survival, and decreased serum prostate-specific antigen levels only in the presence of adoptively transferred human T cells. On the basis of these preclinical findings, MOR209/ES414 warrants further investigation as a potential therapeutic for the treatment of CRPC. Mol Cancer Ther; 15(9); 2155–65. ©2016 AACR.
- Published
- 2016
- Full Text
- View/download PDF
6. Abstract LB-199: APVO436, a bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecule for redirected T-cell cytotoxicity with limited cytokine release, is well tolerated in repeat dose toxicology studies in cynomolgus macaques
- Author
-
Michael R. Comeau, Bannink Jeannette, Mollie Daugherty, David Jablonski, Lynda Misher, Catherine J. McMahan, David Bienvenue, Lara Parr, Rebecca Gottschalk, Gabriela H. Hoyos, Starrla Johnson, Toddy Sewell, Jane A. Gross, Melissa DeFrancesco, and John Kumer
- Subjects
0301 basic medicine ,Cancer Research ,biology ,medicine.diagnostic_test ,business.industry ,medicine.medical_treatment ,CD3 ,medicine.disease ,Flow cytometry ,03 medical and health sciences ,Cytokine release syndrome ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,biology.protein ,Cytotoxic T cell ,Interleukin-3 receptor ,Antibody ,Cytotoxicity ,business - Abstract
Introduction: Depletion of CD123 over-expressing tumor cells may improve patient outcomes in several hematological malignancies including AML, MDS, ALL, CML, HCL and BPDCN. CD123 is infrequently expressed by normal cells making it an attractive tumor target, currently being pursued using several different approaches including T-cell engaging immunotherapies. Cytokine release syndrome (CRS) is a concern with T-cell engaging immunotherapies that may require complex clinical development strategies to manage safely. We have developed APVO436, a bispecific anti-CD123 x anti-CD3 ADAPTIR molecule for redirecting T-cell cytotoxicity to CD123-expressing tumor cells, currently in Phase I clinical testing in AML and MDS. A potential advantage of the ADAPTIR platform is reduced cytokine release compared to other formats (Mol Cancer Ther; 15(9):2155-65). We previously compared APVO436 activity to another CD123 x CD3 bispecific antibody containing the amino acid sequence of MGD006. We also demonstrated that APVO436 induces activation of AML patient T cells to kill endogenous tumor cells. Here we extend these studies to test the capacity of APVO436 to induce memory T-cell generation and describe the results of repeat dose toxicology studies in cynomolgus macaques. Methods: Binding, T-cell activation and proliferation were assessed using flow cytometry. Cytotoxic activity was determined using chromium release and flow cytometry assays. Cytokine levels were assessed using multi-plex technology. Repeat dose toxicology studies were performed in cynomolgus macaques. Results: In vitro, APVO436 induced lower levels of several T-cell cytokines while demonstrating similar levels of tumor cell lysis potency compared to other CD123 x CD3 bispecific antibodies. APVO436 induced extensive T-cell proliferation and the development of fully functional memory T cells with cytolytic function. Repeat-dose administration of APVO436 in NHP for 4 weeks by IV bolus injection induced transient changes in T lymphocytes and CD123+ leukocytes. Terminal elimination half-life ranged up to 108 hours with dose proportional exposure after first and last dose; some impact on serum concentration was observed from anti-drug antibodies at late time points. Conclusions: APVO436 induced lower levels of T-cell cytokines associated with CRS compared to other CD123 x CD3 targeting bispecific antibodies and induced the generation of functional memory T cells. Repeat-dose toxicology studies in cynomolgus monkeys demonstrate that APVO436 has antibody-like clearance and volume of distribution parameters and a serum half-life of about 4.5 days. These data support clinical studies with APVO436 as a potential treatment for AML and other hematological malignancies with possible safety advantages over other CD123 targeting therapies. Citation Format: Michael R. Comeau, Rebecca Gottschalk, Mollie Daugherty, Toddy Sewell, Lynda Misher, Bannink Jeannette, Starrla Johnson, Lara Parr, John Kumer, David Jablonski, Melissa DeFrancesco, David Bienvenue, Gabriela H. Hoyos, Catherine J. McMahan, Jane A. Gross. APVO436, a bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecule for redirected T-cell cytotoxicity with limited cytokine release, is well tolerated in repeat dose toxicology studies in cynomolgus macaques [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr LB-199.
- Published
- 2019
- Full Text
- View/download PDF
7. Abstract 1786: APVO436, a bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecule for redirected T-cell cytotoxicity, induces potent T-cell activation, proliferation and cytotoxicity with limited cytokine release
- Author
-
Robert E. Miller, David Bienvenue, Catherine J. McMahan, Toddy Sewell, Gabriela H. Hoyos, Jane A. Gross, Melissa DeFrancesco, Robert Bader, Lynda Misher, Lara Parr, Mollie Daugherty, Rebecca Gottschalk, and Michael R. Comeau
- Subjects
0301 basic medicine ,Cancer Research ,biology ,Chemistry ,CD3 ,medicine.medical_treatment ,T cell ,Immunotherapy ,medicine.disease ,03 medical and health sciences ,Cytokine release syndrome ,030104 developmental biology ,0302 clinical medicine ,Cytokine ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,medicine ,Cancer research ,Cytotoxic T cell ,Interleukin-3 receptor ,Cytotoxicity - Abstract
Introduction: Depletion of CD123 over-expressing malignant cells provides a potential new treatment option which may improve patient outcomes in several hematological malignancies. CD123 is over-expressed in AML, MDS, ALL, CML, HCL and BPDCN and infrequently expressed by normal cells making it an attractive target which is being pursued using a number of different approaches including T-cell engaging immunotherapy. Cytokine release syndrome is a significant concern with T-cell activating therapeutics which has led to severe complications in clinical trials. We have developed APVO436, a bispecific anti-CD123 x anti-CD3 ADAPTIR molecule for redirecting T-cell cytotoxicity to CD123-expressing tumor cells. A potential advantage of the ADAPTIR platform is reduced cytokine release upon T-cell engagement compared to other formats (Mol Cancer Ther. 2016 Sep;15(9):2155-65). Here we present in vitro and in vivo activity of APVO436 and compare the activity of APVO436 to another anti-CD123 x anti-CD3 bispecific containing the amino acid sequence of MGD006.Methods: Binding, T-cell activation and proliferation were assessed using multi-color flow cytometry. Cytotoxic activity was determined using chromium release assays and flow cytometry. PBMC samples were obtained from normal donors and AML patients. In vivo studies were performed using NSG mice transplanted with human PBMC's. The CD123 and CD3 binding domain sequences for flotuzumab (MGD006) were obtained from patent W02015026892 engineered in Macrogenic's dual-affinity re-targeting format as reported in Sci Transl Med. 2015 May 27;7(289):289ra82.Results: Dose-dependent cytotoxicity of CD123 expressing tumor cell lines and primary AML cells was induced by APVO436 at low effector to target ratios, accompanied by T-cell activation and proliferation. APVO436 induced significantly lower levels of several T-cell cytokines including IFNγ, IL-2, and TNFα compared to the molecule in the dual-affinity re-targeting format. In vivo, APVO436 significantly reduced established tumor burden in xenograft murine models.Conclusions: APVO436 potently induces T-cell activation, proliferation and CD123+ cell depletion with AML and normal donor samples and CD123 expressing tumor cell lines with limited levels of T-cell cytokine release compared to another CD123 x CD3 targeting bispecific format suggesting a potential safety advantage. APVO436 inhibits tumor-growth in sub-cutaneous tumor models with IV-implanted human T cells, indicating migration and engagement of T cells at the tumor site. These data are supportive of further investigation of APVO436 as a potential treatment option for AML and other hematological malignancies. GLP toxicology studies have been completed in non-human primates and APVO436 is advancing to clinical testing. Citation Format: Michael R. Comeau, Robert E. Miller, Robert Bader, Rebecca Gottschalk, Mollie Daugherty, Toddy Sewell, Lynda Misher, Lara Parr, Melissa DeFrancesco, David Bienvenue, Catherine J. McMahan, Gabriela H. Hoyos, Jane A. Gross. APVO436, a bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecule for redirected T-cell cytotoxicity, induces potent T-cell activation, proliferation and cytotoxicity with limited cytokine release [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 1786.
- Published
- 2018
- Full Text
- View/download PDF
8. Abstract 597: Bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecules for redirected T-cell cytotoxicity in hematological malignancies
- Author
-
Maria M. Dasovich, Robert E. Miller, Michael R. Comeau, Peter Pavlik, Megan Aguilar, Gabriela H. Hoyos, Hang Fang, Danielle Mitchell, Starrla Johnson, Nicole Zhang, Jane A. Gross, Rebecca Gottschalk, Brian Woodruff, Mollie Daugherty, Gary Li, Lynda Misher, Catherine J. McMahan, John W. Blankenship, Jeannette Bannink, Robert Bader, Toddy Sewell, Lara Parr, and David Bienvenue
- Subjects
Cancer Research ,biology ,Chemistry ,CD3 ,Fc receptor ,In vitro ,medicine.anatomical_structure ,Oncology ,Cell culture ,In vivo ,Immunology ,biology.protein ,Cancer research ,medicine ,Cytotoxic T cell ,Bone marrow ,Stem cell - Abstract
Introduction: CD123 is a component of the IL-3 receptor expressed in several hematological malignancies including AML, ALL, HCL, and MDS. CD123 is a compelling target in AML due to its overexpression on AML blasts as well as leukemic stem cells, which are thought to be resistant to chemotherapy and may be responsible for relapse of disease following treatment. While CD123 is expressed by some normal leukocyte populations in circulation and hematopoietic progenitor cells in the bone marrow, the low frequency of expression on normal cell types provides a therapeutic window for targeting CD123 in tumor settings with the potential for durable response and reversible side effects. We have developed bispecific anti-CD123 x anti-CD3 ADAPTIR molecules APVO436 and APVO437 for redirecting T-cell cytotoxicity to CD123-expressing tumor cells. Results are presented that examine the in vitro and in vivo activity of these molecules in preclinical models of AML. Methods: APVO436 and APVO437 proteins were expressed in CHO cells. Affinity SPR studies were performed using recombinant CD123-ectodomain. In vitro functional studies were conducted with CD123+ AML tumor cell lines and primary human and cynomolgus macaque T-cell populations. Cytotoxic activity was determined using chromium release assays. On-cell binding, T-cell activation and proliferation were assessed using multi-color flow cytometry. Pharmacokinetic parameters were determined in BALB/c mice using a single IV dose of approximately 10 mg/kg. In vivo studies to examine tumor growth inhibition activity were performed with NOD/SCID mice co-implanted subcutaneously with AML tumor cells and human T-cells followed by treatment with APVO436 or APVO437. Tumor growth was assessed by measuring tumor volume and Bioluminescent Imaging. Results: APVO436 and APVO437 bound human CD123 protein with high affinity and binding to CD123 and CD3 expressing cell lines was confirmed by flow cytometry. Both APVO436 and APVO437 induced concentration-dependent lysis of CD123+ AML cell lines with primary human effector T-cells, accompanied by T-cell activation and proliferation. Comparable redirected T-cell cytotoxicity function was observed using primary cynomolgus macaque T cells. These activities were dependent on the expression of CD123 by the tumor target cells. APVO436 and APVO437 demonstrated an extended elimination half-life in mouse serum, typical of molecules capable of binding the neo-natal Fc receptor. In vivo, growth of AML tumor cells was inhibited by treatment with low doses of APVO436 and APVO437, significantly improving host survival. Conclusion: Taken together these data demonstrate potent in vitro and in vivo activity of APVO436 and APVO437 against CD123 expressing tumor cells and are supportive of further investigation of this approach as a potential treatment option for AML and other hematological malignancies. Citation Format: Michael R. Comeau, Danielle Mitchell, Rebecca Gottschalk, Lynda Misher, Mollie Daugherty, Lara Parr, Peter Pavlik, Brian Woodruff, Hang Fang, Megan Aguilar, Jeannette Bannink, Starrla Johnson, Gary Li, Robert E. Miller, Robert Bader, Nicole Zhang, Toddy Sewell, Maria Dasovich, Gabriela H. Hoyos, John W. Blankenship, Catherine McMahan, David Bienvenue, Jane A. Gross. Bispecific anti-CD123 x anti-CD3 ADAPTIR™ molecules for redirected T-cell cytotoxicity in hematological malignancies [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 597. doi:10.1158/1538-7445.AM2017-597
- Published
- 2017
- Full Text
- View/download PDF
9. Abstract 4995: anti-ROR1 x anti-CD3 ADAPTIR™ molecule, ES425, redirects T-cell cytotoxicity and inhibits tumor growth in preclinical models of triple-negative breast cancer
- Author
-
John W. Blankenship, Hang Fang, Megan Aguilar, Brian Woodruff, Mollie Daugherty, Nicole Zhang, Jeannette Bannink, Catherine J. McMahan, Padma Ravikumar, Robert E. Miller, Maria M. Dasovich, Lara Parr, Robert Bader, Carina Xu, Gabriela H. Hoyos, Jane A. Gross, Philip Tan, Lynda Misher, Danielle Mitchell, and David Bienvenue
- Subjects
0301 basic medicine ,Cancer Research ,medicine.diagnostic_test ,business.industry ,Cancer ,medicine.disease ,Peripheral blood mononuclear cell ,Flow cytometry ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,Cell culture ,In vivo ,030220 oncology & carcinogenesis ,Immunology ,Cancer research ,medicine ,Cytotoxic T cell ,Oncofetal antigen ,Cytotoxicity ,business - Abstract
Background: Effective treatment of metastatic, triple-negative breast cancer (TNBC) remains a highly unmet medical need. We have developed ES425, a bispecific ADAPTIR™ (modular protein technology) molecule that redirects T-cell cytotoxicity to tumor cells expressing ROR1 (receptor tyrosine kinase-like orphan receptor 1), an oncofetal antigen expressed on TNBC and other malignancies. Results are presented for studies run to examine in vitro and in vivo activity of ES425 in preclinical models of TNBC. Materials and Methods: Target-dependent cytotoxic activity was examined in vitro by treating ROR1(+) cell lines and ROR1(−) cell lines with ES425 in the presence of purified human T cells or human peripheral blood mononuclear cells (PBMCs). Cytotoxic activity was determined using chromium release assays. T cells were assessed for activation and proliferation using multi-color flow cytometry. Pharmacokinetics of ES425 in NOD/SCID gamma (NSG) mice was determined using single intravenous dose of approximately 10 mg/kg. Serum concentrations at time points ranging from 15 minutes to 504 hours were used to calculate the terminal elimination half-life of ES425. To assess activity in vivo, NOD/SCID mice were implanted subcutaneously with the ROR1(+) TNBC tumor cell line MDA-MB-231 and purified human T cells and treated with ES425. This model was run twice with T cells from different human donors. Tumor growth was assessed by measuring tumor volume. Results: ES425 efficiently redirected T cell cytotoxicity against ROR1(+) cell lines at low picomolar concentrations in vitro. Cytotoxic activity was dependent on expression of ROR1 by the target cells. T cells were activated and proliferated in response to ES425 in the presence of ROR1(+) target cells; proliferation was not observed in response to ROR1(−) cells. In vivo, pharmacokinetic analysis showed a serum half-life of approximately 7 days in NSG mice, and ES425 inhibited growth of MDA-MB-231 tumors in mouse xenografts. Repeat experiments showed similar inhibition of tumor growth and an improvement in overall survival. Conclusions: These studies show that ES425 may be an efficient agent for redirecting T cell cytotoxicity in preclinical TNBC models and merits investigation as a potential therapeutic in TNBC and other malignancies. Citation Format: John W. Blankenship, Lynda Misher, Danielle Mitchell, Nicole Zhang, Philip Tan, Gabriela H. Hoyos, Padma Ravikumar, Robert Bader, Catherine J. McMahan, Robert E. Miller, Jeannette Bannink, Hang Fang, Lara Parr, Maria Dasovich, David Bienvenue, Megan Aguilar, Carina Xu, Mollie Daugherty, Brian Woodruff, Jane A. Gross. anti-ROR1 x anti-CD3 ADAPTIR™ molecule, ES425, redirects T-cell cytotoxicity and inhibits tumor growth in preclinical models of triple-negative breast cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 4995.
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.