84 results on '"Klara Totpal"'
Search Results
2. Data from Relative Target Affinities of T-Cell–Dependent Bispecific Antibodies Determine Biodistribution in a Solid Tumor Mouse Model
- Author
-
C. Andrew Boswell, Teemu T. Junttila, Saileta Prabhu, Mark S. Dennis, Sean B. Joseph, Kedan Lin, Klara Totpal, Robyn Clark, Maria Hristopoulos, Jason Ho, Dionysos Slaga, Jeffrey Eastham-Anderson, Ji Li, Amy A. Lo, Nene Takahashi, and Danielle Mandikian
- Abstract
Anti-HER2/CD3, a T-cell–dependent bispecific antibody (TDB) construct, induces T-cell–mediated cell death in cancer cells expressing HER2 by cross-linking tumor HER2 with CD3 on cytotoxic T cells, thereby creating a functional cytolytic synapse. TDB design is a very challenging process that requires consideration of multiple parameters. Although therapeutic antibody design strategy is commonly driven by striving for the highest attainable antigen-binding affinity, little is known about how the affinity of each TDB arm can affect the targeting ability of the other arm and the consequent distribution and efficacy. To our knowledge, no distribution studies have been published using preclinical models wherein the T-cell–targeting arm of the TDB is actively bound to T cells. We used a combined approach involving radiochemistry, invasive biodistribution, and noninvasive single-photon emission tomographic (SPECT) imaging to measure TDB distribution and catabolism in transgenic mice with human CD3ϵ expression on T cells. Using CD3 affinity variants, we assessed the impact of CD3 affinity on short-term pharmacokinetics, tissue distribution, and cellular uptake. Our experimental approach determined the relative effects of (i) CD3 targeting to normal tissues, (ii) HER2 targeting to HER2-expressing tumors, and (iii) relative HER2/CD3 affinity, all as critical drivers for TDB distribution. We observed a strong correlation between CD3 affinity and distribution to T-cell–rich tissues, with higher CD3 affinity reducing systemic exposure and shifting TDB distribution away from tumor to T-cell–containing tissues. These observations have important implications for clinical translation of bispecific antibodies for cancer immunotherapy. Mol Cancer Ther; 17(4); 776–85. ©2018 AACR.
- Published
- 2023
3. Supplementary Data from Anti-LYPD1/CD3 T-Cell-Dependent Bispecific Antibody for the Treatment of Ovarian Cancer
- Author
-
Teemu T. Junttila, Yan Wu, Jian Payandeh, C. Andrew Boswell, Genee Lee, Klara Totpal, James Ziai, Diego Ellerman, Dionysos Slaga, Elaine Mai, Daniel Bravo-Perez, Meric Ovacik, Hao Cai, Thinh Pham, Debra Dunlap, Kathy Hötzel, Wei-Ching Liang, Dorothee Nickles, Robyn Clark, Maria Hristopoulos, Danielle Mandikian, Ji Li, Jennifer Johnston, and Amy A. Lo
- Abstract
Supplementary Data
- Published
- 2023
4. Supplemental Methods and Figures 1-5 from Relative Target Affinities of T-Cell–Dependent Bispecific Antibodies Determine Biodistribution in a Solid Tumor Mouse Model
- Author
-
C. Andrew Boswell, Teemu T. Junttila, Saileta Prabhu, Mark S. Dennis, Sean B. Joseph, Kedan Lin, Klara Totpal, Robyn Clark, Maria Hristopoulos, Jason Ho, Dionysos Slaga, Jeffrey Eastham-Anderson, Ji Li, Amy A. Lo, Nene Takahashi, and Danielle Mandikian
- Abstract
Supplemental Methods, followed by Supplemental Figures S1, S2, S3, S4 and S5. Supplementary Fig. S1 In vitro binding of TDBs to HER2 and CD3 expressing cell lines. Supplementary Fig. S2 HER2 expression in model cell lines in vitro and tumors in vivo. Supplementary Fig. S3 HER2, CD3� and MECA-32 staining and vascular characterization of HER2 positive and negative tumors. Supplementary Fig. S4 T cell infiltration and CD3 expression in murine CT26 and CT26-HER2 tumors. Supplementary Fig. S5 Pharmacokinetic data from whole blood and cell pellet fractions
- Published
- 2023
5. Data from Anti-LYPD1/CD3 T-Cell-Dependent Bispecific Antibody for the Treatment of Ovarian Cancer
- Author
-
Teemu T. Junttila, Yan Wu, Jian Payandeh, C. Andrew Boswell, Genee Lee, Klara Totpal, James Ziai, Diego Ellerman, Dionysos Slaga, Elaine Mai, Daniel Bravo-Perez, Meric Ovacik, Hao Cai, Thinh Pham, Debra Dunlap, Kathy Hötzel, Wei-Ching Liang, Dorothee Nickles, Robyn Clark, Maria Hristopoulos, Danielle Mandikian, Ji Li, Jennifer Johnston, and Amy A. Lo
- Abstract
Ovarian cancer is a diverse class of tumors with very few effective treatment options and suboptimal response rates in early clinical studies using immunotherapies. Here we describe LY6/PLAUR domain containing 1 (LYPD1) as a novel target for therapeutic antibodies for the treatment of ovarian cancer. LYPD1 is broadly expressed in both primary and metastatic ovarian cancer with ∼70% prevalence in the serous cancer subset. Bispecific antibodies targeting CD3 on T cells and a tumor antigen on cancer cells have demonstrated significant clinical activity in hematologic cancers. We have developed an anti-LYPD1/CD3 T-cell-dependent bispecific antibody (TDB) to redirect T-cell responses to LYPD1 expressing ovarian cancer. Here we characterize the nonclinical pharmacology of anti-LYPD1/CD3 TDB and show induction of a robust polyclonal T-cell activation and target dependent killing of LYPD1 expressing ovarian cancer cells resulting in efficient in vivo antitumor responses in PBMC reconstituted immune-deficient mice and human CD3 transgenic mouse models. Anti-LYPD1/CD3 TDB is generally well tolerated at high-dose levels in mice, a pharmacologically relevant species, and showed no evidence of toxicity or damage to LYPD1 expressing tissues.
- Published
- 2023
6. Supplementary Fig. S1 from Antixenograft tumor activity of a humanized anti-insulin-like growth factor-I receptor monoclonal antibody is associated with decreased AKT activation and glucose uptake
- Author
-
Jiping Zha, Avi Ashkenazi, Jean-Philippe Stephan, Yan Wu, An Song, Sang Hoon Lee, Congfen Li, Sarajane Ross, Janet Tien, Klaus Hoeflich, Kenji Kozuka, Alexander Vanderbilt, Siao Ping Tsai, Wei-Ching Liang, Christine Tan, Zora Modrusan, Zhijun Tang, Jihong Yang, Simon Williams, Klara Totpal, Mark R. Lackner, Gail Phillips, Suzie J. Scales, Jennifer Batson, Yifan Mao, and Yonglei Shang
- Abstract
Supplementary Fig. S1 from Antixenograft tumor activity of a humanized anti-insulin-like growth factor-I receptor monoclonal antibody is associated with decreased AKT activation and glucose uptake
- Published
- 2023
7. Data from IRE1α Disruption in Triple-Negative Breast Cancer Cooperates with Antiangiogenic Therapy by Reversing ER Stress Adaptation and Remodeling the Tumor Microenvironment
- Author
-
Avi Ashkenazi, Peter Walter, Hartmut Koeppen, Joachim Rudolph, Zora Modrusan, Margaret Solon, Monika Dohse, Kathryn Mesh, Lisa M. Crocker, Klara Totpal, Mark Merchant, Ehud Segal, David Kan, David A. Lawrence, Anna Shemorry, Scot A. Marsters, Thomas D. Wu, Ofer Guttman, Mike Reichelt, Adrien Le Thomas, and Jonathan M. Harnoss
- Abstract
Cancer cells exploit the unfolded protein response (UPR) to mitigate endoplasmic reticulum (ER) stress caused by cellular oncogene activation and a hostile tumor microenvironment (TME). The key UPR sensor IRE1α resides in the ER and deploys a cytoplasmic kinase–endoribonuclease module to activate the transcription factor XBP1s, which facilitates ER-mediated protein folding. Studies of triple-negative breast cancer (TNBC)—a highly aggressive malignancy with a dismal posttreatment prognosis—implicate XBP1s in promoting tumor vascularization and progression. However, it remains unknown whether IRE1α adapts the ER in TNBC cells and modulates their TME, and whether IRE1α inhibition can enhance antiangiogenic therapy—previously found to be ineffective in patients with TNBC. To gauge IRE1α function, we defined an XBP1s-dependent gene signature, which revealed significant IRE1α pathway activation in multiple solid cancers, including TNBC. IRE1α knockout in TNBC cells markedly reversed substantial ultrastructural expansion of their ER upon growth in vivo. IRE1α disruption also led to significant remodeling of the cellular TME, increasing pericyte numbers while decreasing cancer-associated fibroblasts and myeloid-derived suppressor cells. Pharmacologic IRE1α kinase inhibition strongly attenuated growth of cell line–based and patient-derived TNBC xenografts in mice and synergized with anti-VEGFA treatment to cause tumor stasis or regression. Thus, TNBC cells critically rely on IRE1α to adapt their ER to in vivo stress and to adjust the TME to facilitate malignant growth. TNBC reliance on IRE1α is an important vulnerability that can be uniquely exploited in combination with antiangiogenic therapy as a promising new biologic approach to combat this lethal disease.Significance:Pharmacologic IRE1α kinase inhibition reverses ultrastructural distension of the ER, normalizes the tumor vasculature, and remodels the cellular TME, attenuating TNBC growth in mice.
- Published
- 2023
8. Supplementary Table S1 from Antixenograft tumor activity of a humanized anti-insulin-like growth factor-I receptor monoclonal antibody is associated with decreased AKT activation and glucose uptake
- Author
-
Jiping Zha, Avi Ashkenazi, Jean-Philippe Stephan, Yan Wu, An Song, Sang Hoon Lee, Congfen Li, Sarajane Ross, Janet Tien, Klaus Hoeflich, Kenji Kozuka, Alexander Vanderbilt, Siao Ping Tsai, Wei-Ching Liang, Christine Tan, Zora Modrusan, Zhijun Tang, Jihong Yang, Simon Williams, Klara Totpal, Mark R. Lackner, Gail Phillips, Suzie J. Scales, Jennifer Batson, Yifan Mao, and Yonglei Shang
- Abstract
Supplementary Table S1 from Antixenograft tumor activity of a humanized anti-insulin-like growth factor-I receptor monoclonal antibody is associated with decreased AKT activation and glucose uptake
- Published
- 2023
9. Supplementary Data from IRE1α Disruption in Triple-Negative Breast Cancer Cooperates with Antiangiogenic Therapy by Reversing ER Stress Adaptation and Remodeling the Tumor Microenvironment
- Author
-
Avi Ashkenazi, Peter Walter, Hartmut Koeppen, Joachim Rudolph, Zora Modrusan, Margaret Solon, Monika Dohse, Kathryn Mesh, Lisa M. Crocker, Klara Totpal, Mark Merchant, Ehud Segal, David Kan, David A. Lawrence, Anna Shemorry, Scot A. Marsters, Thomas D. Wu, Ofer Guttman, Mike Reichelt, Adrien Le Thomas, and Jonathan M. Harnoss
- Abstract
Supplementary Data, including Supplementary Materials and Methods, References for Supplementary Data, and Figs. S1 to S5 including Figure Legends
- Published
- 2023
10. Data from IFNγ-induced Chemokines Are Required for CXCR3-mediated T-Cell Recruitment and Antitumor Efficacy of Anti-HER2/CD3 Bispecific Antibody
- Author
-
Teemu T. Junttila, Kevin B. Walsh, Melissa R. Junttila, Klara Totpal, James Ziai, Alfonso Arrazate, Patricia De Almeida, Aurelie Herault, Judy Mak, Ryan Ybarra, and Ji Li
- Abstract
Purpose:The response to cancer immune therapy is dependent on endogenous tumor-reactive T cells. To bypass this requirement, CD3-bispecific antibodies have been developed to induce a polyclonal T-cell response against the tumor. Anti-HER2/CD3 T-cell–dependent bispecific (TDB) antibody is highly efficacious in the treatment of HER2-overexpressing tumors in mice. Efficacy and immunologic effects of anti-HER2/CD3 TDB were investigated in mammary tumor model with very few T cells prior treatment. We further describe the mechanism for TDB-induced T-cell recruitment to tumors.Experimental Design:The immunologic effects and the mechanism of CD3-bispecific antibody-induced T-cell recruitment into spontaneous HER2-overexpressing mammary tumors was studied using human HER2 transgenic, immunocompetent mouse models.Results:Anti-HER2/CD3 TDB treatment induced an inflammatory response in tumors converting them from poorly infiltrated to an inflamed, T-cell abundant, phenotype. Multiple mechanisms accounted for the TDB-induced increase in T cells within tumors. TDB treatment induced CD8+ T-cell proliferation. T cells were also actively recruited post-TDB treatment by IFNγ-dependent T-cell chemokines mediated via CXCR3. This active T-cell recruitment by TDB-induced chemokine signaling was the dominant mechanism and necessary for the therapeutic activity of anti-HER2/CD3 TDB.Conclusions:In summary, we demonstrate that the activity of anti-HER2/CD3 TDB was not dependent on high-level baseline T-cell infiltration. Our results suggest that anti-HER2/CD3 TDB may be efficacious in patients and indications that respond poorly to checkpoint inhibitors. An active T-cell recruitment mediated by TDB-induced chemokine signaling was the major mechanism for T-cell recruitment.
- Published
- 2023
11. Data from Antixenograft tumor activity of a humanized anti-insulin-like growth factor-I receptor monoclonal antibody is associated with decreased AKT activation and glucose uptake
- Author
-
Jiping Zha, Avi Ashkenazi, Jean-Philippe Stephan, Yan Wu, An Song, Sang Hoon Lee, Congfen Li, Sarajane Ross, Janet Tien, Klaus Hoeflich, Kenji Kozuka, Alexander Vanderbilt, Siao Ping Tsai, Wei-Ching Liang, Christine Tan, Zora Modrusan, Zhijun Tang, Jihong Yang, Simon Williams, Klara Totpal, Mark R. Lackner, Gail Phillips, Suzie J. Scales, Jennifer Batson, Yifan Mao, and Yonglei Shang
- Abstract
The insulin-like growth factor (IGF) system consists of two ligands (IGF-I and IGF-II), which both signal through IGF-I receptor (IGF-IR) to stimulate proliferation and inhibit apoptosis, with activity contributing to malignant growth of many types of human cancers. We have developed a humanized, affinity-matured anti-human IGF-IR monoclonal antibody (h10H5), which binds with high affinity and specificity to the extracellular domain. h10H5 inhibits IGF-IR-mediated signaling by blocking IGF-I and IGF-II binding and by inducing cell surface receptor down-regulation via internalization and degradation, with the extracellular and intracellular domains of IGF-IR being differentially affected by the proteasomal and lysosomal inhibitors. In vitro, h10H5 exhibits antiproliferative effects on cancer cell lines. In vivo, h10H5 shows single-agent antitumor efficacy in human SK-N-AS neuroblastoma and SW527 breast cancer xenograft models and even greater efficacy in combination with the chemotherapeutic agent docetaxel or an anti–vascular endothelial growth factor antibody. Antitumor activity of h10H5 is associated with decreased AKT activation and glucose uptake and a 316-gene transcription profile with significant changes involving DNA metabolic and cell cycle machineries. These data support the clinical testing of h10H5 as a biotherapeutic for IGF-IR-dependent human tumors and furthermore illustrate a new method of monitoring its activity noninvasively in vivo via 2-fluoro-2-deoxy-d-glucose-positron emission tomography imaging. [Mol Cancer Ther 2008;7(9):2599–608]
- Published
- 2023
12. Supplemental Figure 1-5 from IFNγ-induced Chemokines Are Required for CXCR3-mediated T-Cell Recruitment and Antitumor Efficacy of Anti-HER2/CD3 Bispecific Antibody
- Author
-
Teemu T. Junttila, Kevin B. Walsh, Melissa R. Junttila, Klara Totpal, James Ziai, Alfonso Arrazate, Patricia De Almeida, Aurelie Herault, Judy Mak, Ryan Ybarra, and Ji Li
- Abstract
Fig. S1. Subset analysis of tumor infiltrating and splenic CD8+ T cells after treatment with anti-HER2/CD3 TDB; Fig. S2. Anti-HER2/CD3 TDB increases intra-tumoral lymphocyte infiltration in Fo5 allograft model of HER2 overexpressing breast cancer within immunocompetent mice; Fig. S3. Anti-HER2/CD3 treatment did not induce peripheral CD8+ T cell proliferation; Fig. S4. Anti-HER2/CD3 TDB induced acute release of pro-inflammatory cytokines in serum; Fig. S5. Induction of CXCR3 ligands within Fo5 tumors in response to anti-HER2/CD3.
- Published
- 2023
13. Supplementary Data 1 from Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer
- Author
-
Mark X. Sliwkowski, Robert F. Kelley, Klara Totpal, Gloria Meng, Tomasz Baginski, Oliver Pabonan, Lisa Crocker, Julie Theriault, Yan Xin, Yanmei Lu, Christine Olsson, Kathryn Parsons, and Teemu T. Junttila
- Abstract
Supplementary Data 1 from Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer
- Published
- 2023
14. Supplemental Figures 1 - 8 from Antitumor Efficacy of a Bispecific Antibody That Targets HER2 and Activates T Cells
- Author
-
Allen Ebens, Klara Totpal, Robert Kelley, Mark S. Dennis, Melissa R. Junttila, Justin M. Scheer, Bryan Irving, Andrew Polson, Christoph Spiess, Eric Stefanich, Gail Lewis Phillips, Elizabeth Luis, Judy Young, Guangmin Li, Mary Mathieu, Yijin Li, Bu-Er Wang, Diego Ellerman, Robyn Clark, Maria Hristopoulos, Jennifer Johnston, Ji Li, and Teemu T. Junttila
- Abstract
Supplemental Fig. 1: Characteristics of T cell activation and killing induced by HER2-TDB. Supplemental Fig 2: Activation of T cells by HER2-TDB induces T cell proliferation. Supplemental Figure 3. Activity of HER2-TDB in NOD-SCID mice is dependent on human PBMCs. Supplemental Figure 4. TDB mediated killing by CD3-TG splenic T cells. Supplemental Figure 5. Effect of 4D5 on established mammary tumors in MMTV-huHER2 transgenic mice. CD3-TG T cells express both mouse and human CD3 on approximately 50% level of respective Balb/c mouse or human T cells. Supplemental Figure 6. Anti-tumor activity of HER2-TDB is T cell dependent. Supplemental Figure 7. T cells in CT26-HER2 tumors display CD69 activation marker. Supplemental Figure 8. CT-26-HER2 tumor infiltrating T cells express PD-1. CT-26-HER2 tumor cells express PD-L1.
- Published
- 2023
15. Data from Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer
- Author
-
Mark X. Sliwkowski, Robert F. Kelley, Klara Totpal, Gloria Meng, Tomasz Baginski, Oliver Pabonan, Lisa Crocker, Julie Theriault, Yan Xin, Yanmei Lu, Christine Olsson, Kathryn Parsons, and Teemu T. Junttila
- Abstract
The enhancement of immune effector functions has been proposed as a potential strategy for increasing the efficacy of therapeutic antibodies. Here, we show that removing fucose from trastuzumab (Herceptin) increased its binding to FcγRIIIa, enhanced antibody-dependent cell-mediated cytotoxicity, and more than doubled the median progression-free survival when compared with conventional trastuzumab in treating preclinical models of HER2-amplified breast cancer. Our results show that afucosylated trastuzumab has superior efficacy in treating in vivo models of HER2-amplified breast cancer and support the development of effector function–enhanced antibodies for solid tumor therapy. Cancer Res; 70(11); 4481–9. ©2010 AACR.
- Published
- 2023
16. Supplementary Figure Legends 1-3 from Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer
- Author
-
Mark X. Sliwkowski, Robert F. Kelley, Klara Totpal, Gloria Meng, Tomasz Baginski, Oliver Pabonan, Lisa Crocker, Julie Theriault, Yan Xin, Yanmei Lu, Christine Olsson, Kathryn Parsons, and Teemu T. Junttila
- Abstract
Supplementary Figure Legends 1-3 from Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer
- Published
- 2023
17. Supplementary Data 2 from Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer
- Author
-
Mark X. Sliwkowski, Robert F. Kelley, Klara Totpal, Gloria Meng, Tomasz Baginski, Oliver Pabonan, Lisa Crocker, Julie Theriault, Yan Xin, Yanmei Lu, Christine Olsson, Kathryn Parsons, and Teemu T. Junttila
- Abstract
Supplementary Data 2 from Superior In vivo Efficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer
- Published
- 2023
18. Novel Anti-LY6G6D/CD3 T-Cell–Dependent Bispecific Antibody for the Treatment of Colorectal Cancer
- Author
-
Peiyin Wang, Liping L. Sun, Robyn Clark, Maria Hristopoulos, Cecilia P.C. Chiu, Michael Dillon, WeiYu Lin, Amy A. Lo, Sreedevi Chalsani, Meghna Das Thakur, Kristin M. Zimmerman Savill, Lionel Rougé, Patrick Lupardus, Robert Piskol, Bushra Husain, Diego Ellerman, Vittal Shivva, Steven R. Leong, Meric Ovacik, Klara Totpal, Yan Wu, Christoph Spiess, Genee Lee, Douglas D. Leipold, and Andrew G. Polson
- Subjects
Mice ,Cancer Research ,Oncology ,T-Lymphocytes ,Antibodies, Bispecific ,Animals ,Humans ,Immunoglobulins ,Microsatellite Instability ,Colorectal Neoplasms ,Immune Checkpoint Inhibitors ,digestive system diseases - Abstract
New therapeutics and combination regimens have led to marked clinical improvements for the treatment of a subset of colorectal cancer. Immune checkpoint inhibitors have shown clinical efficacy in patients with mismatch-repair–deficient or microsatellite instability–high (MSI-H) metastatic colorectal cancer (mCRC). However, patients with microsatellite-stable (MSS) or low levels of microsatellite instable (MSI-L) colorectal cancer have not benefited from these immune modulators, and the survival outcome remains poor for the majority of patients diagnosed with mCRC. In this article, we describe the discovery of a novel T-cell–dependent bispecific antibody (TDB) targeting tumor-associated antigen LY6G6D, LY6G6D-TDB, for the treatment of colorectal cancer. RNAseq analysis showed that LY6G6D was differentially expressed in colorectal cancer with high prevalence in MSS and MSI-L subsets, whereas LY6G6D expression in normal tissues was limited. IHC confirmed the elevated expression of LY6G6D in primary and metastatic colorectal tumors, whereas minimal or no expression was observed in most normal tissue samples. The optimized LY6G6D-TDB, which targets a membrane-proximal epitope of LY6G6D and binds to CD3 with high affinity, exhibits potent antitumor activity both in vitro and in vivo. In vitro functional assays show that LY6G6D-TDB–mediated T-cell activation and cytotoxicity are conditional and target dependent. In mouse xenograft tumor models, LY6G6D-TDB demonstrates antitumor efficacy as a single agent against established colorectal tumors, and enhanced efficacy can be achieved when LY6G6D-TDB is combined with PD-1 blockade. Our studies provide evidence for the therapeutic potential of LY6G6D-TDB as an effective treatment option for patients with colorectal cancer.
- Published
- 2022
19. T cell‐dependent bispecific antibodies alter organ‐specific endothelial cell–T cell interaction
- Author
-
Patricia Himmels, Thi Thu Thao Nguyen, Maresa Caunt Mitzner, Alfonso Arrazate, Stacey Yeung, Jeremy Burton, Robyn Clark, Klara Totpal, Raj Jesudason, Angela Yang, Margaret Solon, Jeffrey Eastham, Zora Modrusan, Joshua D Webster, Amy A Lo, Robert Piskol, and Weilan Ye
- Subjects
Genetics ,Molecular Biology ,Biochemistry - Abstract
Preclinical and clinical studies demonstrate that T cell-dependent bispecific antibodies (TDBs) induce systemic changes in addition to tumor killing, leading to adverse events. Here, we report an in-depth characterization of acute responses to TDBs in tumor-bearing mice. Contrary to modest changes in tumors, rapid and substantial lymphocyte accumulation and endothelial cell (EC) activation occur around large blood vessels in normal organs including the liver. We hypothesize that organ-specific ECs may account for the differential responses in normal tissues and tumors, and we identify a list of genes selectively upregulated by TDB in large liver vessels. Using one of the genes as an example, we demonstrate that CD9 facilitates ICAM-1 to support T cell-EC interaction in response to soluble factors released from a TDB-mediated cytotoxic reaction. Our results suggest that multiple factors may cooperatively promote T cell infiltration into normal organs as a secondary response to TDB-mediated tumor killing. These data shed light on how different vascular beds respond to cancer immunotherapy and may help improve their safety and efficacy.
- Published
- 2023
20. The Tumor Suppressor BAP1 Regulates the Hippo Pathway in Pancreatic Ductal Adenocarcinoma
- Author
-
Allen G. Cai, Jeffrey Eastham-Anderson, Wyne P. Lee, Thijs J. Hagenbeek, Janet Tao, Ho-June Lee, Joshua D. Webster, Michele Carbone, Klara Totpal, Xiumin Wu, Trang H. Pham, Boris C. Bastian, Dwight Barnes, Anwesha Dey, Matthew T. Chang, Xu Chen, Rajkumar Noubade, and Christopher Tran
- Subjects
0301 basic medicine ,Cancer Research ,Pancreatic Intraepithelial Neoplasia ,Adenocarcinoma ,Protein Serine-Threonine Kinases ,Biology ,Deubiquitinating enzyme ,law.invention ,03 medical and health sciences ,0302 clinical medicine ,law ,medicine ,Humans ,Hippo Signaling Pathway ,Mesothelioma ,Hippo signaling pathway ,BAP1 ,Tumor Suppressor Proteins ,medicine.disease ,Pancreatic Neoplasms ,030104 developmental biology ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Cancer research ,biology.protein ,Suppressor ,Ubiquitin Thiolesterase ,Signal Transduction - Abstract
The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk for mesothelioma and melanocytic tumors. Here, we show that pancreatic intraepithelial neoplasia driven by oncogenic mutant KrasG12D progressed to pancreatic adenocarcinoma in the absence of BAP1. The Hippo pathway was deregulated in BAP1-deficient pancreatic tumors, with the tumor suppressor LATS exhibiting enhanced ubiquitin-dependent proteasomal degradation. Therefore, BAP1 may limit tumor progression by stabilizing LATS and thereby promoting activity of the Hippo tumor suppressor pathway. Significance: BAP1 is mutated in a broad spectrum of tumors. Pancreatic Bap1 deficiency causes acinar atrophy but combines with oncogenic Ras to produce pancreatic tumors. BAP1-deficient tumors exhibit deregulation of the Hippo pathway. See related commentary by Brekken, p. 1624
- Published
- 2020
21. Abstract P6-17-20: Withdrawn
- Author
-
Robyn Clark, Rodney A. Prell, Jennifer Johnston, G Lee, Ji Li, Christoph Spiess, Klara Totpal, Teemu T. Junttila, Diego Ellerman, Rajesh Vij, Maria Hristopoulos, Elaine Mai, N Lombana, Amy L. Shelton, K Gadkar, Aa Lo, Dion Slaga, and JT Koerber
- Subjects
Cancer Research ,Oncology - Abstract
This abstract was withdrawn by the authors. Citation Format: Junttila TT, Ellerman D, Lombana N, Hristopoulos M, Clark R, Li J, Vij R, Koerber JT, Johnston J, Shelton A, Mai E, Gadkar K, Lo AA, Totpal K, Prell R, Lee G, Spiess C, Slaga D. Withdrawn [abstract]. In: Proceedings of the 2018 San Antonio Breast Cancer Symposium; 2018 Dec 4-8; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2019;79(4 Suppl):Abstract nr P6-17-20.
- Published
- 2019
22. Anti-LYPD1/CD3 T-Cell-Dependent Bispecific Antibody for the Treatment of Ovarian Cancer
- Author
-
Amy C. Y. Lo, C. Andrew Boswell, Teemu T. Junttila, Thinh Pham, Genee Lee, Jian Payandeh, Danielle Mandikian, Maria Hristopoulos, James Ziai, Elaine Mai, Klara Totpal, Wei-Ching Liang, Yan Wu, Dionysos Slaga, Kathy Hötzel, Hao Cai, Ji Li, Meric A. Ovacik, Dorothee Nickles, Jennifer Johnston, Diego Ellerman, Daniel Bravo-Perez, Robyn Clark, and Debra Dunlap
- Subjects
0301 basic medicine ,Genetically modified mouse ,Cancer Research ,CD3 Complex ,CD3 ,T cell ,Mice, Transgenic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cell Line, Tumor ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,Amino Acid Sequence ,Ovarian Neoplasms ,biology ,business.industry ,Cancer ,medicine.disease ,Tumor antigen ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research ,Female ,Antibody ,Ovarian cancer ,business - Abstract
Ovarian cancer is a diverse class of tumors with very few effective treatment options and suboptimal response rates in early clinical studies using immunotherapies. Here we describe LY6/PLAUR domain containing 1 (LYPD1) as a novel target for therapeutic antibodies for the treatment of ovarian cancer. LYPD1 is broadly expressed in both primary and metastatic ovarian cancer with ∼70% prevalence in the serous cancer subset. Bispecific antibodies targeting CD3 on T cells and a tumor antigen on cancer cells have demonstrated significant clinical activity in hematologic cancers. We have developed an anti-LYPD1/CD3 T-cell-dependent bispecific antibody (TDB) to redirect T-cell responses to LYPD1 expressing ovarian cancer. Here we characterize the nonclinical pharmacology of anti-LYPD1/CD3 TDB and show induction of a robust polyclonal T-cell activation and target dependent killing of LYPD1 expressing ovarian cancer cells resulting in efficient in vivo antitumor responses in PBMC reconstituted immune-deficient mice and human CD3 transgenic mouse models. Anti-LYPD1/CD3 TDB is generally well tolerated at high-dose levels in mice, a pharmacologically relevant species, and showed no evidence of toxicity or damage to LYPD1 expressing tissues.
- Published
- 2020
23. IRE1α disruption in triple-negative breast cancer cooperates with anti-angiogenic therapy by reversing ER stress adaptation and remodeling the tumor microenvironment
- Author
-
Thomas D. Wu, Margaret Solon, Zora Modrusan, Monika Dohse, David A. Lawrence, Jonathan M. Harnoss, Peter Walter, David Kan, Ofer Guttman, Mike Reichelt, Ehud Segal, Adrien Le Thomas, Anna Shemorry, Avi Ashkenazi, Joachim Rudolph, Mark Merchant, Lisa Crocker, Hartmut Koeppen, Scot A. Marsters, Kathryn Mesh, and Klara Totpal
- Subjects
0301 basic medicine ,Vascular Endothelial Growth Factor A ,X-Box Binding Protein 1 ,Cancer Research ,Messenger ,Angiogenesis Inhibitors ,Triple Negative Breast Neoplasms ,Mice, SCID ,Gene Knockout Techniques ,Mice ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Tumor Microenvironment ,Triple-negative breast cancer ,Tumor ,Neovascularization, Pathologic ,Protein-Serine-Threonine Kinases ,Endoplasmic Reticulum Stress ,medicine.anatomical_structure ,Immunological ,Oncology ,030220 oncology & carcinogenesis ,Female ,Pericyte ,Oncology and Carcinogenesis ,Antineoplastic Agents ,Protein Serine-Threonine Kinases ,SCID ,Article ,Cell Line ,03 medical and health sciences ,Cell Line, Tumor ,Endoribonucleases ,medicine ,Animals ,Humans ,Oncology & Carcinogenesis ,RNA, Messenger ,Transcription factor ,Neovascularization ,Pathologic ,Tumor microenvironment ,business.industry ,Endoplasmic reticulum ,Gene signature ,Xenograft Model Antitumor Assays ,030104 developmental biology ,Cancer cell ,Unfolded protein response ,Cancer research ,RNA ,business - Abstract
Cancer cells exploit the unfolded protein response (UPR) to mitigate endoplasmic reticulum (ER) stress caused by cellular oncogene activation and a hostile tumor microenvironment (TME). The key UPR sensor IRE1α resides in the ER and deploys a cytoplasmic kinase–endoribonuclease module to activate the transcription factor XBP1s, which facilitates ER-mediated protein folding. Studies of triple-negative breast cancer (TNBC)—a highly aggressive malignancy with a dismal posttreatment prognosis—implicate XBP1s in promoting tumor vascularization and progression. However, it remains unknown whether IRE1α adapts the ER in TNBC cells and modulates their TME, and whether IRE1α inhibition can enhance antiangiogenic therapy—previously found to be ineffective in patients with TNBC. To gauge IRE1α function, we defined an XBP1s-dependent gene signature, which revealed significant IRE1α pathway activation in multiple solid cancers, including TNBC. IRE1α knockout in TNBC cells markedly reversed substantial ultrastructural expansion of their ER upon growth in vivo. IRE1α disruption also led to significant remodeling of the cellular TME, increasing pericyte numbers while decreasing cancer-associated fibroblasts and myeloid-derived suppressor cells. Pharmacologic IRE1α kinase inhibition strongly attenuated growth of cell line–based and patient-derived TNBC xenografts in mice and synergized with anti-VEGFA treatment to cause tumor stasis or regression. Thus, TNBC cells critically rely on IRE1α to adapt their ER to in vivo stress and to adjust the TME to facilitate malignant growth. TNBC reliance on IRE1α is an important vulnerability that can be uniquely exploited in combination with antiangiogenic therapy as a promising new biologic approach to combat this lethal disease. Significance: Pharmacologic IRE1α kinase inhibition reverses ultrastructural distension of the ER, normalizes the tumor vasculature, and remodels the cellular TME, attenuating TNBC growth in mice.
- Published
- 2020
24. The tumor suppressor <scp>BAP</scp> 1 cooperates with <scp>BRAFV</scp> 600E to promote tumor formation in cutaneous melanoma
- Author
-
Sara Chan, Eric Stawiski, Christopher Tran, Mira S. Chaurushiya, Wyne P. Lee, Anwesha Dey, Boris C. Bastian, Xiumin Wu, Kathy Hötzel, Trang H. Pham, Klara Totpal, Sreedevi Chalasani, Ho-June Lee, Mengshu Xu, Jeffrey Hung, Zora Modrusan, Nicolas W. Hughes, Vishva M. Dixit, Eric Talevich, Neeraj Sharma, Matthew T. Chang, Alan Hunter Shain, Philamer C. Calses, Jennie R. Lill, Joshua D. Webster, and Zhongwu Li
- Subjects
Proto-Oncogene Proteins B-raf ,0301 basic medicine ,Epithelial-Mesenchymal Transition ,Lung Neoplasms ,Skin Neoplasms ,Transcription, Genetic ,Carcinogenesis ,DNA damage ,Dermatology ,General Biochemistry, Genetics and Molecular Biology ,law.invention ,Histones ,03 medical and health sciences ,0302 clinical medicine ,law ,Cell Line, Tumor ,Histone H2A ,Animals ,Medicine ,Mesothelioma ,Melanoma ,neoplasms ,Cell Proliferation ,Mice, Knockout ,BAP1 ,business.industry ,Tumor Suppressor Proteins ,Ubiquitination ,medicine.disease ,Gene Expression Regulation, Neoplastic ,Mice, Inbred C57BL ,030104 developmental biology ,Oncology ,Tumor progression ,030220 oncology & carcinogenesis ,Mutation ,Cutaneous melanoma ,Cancer research ,Melanocytes ,Suppressor ,business ,Ubiquitin Thiolesterase ,Gene Deletion ,DNA Damage - Abstract
The deubiquitinating enzyme BAP1 is mutated in a hereditary cancer syndrome with a high risk of mesothelioma and melanocytic tumors. Here, we show that Bap1 deletion in melanocytes cooperates with the constitutively active, oncogenic form of BRAF (BRAFV600E ) and UV to cause melanoma in mice, albeit at very low frequency. In addition, Bap1-null melanoma cells derived from mouse tumors are more aggressive and colonize and grow at distant sites more than their wild-type counterparts. Molecularly, Bap1-null melanoma cell lines have increased DNA damage measured by γH2aX and hyperubiquitination of histone H2a. Therapeutically, these Bap1-null tumors are completely responsive to BRAF- and MEK-targeted therapies. Therefore, BAP1 functions as a tumor suppressor and limits tumor progression in melanoma.
- Published
- 2018
25. An anti-CD3/anti–CLL-1 bispecific antibody for the treatment of acute myeloid leukemia
- Author
-
Andrew Polson, Steven T Laing, Elizabeth Lu, Klara Totpal, Lucinda Tam, Genee Lee, Crystal Zhang, Allen Nguyen, Mark S. Dennis, Kedan Lin, Yu-Waye Chu, Mary Mathieu, Robert J. Newman, Alfred Wong, Bing Zheng, Rodney A. Prell, Maria Hristopoulos, Brian R. Vuillemenot, Diego Ellerman, Siddharth Sukumaran, Steven R. Leong, Chen Gu, and Shannon Stainton
- Subjects
Myeloid ,Sialic Acid Binding Ig-like Lectin 3 ,Immunology ,CD33 ,Antineoplastic Agents ,Mice, Transgenic ,Biology ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,immune system diseases ,Cell Line, Tumor ,hemic and lymphatic diseases ,Acute lymphocytic leukemia ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,Cytotoxic T cell ,Lectins, C-Type ,neoplasms ,Myeloid Neoplasia ,Myeloid leukemia ,Cell Biology ,Hematology ,medicine.disease ,Mice, Inbred C57BL ,Leukemia, Myeloid, Acute ,Macaca fascicularis ,Haematopoiesis ,Leukemia ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Blinatumomab ,030215 immunology ,medicine.drug - Abstract
Acute myeloid leukemia (AML) is a major unmet medical need. Most patients have poor long-term survival, and treatment has not significantly changed in 40 years. Recently, bispecific antibodies that redirect the cytotoxic activity of effector T cells by binding to CD3, the signaling component of the T-cell receptor, and a tumor target have shown clinical activity. Notably, blinatumomab is approved to treat relapsed/refractory acute lymphoid leukemia. Here we describe the design, discovery, pharmacologic activity, pharmacokinetics, and safety of a CD3 T cell-dependent bispecific (TDB) full-length human IgG1 therapeutic antibody targeting CLL-1 that could potentially be used in humans to treat AML. CLL-1 is prevalent in AML and, unlike other targets such as CD33 and CD123, is not expressed on hematopoietic stem cells providing potential hematopoietic recovery. We selected a high-affinity monkey cross-reactive anti-CLL-1 arm and tested several anti-CD3 arms that varied in affinity, and determined that the high-affinity CD3 arms were up to 100-fold more potent in vitro. However, in mouse models, the efficacy differences were less pronounced, probably because of prolonged exposure to TDB found with lower-affinity CD3 TDBs. In monkeys, assessment of safety and target cell depletion by the high- and low-affinity TDBs revealed that only the low-affinity CD3/CLL1 TDB was well tolerated and able to deplete target cells. Our data suggest that an appropriately engineered CLL-1 TDB could be effective in the treatment of AML.
- Published
- 2017
26. Disruption of IRE1α through its kinase domain attenuates multiple myeloma
- Author
-
Heidi J.A. Wallweber, Diego Acosta-Alvear, Yung-Chia Ariel Chen, Maria N. Lorenzo, Jiansheng Wu, Mark Merchant, Anna Shemorry, Michael J VanWyngarden, Klara Totpal, Jonathan M. Harnoss, Alvin Gogineni, Adrien Le Thomas, Daniel W. Sherbenou, Susan Kaufman, Robby M. Weimer, Marie-Gabrielle Braun, David A. Lawrence, Weiru Wang, Kevin R Clark, Mike Reichelt, Tom De Bruyn, Steven T Laing, Min Lu, Avi Ashkenazi, David Kan, Benjamin Haley, Jing Qing, Maureen Beresini, Justin Ly, Amy Heidersbach, Ehud Segal, Wendy Sandoval, Martine Amiot, Peter Walter, Scot A. Marsters, Patricia Gomez-Bougie, Dong Lee, Joachim Rudolph, Department of Cancer Immunology, Genentech, Inc. [San Francisco], Translational Oncology [South San Francisco, CA, USA], Pathology [South San Francisco, CA, USA], Structural Biology [South San Francisco, CA, USA], Biochemical and Cellular Pharmacology [South San Francisco, CA, USA], Microchemistry, Proteomics and Lipidomics [South San Francisco, CA, USA], Protein Chemistry [South San Francisco, CA, USA], Drug Metabolism and Pharmacokinetics [South San Francisco, CA, USA], Molecular Biology [South San Francisco, CA, USA], Biomolecular Imaging [South San Francisco, CA, USA], Safety Assessment [South San Francisco, CA, USA], Department of Discovery Chemistry, Division of Hematology [Aurora, CO, USA] (Department of Medicine), University of Colorado Cancer Center [Aurora, CO, USA], Regulation of Bcl2 and p53 Networks in Multiple Myeloma and Mantle Cell Lymphoma (CRCINA-ÉQUIPE 10), Centre de Recherche en Cancérologie et Immunologie Nantes-Angers (CRCINA), Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Université de Nantes - UFR de Médecine et des Techniques Médicales (UFR MEDECINE), Université de Nantes (UN)-Université de Nantes (UN)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Centre National de la Recherche Scientifique (CNRS)-Université d'Angers (UA), Service d'Hématologie Clinique [CHU Nantes] (Unité d'Investigation Clinique), Centre hospitalier universitaire de Nantes (CHU Nantes), Department of Biochemistry and Biophysics [San Francisco], University of California, Howard Hughes Medical Institute [San Francisco, CA, USA], University of California [San Francisco] (UCSF), University of California-University of California, Bernardo, Elizabeth, Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes)-Université d'Angers (UA)-Université de Nantes (UN)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre hospitalier universitaire de Nantes (CHU Nantes), University of California (UC), University of California [San Francisco] (UC San Francisco), and University of California (UC)-University of California (UC)
- Subjects
Male ,X-Box Binding Protein 1 ,Chemokine ,Bortezomib ,Mice ,0302 clinical medicine ,kinase inhibitors ,Lenalidomide ,Multiple myeloma ,media_common ,0303 health sciences ,Multidisciplinary ,biology ,unfolded protein response ,Biological Sciences ,Middle Aged ,Protein-Serine-Threonine Kinases ,Endoplasmic Reticulum Stress ,3. Good health ,Gene Expression Regulation, Neoplastic ,multiple myeloma ,PNAS Plus ,030220 oncology & carcinogenesis ,Female ,Antibody ,medicine.drug ,Signal Transduction ,inositol-requiring enzyme 1 ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,Protein Serine-Threonine Kinases ,03 medical and health sciences ,[SDV.CAN] Life Sciences [q-bio]/Cancer ,Endoribonucleases ,medicine ,media_common.cataloged_instance ,Animals ,Humans ,Secretion ,European union ,Protein Kinase Inhibitors ,030304 developmental biology ,Aged ,Neoplastic ,business.industry ,Endoplasmic reticulum ,Cell Biology ,medicine.disease ,Xenograft Model Antitumor Assays ,Gene Expression Regulation ,Cancer research ,biology.protein ,business ,Homeostasis - Abstract
Significance Multiple myeloma (MM) is a lethal malignancy arising from plasma cells. MM cells experience endoplasmic reticulum (ER) stress due to immunoglobulin hyperproduction. The ER-resident sensor IRE1α mitigates ER stress by expanding protein-folding and secretion capacity, while supporting proteasomal degradation of ER misfolded proteins. IRE1α elaborates these functions by deploying a cytoplasmic kinase–RNase module to activate the transcription factor XBP1s. Although IRE1α has been implicated in MM, its validity as a potential therapeutic target—particularly as a kinase—has been unclear. Using genetic and pharmacologic disruption, we demonstrate that the IRE1α–XBP1s pathway is critical for MM tumor growth. We further show that the kinase domain of IRE1α is an effective and safe potential small-molecule target for MM therapy., Multiple myeloma (MM) arises from malignant immunoglobulin (Ig)-secreting plasma cells and remains an incurable, often lethal disease despite therapeutic advances. The unfolded-protein response sensor IRE1α supports protein secretion by deploying a kinase–endoribonuclease module to activate the transcription factor XBP1s. MM cells may co-opt the IRE1α–XBP1s pathway; however, the validity of IRE1α as a potential MM therapeutic target is controversial. Genetic disruption of IRE1α or XBP1s, or pharmacologic IRE1α kinase inhibition, attenuated subcutaneous or orthometastatic growth of MM tumors in mice and augmented efficacy of two established frontline antimyeloma agents, bortezomib and lenalidomide. Mechanistically, IRE1α perturbation inhibited expression of key components of the endoplasmic reticulum-associated degradation machinery, as well as secretion of Ig light chains and of cytokines and chemokines known to promote MM growth. Selective IRE1α kinase inhibition reduced viability of CD138+ plasma cells while sparing CD138− cells derived from bone marrows of newly diagnosed or posttreatment-relapsed MM patients, in both US- and European Union-based cohorts. Effective IRE1α inhibition preserved glucose-induced insulin secretion by pancreatic microislets and viability of primary hepatocytes in vitro, as well as normal tissue homeostasis in mice. These results establish a strong rationale for developing kinase-directed inhibitors of IRE1α for MM therapy.
- Published
- 2019
27. CD3 bispecific antibody-induced cytokine release is dispensable for cytotoxic T cell activity
- Author
-
Teemu T. Junttila, Robyn Clark, Maria Hristopoulos, Ji Li, Robert Piskol, Ryan Ybarra, Klara Totpal, Ying-Jiun J. Chen, Jason Li, Melissa R. Junttila, Zora Modrusan, and Dionysos Slaga
- Subjects
Cytotoxicity, Immunologic ,CD3 Complex ,Receptor, ErbB-2 ,medicine.medical_treatment ,T cell ,CD3 ,Mice, Transgenic ,Monocytes ,Antibodies, Bispecific ,medicine ,Cytotoxic T cell ,Animals ,Humans ,biology ,Chemistry ,Tumor Necrosis Factor-alpha ,Monocyte ,Macrophages ,General Medicine ,medicine.disease ,Cytokine release syndrome ,Cytolysis ,medicine.anatomical_structure ,Cytokine ,Cancer research ,biology.protein ,Cytokines ,Tumor necrosis factor alpha ,T-Lymphocytes, Cytotoxic - Abstract
T cell–retargeting therapies have transformed the therapeutic landscape of oncology. Regardless of the modality, T cell activating therapies are commonly accompanied by systemic cytokine release, which can progress to deadly cytokine release syndrome (CRS). Because of incomplete mechanistic understanding of the relationship between T cell activation and systemic cytokine release, optimal toxicity management that retains full therapeutic potential remains unclear. Here, we report the cell type–specific cellular mechanisms that link CD3 bispecific antibody–mediated killing to toxic cytokine release. The immunologic cascade is initiated by T cell triggering, whereas monocytes and macrophages are the primary source of systemic toxic cytokine release. We demonstrate that T cell–generated tumor necrosis factor–α (TNF-α) is the primary mechanism mediating monocyte activation and systemic cytokine release after CD3 bispecific treatment. Prevention of TNF-α release is sufficient to impair systemic release of monocyte cytokines without affecting antitumor efficacy. Systemic cytokine release is only observed upon initial exposure to CD3 bispecific antibody not subsequent doses, indicating a biological distinction between doses. Despite impaired cytokine release after second exposure, T cell cytotoxicity remained unaffected, demonstrating that cytolytic activity of T cells can be achieved in the absence of cytokine release. The mechanistic uncoupling of toxic cytokines and T cell cytolytic activity in the context of CD3 bispecifics provides a biological rationale to clinically explore preventative treatment approaches to mitigate toxicity.
- Published
- 2019
28. Disruption of IRElα through its Kinase Domain Attenuates Multiple Myeloma
- Author
-
David A. Lawrence, Kevin R Clark, Tom De Bruyn, Justin Ly, Dong Lee, Yung-Chia Ariel Chen, Amy Heidersbachh, Joachim Rudolph, Wendy Sandoval, Alvin Gogineni, Klara Totpal, Michael J VanWyngarden, Martine Amiot, David Kan, Benjamin Haley, Peter Walter, Adrien Le Thomas, Robby M. Weimer, Scot A. Marsters, Patricia Gomez-Bougie, Weiru Wang, Diego Acosta-Alvear, Jiansheng Wu, Maria N. Lorenzo, Min Lu, Heidi J.A. Wallweber, Avi Ashkenazi, Ehud Segal, Jing Qing, Daniel W. Sherbenou, Marie-Gabrielle Braun, Susan Kaufman, Jonathan M. Harnoss, and Maureen Beresini
- Subjects
Chemokine ,biology ,business.industry ,Kinase ,Bortezomib ,medicine.disease ,medicine.anatomical_structure ,Protein kinase domain ,medicine ,biology.protein ,Cancer research ,Bone marrow ,business ,Transcription factor ,Multiple myeloma ,Lenalidomide ,medicine.drug - Abstract
Multiple myeloma (MM) arises from malignant immunoglobulin-secreting plasma cells and remains an incurable, often lethal disease despite recent therapeutic advances. The unfolded-protein response sensor IRE1α supports protein secretion by deploying a kinase-endoribonuclease module to activate the transcription factor XBP1s. MM cells may coopt the IRE1α-XBP1s pathway; however, the validity of IRE1α as a potential MM therapeutic target is controversial. Here we show that genetic disruption of IRE1α or XBP1s, or pharmacologic IRE1α kinase inhibition, attenuated subcutaneous or orthometastatic growth of MM tumors in mice, and augmented efficacy of two well-established frontline antimyeloma agents, bortezomib or lenalidomide. Mechanistically, IRE1α perturbation inhibited expression of key components of the ER-associated degradation machinery, as well as cytokines and chemokines known to promote MM growth. Selective IRE1α kinase inhibition reduced viability of CD138+ plasma cells while sparing CD138− cells from bone marrow of newly diagnosed MM patients or patients whose disease relapsed after 1 - 4 lines of treatment in both US- and EU-based cohorts. IRE1α inhibition preserved survival and glucose-induced insulin secretion by pancreatic microislets. Together, these results establish a strong therapeutic rationale for targeting IRE1α with kinase-based small-molecule inhibitors in MM.Significance statementMultiple myeloma (MM) is a lethal malignancy of plasma cells. MM cells have an expanded endoplasmic reticulum (ER) that is constantly under stress due to immunoglobulin hyperproduction. The ER-resident sensor IRE1α mitigates ER stress by expanding the ER’s protein-folding capacity while supporting proteasomal degradation of misfolded ER proteins. IRE1α elaborates these functions by deploying its cytoplasmic kinase-RNase module to activate the transcription factor XBP1s. The validity of IRE1α as a potential therapeutic target in MM has been questioned. Using genetic and pharmacologic disruption in vitro and in vivo, we demonstrate that the IRE1α-XBP1s pathway plays a critical role in MM growth. We further show that IRE1α’s kinase domain is an effective and safe potential small-molecule target for MM therapy.
- Published
- 2018
- Full Text
- View/download PDF
29. Immunization associated with primary tumor growth leads to rejection of commonly used syngeneic tumors upon tumor rechallenge
- Author
-
Stephen E. Gould, Amy M Berkley, Bruno Alicke, Sophie M. Lehar, Jill Schartner, Klara Totpal, Rafael Cubas, and Aude-Hélène Capietto
- Subjects
Male ,Cancer Research ,medicine.medical_treatment ,T cell ,Immunology ,Short Report ,Mice ,Immune system ,Immunity ,Neoplasms ,medicine ,Animals ,Humans ,Immunology and Allergy ,drug evaluation, preclinical ,RC254-282 ,Pharmacology ,immunologic memory ,business.industry ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Immunosuppression ,Immunotherapy ,medicine.disease ,Primary tumor ,medicine.anatomical_structure ,Oncology ,Immunization ,Cancer cell ,Cancer research ,Molecular Medicine ,Female ,immunotherapy ,business - Abstract
The recent success of multiple immunomodulating drugs in oncology highlights the potential of relieving immunosuppression by directly engaging the immune system in the tumor bed to target cancer cells. Durable responses to immune checkpoint inhibitors experienced by some patients may be indicative of the formation of a T cell memory response. This has prompted the search for preclinical evidence of therapy-induced long-term immunity as part of the evaluation of novel therapeutics. A common preclinical method used to document long-term immunity is the use of tumor rechallenge experiments in which tumor growth is assessed in mice that have previously rejected tumors in response to therapy. Failure of rechallenge engraftment, typically alongside successful engraftment of the same tumor in naive animals as a control, is often presented as evidence of therapy-induced tumor immunity. Here, we present evidence that formation of tumor immunity often develops independent of therapy. We observed elevated rates of rechallenge rejection following surgical resection of primary tumors for four of five commonly used models and that such postexcision immunity could be adoptively transferred to treatment-naïve mice. We also show that tumor-specific cytolytic T cells are induced on primary tumor challenge independent of therapeutic intervention. Taken together these data call into question the utility of tumor rechallenge studies and the use of naïve animals as controls to demonstrate therapy-induced formation of long-term tumor immunity.
- Published
- 2020
30. The Hippo pathway effector TAZ induces TEAD-dependent liver inflammation and tumors
- Author
-
Thijs J. Hagenbeek, Xiumin Wu, Matthew T. Chang, Wyne P. Lee, Christiaan Klijn, Shuqun Yang, Joshua D. Webster, Noelyn M. Kljavin, Kevin Walsh, Qunsheng Ji, Zhixiang Zhang, Anwesha Dey, Dae-Sik Lim, Buvana Ravishankar, Da-Hye Lee, Georgia Hatzivassiliou, Cecile de la Cruz, Stephen E. Gould, Naiying Yang, Qingyang Gu, Ho-June Lee, Trang H. Pham, Erica L. Jackson, Klara Totpal, and Allen G. Cai
- Subjects
0301 basic medicine ,Myeloid ,Cell ,Transplantation, Heterologous ,Inflammation ,Cell Cycle Proteins ,Biology ,Protein Serine-Threonine Kinases ,Biochemistry ,Proinflammatory cytokine ,03 medical and health sciences ,medicine ,Animals ,Humans ,Secretion ,Hippo Signaling Pathway ,Molecular Biology ,Hippo signaling pathway ,Effector ,Gene Expression Profiling ,Liver Neoplasms ,Intracellular Signaling Peptides and Proteins ,Nuclear Proteins ,TEA Domain Transcription Factors ,Cell Biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Liver ,Transcriptional Coactivator with PDZ-Binding Motif Proteins ,Mutation ,Cancer research ,Trans-Activators ,Cytokines ,medicine.symptom ,Homeostasis ,Signal Transduction ,Transcription Factors - Abstract
The Hippo signaling pathway regulates organ size and plays critical roles in maintaining tissue growth, homeostasis, and regeneration. Dysregulated in a wide spectrum of cancers, in mammals, this pathway is regulated by two key effectors, YAP and TAZ, that may functionally overlap. We found that TAZ promoted liver inflammation and tumor development. The expression of TAZ, but not YAP, in human liver tumors positively correlated with the expression of proinflammatory cytokines. Hyperactivated TAZ induced substantial myeloid cell infiltration into the liver and the secretion of proinflammatory cytokines through a TEAD-dependent mechanism. Furthermore, tumors with hyperactivated YAP and TAZ had distinct transcriptional signatures, which included the increased expression of inflammatory cytokines in TAZ-driven tumors. Our study elucidated a previously uncharacterized link between TAZ activity and inflammatory responses that influence tumor development in the liver.
- Published
- 2018
31. IFNγ-induced Chemokines Are Required for CXCR3-mediated T-Cell Recruitment and Antitumor Efficacy of Anti-HER2/CD3 Bispecific Antibody
- Author
-
Kevin Walsh, Melissa R. Junttila, Aurelie Herault, James Ziai, Judy Mak, Ji Li, Ryan Ybarra, Klara Totpal, Patrícia de Almeida, Teemu T. Junttila, and Alfonso Arrazate
- Subjects
0301 basic medicine ,Cancer Research ,Chemokine ,Adoptive cell transfer ,Receptors, CXCR3 ,CD3 Complex ,Receptor, ErbB-2 ,CD3 ,T cell ,T-Lymphocytes ,CD8-Positive T-Lymphocytes ,CXCR3 ,Lymphocyte Activation ,03 medical and health sciences ,Interferon-gamma ,Mice ,0302 clinical medicine ,Lymphocytes, Tumor-Infiltrating ,Cell Line, Tumor ,Neoplasms ,Antibodies, Bispecific ,Medicine ,Animals ,Humans ,Mammary tumor ,biology ,business.industry ,Adoptive Transfer ,Xenograft Model Antitumor Assays ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,biology.protein ,Cancer research ,Cytokines ,Female ,Antibody ,Chemokines ,Inflammation Mediators ,business ,CD8 ,Signal Transduction - Abstract
Purpose: The response to cancer immune therapy is dependent on endogenous tumor-reactive T cells. To bypass this requirement, CD3-bispecific antibodies have been developed to induce a polyclonal T-cell response against the tumor. Anti-HER2/CD3 T-cell–dependent bispecific (TDB) antibody is highly efficacious in the treatment of HER2-overexpressing tumors in mice. Efficacy and immunologic effects of anti-HER2/CD3 TDB were investigated in mammary tumor model with very few T cells prior treatment. We further describe the mechanism for TDB-induced T-cell recruitment to tumors. Experimental Design: The immunologic effects and the mechanism of CD3-bispecific antibody-induced T-cell recruitment into spontaneous HER2-overexpressing mammary tumors was studied using human HER2 transgenic, immunocompetent mouse models. Results: Anti-HER2/CD3 TDB treatment induced an inflammatory response in tumors converting them from poorly infiltrated to an inflamed, T-cell abundant, phenotype. Multiple mechanisms accounted for the TDB-induced increase in T cells within tumors. TDB treatment induced CD8+ T-cell proliferation. T cells were also actively recruited post-TDB treatment by IFNγ-dependent T-cell chemokines mediated via CXCR3. This active T-cell recruitment by TDB-induced chemokine signaling was the dominant mechanism and necessary for the therapeutic activity of anti-HER2/CD3 TDB. Conclusions: In summary, we demonstrate that the activity of anti-HER2/CD3 TDB was not dependent on high-level baseline T-cell infiltration. Our results suggest that anti-HER2/CD3 TDB may be efficacious in patients and indications that respond poorly to checkpoint inhibitors. An active T-cell recruitment mediated by TDB-induced chemokine signaling was the major mechanism for T-cell recruitment.
- Published
- 2018
32. Avidity-based binding to HER2 results in selective killing of HER2-overexpressing cells by anti-HER2/CD3
- Author
-
Amy L. Shelton, Ji Li, Rodney A. Prell, Jennifer Johnston, Christoph Spiess, Genee Lee, Kapil Gadkar, James T. Koerber, Amy C. Y. Lo, Klara Totpal, Diego Ellerman, Teemu T. Junttila, T. Noelle Lombana, Maria Hristopoulos, Elaine Mai, Robyn Clark, Dionysos Slaga, and Rajesh Vij
- Subjects
0301 basic medicine ,Cytotoxicity, Immunologic ,CD3 Complex ,Receptor, ErbB-2 ,T cell ,CD3 ,Antibody Affinity ,medicine.disease_cause ,Lymphocyte Activation ,Autoimmunity ,03 medical and health sciences ,Immunoglobulin Fab Fragments ,Therapeutic index ,Cell Line, Tumor ,Antibodies, Bispecific ,medicine ,Humans ,Avidity ,skin and connective tissue diseases ,Cytotoxicity ,biology ,Chemistry ,General Medicine ,030104 developmental biology ,medicine.anatomical_structure ,Cell culture ,Immunoglobulin G ,biology.protein ,Cancer research ,Antibody ,Protein Binding - Abstract
A primary barrier to the success of T cell-recruiting bispecific antibodies in the treatment of solid tumors is the lack of tumor-specific targets, resulting in on-target off-tumor adverse effects from T cell autoreactivity to target-expressing organs. To overcome this, we developed an anti-HER2/CD3 T cell-dependent bispecific (TDB) antibody that selectively targets HER2-overexpressing tumor cells with high potency, while sparing cells that express low amounts of HER2 found in normal human tissues. Selectivity is based on the avidity of two low-affinity anti-HER2 Fab arms to high target density on HER2-overexpressing cells. The increased selectivity to HER2-overexpressing cells is expected to mitigate the risk of adverse effects and increase the therapeutic index. Results included in this manuscript not only support the clinical development of anti-HER2/CD3 1Fab-immunoglobulin G TDB but also introduce a potentially widely applicable strategy for other T cell-directed therapies. The potential of this discovery has broad applications to further enable consideration of solid tumor targets that were previously limited by on-target, but off-tumor, autoimmunity.
- Published
- 2018
33. Relative Target Affinities of T-Cell-Dependent Bispecific Antibodies Determine Biodistribution in a Solid Tumor Mouse Model
- Author
-
Danielle Mandikian, C. Andrew Boswell, Jason Ho, Teemu T. Junttila, Kedan Lin, Maria Hristopoulos, Amy C. Y. Lo, Mark S. Dennis, Saileta Prabhu, Robyn Clark, Jeffrey Eastham-Anderson, Ji Li, Klara Totpal, Dionysos Slaga, Nene Takahashi, and Sean B. Joseph
- Subjects
0301 basic medicine ,Cancer Research ,Biodistribution ,CD3 Complex ,Receptor, ErbB-2 ,medicine.medical_treatment ,T cell ,CD3 ,Antibody Affinity ,Mice, Nude ,Mice, Transgenic ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Cancer immunotherapy ,Antibodies, Bispecific ,medicine ,Tumor Cells, Cultured ,Cytotoxic T cell ,Animals ,Humans ,Tissue Distribution ,biology ,Chemistry ,Immunotherapy ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,030220 oncology & carcinogenesis ,Cancer cell ,Colonic Neoplasms ,Cancer research ,biology.protein ,Female ,Antibody ,T-Lymphocytes, Cytotoxic - Abstract
Anti-HER2/CD3, a T-cell–dependent bispecific antibody (TDB) construct, induces T-cell–mediated cell death in cancer cells expressing HER2 by cross-linking tumor HER2 with CD3 on cytotoxic T cells, thereby creating a functional cytolytic synapse. TDB design is a very challenging process that requires consideration of multiple parameters. Although therapeutic antibody design strategy is commonly driven by striving for the highest attainable antigen-binding affinity, little is known about how the affinity of each TDB arm can affect the targeting ability of the other arm and the consequent distribution and efficacy. To our knowledge, no distribution studies have been published using preclinical models wherein the T-cell–targeting arm of the TDB is actively bound to T cells. We used a combined approach involving radiochemistry, invasive biodistribution, and noninvasive single-photon emission tomographic (SPECT) imaging to measure TDB distribution and catabolism in transgenic mice with human CD3ϵ expression on T cells. Using CD3 affinity variants, we assessed the impact of CD3 affinity on short-term pharmacokinetics, tissue distribution, and cellular uptake. Our experimental approach determined the relative effects of (i) CD3 targeting to normal tissues, (ii) HER2 targeting to HER2-expressing tumors, and (iii) relative HER2/CD3 affinity, all as critical drivers for TDB distribution. We observed a strong correlation between CD3 affinity and distribution to T-cell–rich tissues, with higher CD3 affinity reducing systemic exposure and shifting TDB distribution away from tumor to T-cell–containing tissues. These observations have important implications for clinical translation of bispecific antibodies for cancer immunotherapy. Mol Cancer Ther; 17(4); 776–85. ©2018 AACR.
- Published
- 2017
34. Tumour and host cell PD-L1 is required to mediate suppression of anti-tumour immunity in mice
- Author
-
Maike Schmidt, Jeong M. Kim, Bryan Irving, Steve Lianoglou, Patrick Caplazi, Henry Chiu, Hartmut Koeppen, Rebecca Hong, Klara Totpal, Jacqueline McBride, Armando Navarro, Vincent Javinal, Robert L. Yauch, Marcia Belvin, Ira Mellman, Janet Lau, Jane L. Grogan, Laura Sanders, Jeanne Cheung, and Benjamin Haley
- Subjects
0301 basic medicine ,Cell type ,Myeloid ,Science ,Programmed Cell Death 1 Receptor ,General Physics and Astronomy ,General Biochemistry, Genetics and Molecular Biology ,Article ,B7-H1 Antigen ,03 medical and health sciences ,Immune system ,PD-L1 ,Cell Line, Tumor ,Neoplasms ,Blocking antibody ,medicine ,Tumor Microenvironment ,Cytotoxic T cell ,Animals ,Humans ,Receptor ,Antibodies, Blocking ,Mice, Knockout ,Multidisciplinary ,biology ,General Chemistry ,030104 developmental biology ,medicine.anatomical_structure ,Immunology ,biology.protein ,Antibody ,Signal Transduction ,T-Lymphocytes, Cytotoxic - Abstract
Expression of PD-L1, the ligand for T-cell inhibitory receptor PD-1, is one key immunosuppressive mechanism by which cancer avoids eradication by the immune system. Therapeutic use of blocking antibodies to PD-L1 or its receptor PD-1 has produced unparalleled, durable clinical responses, with highest likelihood of response seen in patients whose tumour or immune cells express PD-L1 before therapy. The significance of PD-L1 expression in each cell type has emerged as a central and controversial unknown in the clinical development of immunotherapeutics. Using genetic deletion in preclinical mouse models, here we show that PD-L1 from disparate cellular sources, including tumour cells, myeloid or other immune cells can similarly modulate the degree of cytotoxic T-cell function and activity in the tumour microenvironment. PD-L1 expression in both the host and tumour compartment contribute to immune suppression in a non-redundant fashion, suggesting that both sources could be predictive of sensitivity to therapeutic agents targeting the PD-L1/PD-1 axis., PD-L1, the ligand for T-cell inhibitory receptor PD-1, can be expressed by various cell types in the tumour microenvironment. Here, the authors show that, in mouse models, the expression of PD-L1 from both cancerous and normal host immune cells is important for suppressing anti-tumour immune responses.
- Published
- 2017
35. PAK1 mediates pancreatic cancer cell migration and resistance to MET inhibition
- Author
-
Qian Xiao, Adrian M. Jubb, Robert Grützmann, Wei Zhou, Daniela Aust, Mark Merchant, William F. Forrest, Lei Zheng, Hartmut Koeppen, Elizabeth M. Jaffee, Qinghua Song, Ana De Jesus-Acosta, Klara Totpal, Florian Gnad, Christy C. Ong, Lori Friedman, Karen Lyle, Ling Fu, Mally Romero, Richard M. Neve, Marcia Belvin, Yulei Wang, Yibing Yan, Christian Pilarsky, Rajiv Raja, Klaus P. Hoeflich, Peter M. Haverty, and Rupal Desai
- Subjects
Crizotinib ,Kinase ,Growth factor ,medicine.medical_treatment ,Biology ,medicine.disease ,Pathology and Forensic Medicine ,Metastasis ,PAK1 ,Onartuzumab ,medicine ,Cancer research ,Adenocarcinoma ,Hepatocyte growth factor ,medicine.drug - Abstract
Pancreatic adenocarcinoma (PDAC) is a major unmet medical need and a deeper understanding of molecular drivers is needed to advance therapeutic options for patients. We report here that p21-activated kinase 1 (PAK1) is a central node in PDAC cells downstream of multiple growth factor signalling pathways, including hepatocyte growth factor (HGF) and MET receptor tyrosine kinase. PAK1 inhibition blocks signalling to cytoskeletal effectors and tumour cell motility driven by HGF/MET. MET antagonists, such as onartuzumab and crizotinib, are currently in clinical development. Given that even highly effective therapies have resistance mechanisms, we show that combination with PAK1 inhibition overcomes potential resistance mechanisms mediated either by activation of parallel growth factor pathways or by direct amplification of PAK1. Inhibition of PAK1 attenuated in vivo tumour growth and metastasis in a model of pancreatic adenocarcinoma. In human tissues, PAK1 is highly expressed in a proportion of PDACs (33% IHC score 2 or 3; n = 304) and its expression is significantly associated with MET positivity (p < 0.0001) and linked to a widespread metastatic pattern in patients (p = 0.067). Taken together, our results provide evidence for a functional role of MET/PAK1 signalling in pancreatic adenocarcinoma and support further characterization of therapeutic inhibitors in this indication.
- Published
- 2014
36. Engineering Upper Hinge Improves Stability and Effector Function of a Human IgG1
- Author
-
Ashraf Amanullah, Bartek Nogal, Paul Carter, Shan Chung, Priyanka Gupta, Klara Totpal, Amrita V. Kamath, Martin Vanderlaan, Gabriele Schaefer, Reed J. Harris, Gloria Meng, Jean-Michel Vernes, Craig Emery, Guoying Jiang, Joni Tsukuda, Yuwen Lin, Anne Wong, Daniel Boyd, Boxu Yan, Amy Shen, and Timothy Kaschak
- Subjects
Stereochemistry ,Molecular Sequence Data ,Mutant ,Hinge ,CHO Cells ,Protein degradation ,Protein Engineering ,Immunoglobulin light chain ,Biochemistry ,Cricetulus ,Cricetinae ,Animals ,Humans ,Amino Acid Sequence ,Molecular Biology ,Histidine ,Hydroxyl Radical ,Protein Stability ,Chemistry ,Effector ,Wild type ,Cell Biology ,Protein engineering ,Amino Acid Substitution ,Protein Synthesis and Degradation ,Immunoglobulin G ,Mutation ,Proteolysis ,Biophysics ,Feasibility Studies ,Immunoglobulin Light Chains ,Immunoglobulin Heavy Chains - Abstract
Upper hinge is vulnerable to radical attacks that result in breakage of the heavy-light chain linkage and cleavage of the hinge of an IgG1. To further explore mechanisms responsible for the radical induced hinge degradation, nine mutants were designed to determine the roles that the upper hinge Asp and His play in the radical reactions. The observation that none of these substitutions could inhibit the breakage of the heavy-light chain linkage suggests that the breakage may result from electron transfer from Cys(231) directly to the heavy-light chain linkage upon radical attacks, and implies a pathway separate from His(229)-mediated hinge cleavage. On the other hand, the substitution of His(229) with Tyr showed promising advantages over the native antibody and other substitutions in improving the stability and function of the IgG1. This substitution inhibited the hinge cleavage by 98% and suggests that the redox active nature of Tyr did not enable it to replicate the ability of His to facilitate radical induced degradation. We propose that the lower redox potential of Tyr, a residue that may be the ultimate sink for oxidizing equivalents in proteins, is responsible for the inhibition. More importantly, the substitution increased the antibody's binding to FcγRIII receptors by 2-3-fold, and improved ADCC activity by 2-fold, while maintaining a similar pharmacokinetic profile with respect to the wild type. Implications of these observations for antibody engineering and development are discussed.
- Published
- 2012
37. An Fcγ Receptor-Dependent Mechanism Drives Antibody-Mediated Target-Receptor Signaling in Cancer Cells
- Author
-
Scot A. Marsters, Annie Yang, Stefanie Loeser, Gloria Meng, Avi Ashkenazi, Becky Yang, Robert M. Pitti, Cam Adams, Sarah G. Hymowitz, Dylan Daniel, Jean-Michel Vernes, David A. Lawrence, Bob Kelley, Yun Li, Sarajane Ross, Sharon Yee, Rienk Offringa, Klara Totpal, Nicholas S. Wilson, and Yanmei Lu
- Subjects
Cancer Research ,Apoptosis ,Context (language use) ,Biology ,Antibodies, Monoclonal, Humanized ,Polymorphism, Single Nucleotide ,Mice ,Antigen ,Cell Line, Tumor ,Neoplasms ,Leukocytes ,medicine ,Animals ,Humans ,Myeloid Cells ,CD40 Antigens ,Receptor ,Mice, Knockout ,B-Lymphocytes ,Mice, Inbred BALB C ,Receptor Aggregation ,Receptors, IgG ,NF-kappa B ,Antibodies, Monoclonal ,Cell Biology ,HCT116 Cells ,Xenograft Model Antitumor Assays ,Immunoglobulin Fc Fragments ,Cell biology ,Killer Cells, Natural ,Mice, Inbred C57BL ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Oncology ,Mechanism of action ,Immunoglobulin G ,Mutation ,Cancer cell ,biology.protein ,Female ,medicine.symptom ,Antibody ,Function (biology) ,Protein Binding ,Signal Transduction - Abstract
SummaryAntibodies to cell-surface antigens trigger activatory Fcγ receptor (FcγR)-mediated retrograde signals in leukocytes to control immune effector functions. Here, we uncover an FcγR mechanism that drives antibody-dependent forward signaling in target cells. Agonistic antibodies to death receptor 5 (DR5) induce cancer-cell apoptosis and are in clinical trials; however, their mechanism of action in vivo is not fully defined. Interaction of the DR5-agonistic antibody drozitumab with leukocyte FcγRs promoted DR5-mediated tumor-cell apoptosis. Whereas the anti-CD20 antibody rituximab required activatory FcγRs for tumoricidal function, drozitumab was effective in the context of either activatory or inhibitory FcγRs. A CD40-agonistic antibody required similar FcγR interactions to stimulate nuclear factor-κB activity in B cells. Thus, FcγRs can drive antibody-mediated receptor signaling in target cells.
- Published
- 2011
38. SuperiorIn vivoEfficacy of Afucosylated Trastuzumab in the Treatment of HER2-Amplified Breast Cancer
- Author
-
Teemu T. Junttila, Mark X. Sliwkowski, Oliver Pabonan, Klara Totpal, Tomasz Baginski, Julie Theriault, Christine Olsson, Lisa Crocker, Kathryn Parsons, Robert F. Kelley, Gloria Meng, Yanmei Lu, and Yan Xin
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,Receptor, ErbB-2 ,Antineoplastic Agents ,Breast Neoplasms ,Mice, Transgenic ,Mice, SCID ,Antibodies, Monoclonal, Humanized ,Mice ,Breast cancer ,In vivo ,Trastuzumab ,Cell Line, Tumor ,Internal medicine ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,Cytotoxicity ,neoplasms ,Fucose ,Mice, Inbred BALB C ,biology ,business.industry ,Effector ,Receptors, IgG ,Antibody-Dependent Cell Cytotoxicity ,Antibodies, Monoclonal ,Cancer ,medicine.disease ,Monoclonal ,biology.protein ,Female ,Antibody ,business ,medicine.drug - Abstract
The enhancement of immune effector functions has been proposed as a potential strategy for increasing the efficacy of therapeutic antibodies. Here, we show that removing fucose from trastuzumab (Herceptin) increased its binding to FcγRIIIa, enhanced antibody-dependent cell-mediated cytotoxicity, and more than doubled the median progression-free survival when compared with conventional trastuzumab in treating preclinical models of HER2-amplified breast cancer. Our results show that afucosylated trastuzumab has superior efficacy in treating in vivo models of HER2-amplified breast cancer and support the development of effector function–enhanced antibodies for solid tumor therapy. Cancer Res; 70(11); 4481–9. ©2010 AACR.
- Published
- 2010
39. Cooperation of the Agonistic DR5 Antibody Apomab with Chemotherapy to Inhibit Orthotopic Lung Tumor Growth and Improve Survival
- Author
-
Ralph H. Schwall, Sharon Fong, Klara Totpal, Hartmut Koeppen, Richard A.D. Carano, Zhong Zheng, Avi Ashkenazi, Renhui Yang, Hongkui Jin, Howard M. Stern, Jed Ross, and David S. Lawrence
- Subjects
Cancer Research ,Pathology ,medicine.medical_specialty ,Lung Neoplasms ,Paclitaxel ,medicine.medical_treatment ,Immunoblotting ,Mice, Nude ,Apoptosis ,Carboplatin ,Mice ,chemistry.chemical_compound ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,In Situ Nick-End Labeling ,Carcinoma ,Animals ,Humans ,Medicine ,Lung cancer ,Cell Proliferation ,Chemotherapy ,business.industry ,Antibodies, Monoclonal ,Cancer ,medicine.disease ,Xenograft Model Antitumor Assays ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Oncology ,chemistry ,Tumor progression ,Cancer cell ,Cancer research ,Female ,business - Abstract
Purpose: Apomab is a fully human monoclonal antibody that induces programmed cell death through the proapoptotic receptor DR5 in various cancer cells but not in normal cells. Several lung cancer cell lines express DR5 and exhibit apoptosis in response to apomab in vitro. Experimental Design: We investigated the efficacy of apomab and its interaction with chemotherapy in xenograft models based on human NCI-H460 non–small-cell lung carcinoma cells. In an established model of s.c. tumor xenografts, apomab or Taxol plus carboplatin chemotherapy delayed tumor progression, whereas combined treatment caused tumor regression and a substantially longer growth delay. To test apomab activity in a setting that may more closely mimic lung cancer pathology in patients, we developed a lung orthotopic model. Results: In this model, microcomputed tomography imaging showed that apomab, chemotherapy, or combination treatment significantly inhibited tumor growth compared with vehicle, whereas the combination caused greater inhibition in tumor growth relative to chemotherapy or apomab. Similarly, histologic analysis revealed that apomab, chemotherapy, or the combination significantly reduced tumor size compared with vehicle, whereas the combination induced significantly greater reduction in tumor size than did chemotherapy or apomab. Furthermore, combined treatment improved 105-day survival relative to vehicle (P = 0.0023) as well as to apomab (P = 0.0445) or chemotherapy (P = 0.0415). Conclusion: These results show a positive interaction of apomab with chemotherapy, evidenced by significant inhibition of tumor growth as well as improved survival, thus supporting further investigation of this therapeutic approach in lung cancer patients.
- Published
- 2008
40. Erlotinib Directly Inhibits HER2 Kinase Activation and Downstream Signaling Events in Intact Cells Lacking Epidermal Growth Factor Receptor Expression
- Author
-
Robert W. Akita, Gabriele Schaefer, Lihua Shao, and Klara Totpal
- Subjects
Cancer Research ,DNA, Complementary ,Lung Neoplasms ,Receptor, ErbB-3 ,MAP Kinase Signaling System ,Receptor, ErbB-2 ,Neuregulin-1 ,Cetuximab ,Cell Growth Processes ,Antibodies, Monoclonal, Humanized ,Erlotinib Hydrochloride ,Mice ,Growth factor receptor ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Drug Interactions ,Epidermal growth factor receptor ,Phosphorylation ,skin and connective tissue diseases ,Receptor ,Protein Kinase Inhibitors ,neoplasms ,biology ,Cell growth ,Kinase ,Antibodies, Monoclonal ,respiratory tract diseases ,ErbB Receptors ,Oncology ,Quinazolines ,Cancer research ,biology.protein ,Erlotinib ,Signal transduction ,Proto-Oncogene Proteins c-akt ,medicine.drug - Abstract
Erlotinib (Tarceva), is an orally available, reversible inhibitor of epidermal growth factor receptor (EGFR; HER1) that exhibits inhibitory activity on purified HER2 kinase at much higher concentrations. Despite the minimal activity on purified protein in vitro, in vivo studies show that erlotinib inhibits the growth of HER2-driven systems effectively. Several hypotheses have been put forward to explain this discrepancy. In particular, it has been suggested that erlotinib might indirectly suppress the activity of HER2 by blocking the ability of EGFR to transactivate it when the two receptors are part of a heterodimer complex. However, an alternative possibility that has not been adequately addressed is whether the direct inhibitory action of erlotinib on the HER2 kinase might account for the observed biological responses. To distinguish between a direct effect of erlotinib on HER2 kinase in intact cells or an indirect effect of erlotinib on HER2 activity that is mediated through EGFR, we generated cell lines that express either EGFR-H2 chimeric receptor or HER2 and HER3 receptors in an EGFR-negative background. We show that dose-dependent inhibition of HER2 was achieved at the receptor level, on downstream signaling molecules, and more importantly was also translated into inhibition of cell growth. Our findings imply that the inhibitory effect of erlotinib in HER2-expressing cells may in part be mediated through direct interaction with HER2 rather than indirectly through a process that requires the presence of EGFR. [Cancer Res 2007;67(3):1228–38]
- Published
- 2007
41. Anti-CD20/CD3 T cell-dependent bispecific antibody for the treatment of B cell malignancies
- Author
-
Yu-Waye Chu, Arthur E. Reyes, Mark S. Dennis, Mary Mathieu, Mahrukh Huseni, Justin Scheer, Judy Young, Klara Totpal, Maria Hristopoulos, Clarissa Johnson, Liping L. Sun, Xinhua Wang, Elaine Mai, Xiaocheng Chen, Diego Ellerman, Yijin Li, Teemu T. Junttila, Hong Wang, Allen J. Ebens, Peiyin Wang, Xiao-Jie Yan, Yvonne Chen, Robyn Clark, Eric Stefanich, Nicholas Chiorazzi, and Noel Dybdal
- Subjects
CD3 Complex ,medicine.drug_class ,CD3 ,T cell ,T-Lymphocytes ,Mice, Transgenic ,Monoclonal antibody ,Mice ,Antigen ,immune system diseases ,In vivo ,hemic and lymphatic diseases ,Antibodies, Bispecific ,medicine ,Leukemia, B-Cell ,Animals ,Humans ,B cell ,biology ,General Medicine ,Antigens, CD20 ,Macaca fascicularis ,medicine.anatomical_structure ,Immunology ,biology.protein ,Blinatumomab ,Antibody ,medicine.drug - Abstract
Bispecific antibodies and antibody fragments in various formats have been explored as a means to recruit cytolytic T cells to kill tumor cells. Encouraging clinical data have been reported with molecules such as the anti-CD19/CD3 bispecific T cell engager (BiTE) blinatumomab. However, the clinical use of many reported T cell-recruiting bispecific modalities is limited by liabilities including unfavorable pharmacokinetics, potential immunogenicity, and manufacturing challenges. We describe a B cell-targeting anti-CD20/CD3 T cell-dependent bispecific antibody (CD20-TDB), which is a full-length, humanized immunoglobulin G1 molecule with near-native antibody architecture constructed using "knobs-into-holes" technology. CD20-TDB is highly active in killing CD20-expressing B cells, including primary patient leukemia and lymphoma cells both in vitro and in vivo. In cynomolgus monkeys, CD20-TDB potently depletes B cells in peripheral blood and lymphoid tissues at a single dose of 1 mg/kg while demonstrating pharmacokinetic properties similar to those of conventional monoclonal antibodies. CD20-TDB also exhibits activity in vitro and in vivo in the presence of competing CD20-targeting antibodies. These data provide rationale for the clinical testing of CD20-TDB for the treatment of CD20-expressing B cell malignancies.
- Published
- 2015
42. Role of Protein Kinases and Phosphatases in Receptor Biosynthesis, Shedding and Mediation of Cellular Responses to Tumor Necrosis Factor
- Author
-
Klara Totpal, Masahiro Higuchi, Eva Pocsik, Bharat B. Aggarwal, Kristi Graff, and Warren Liao
- Subjects
Receptor biosynthesis ,Kinase ,Mediation ,Phosphatase ,Tumor necrosis factor alpha ,Biology ,Cell biology - Published
- 2015
43. Synergistic Antiproliferative Effects of TNF and IL-4 against Various Tumor Cell Lines
- Author
-
Klara Totpal and Bharat B. Aggarwal
- Subjects
Chemistry ,Cancer research ,Tumor necrosis factor alpha ,Tumor cells ,Interleukin 4 - Published
- 2015
44. Activation of the Proapoptotic Death Receptor DR5 by Oligomeric Peptide and Antibody Agonists
- Author
-
Stephen J. Russell, Avi Ashkenazi, Andrea G. Cochran, Klara Totpal, Scot A. Marsters, Sachdev S. Sidhu, Sarah G. Hymowitz, Bing Li, and Deanne M. Compaan
- Subjects
Models, Molecular ,Programmed cell death ,Phage display ,Protein Conformation ,medicine.drug_class ,Molecular Sequence Data ,Apoptosis ,Peptide ,Complementarity determining region ,Crystallography, X-Ray ,Monoclonal antibody ,Antibodies ,Receptors, Tumor Necrosis Factor ,Immunoglobulin Fab Fragments ,Peptide Library ,Structural Biology ,Cell Line, Tumor ,medicine ,Humans ,Amino Acid Sequence ,Receptor ,Peptide library ,Molecular Biology ,chemistry.chemical_classification ,Binding Sites ,biology ,Molecular biology ,Cell biology ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,chemistry ,biology.protein ,Antibody ,Oligopeptides - Abstract
The cell-extrinsic apoptotic pathway triggers programmed cell death in response to certain ligands that bind to cell-surface death receptors. Apoptosis is essential for normal development and homeostasis in metazoans, and furthermore, selective activation of the cell-extrinsic pathway in tumor cells holds considerable promise for cancer therapy. We used phage display to identify peptides and synthetic antibodies that specifically bind to the human proapoptotic death receptor DR5. Despite great differences in overall size and structure, the DR5-binding peptides and antibodies shared a tripeptide motif, which was conserved within a disulfide-constrained loop of the peptides and the third complementarity determining region of the antibody heavy chains. The X-ray crystal structure of an antibody in complex with DR5 revealed that the tripeptide motif is buried at the core of the interface, confirming its central role in antigen recognition. We found that certain peptides and antibodies exhibited potent proapoptotic activity against DR5-expressing SK-MES-1 lung carcinoma cells. These phage-derived ligands may be useful for elucidating DR5 activation at the molecular level and for creating synthetic agonists of proapoptotic death receptors.
- Published
- 2006
45. Receptor-selective Mutants of Apoptosis-inducing Ligand 2/Tumor Necrosis Factor-related Apoptosis-inducing Ligand Reveal a Greater Contribution of Death Receptor (DR) 5 than DR4 to Apoptosis Signaling
- Author
-
Stephanie Ho Lindstrom, Sarah G. Hymowitz, Karen Billeci, Robert F. Kelley, Mary Mathieu, Avi Ashkenazi, Klara Totpal, Roger Pai, and Laura DeForge
- Subjects
Models, Molecular ,Programmed cell death ,Protein Conformation ,Apoptosis ,Decoy Receptor 1 ,Biology ,Vascular endothelial growth inhibitor ,Biochemistry ,Receptors, Tumor Necrosis Factor ,Fas ligand ,TNF-Related Apoptosis-Inducing Ligand ,Jurkat Cells ,Humans ,Receptor ,Molecular Biology ,Membrane Glycoproteins ,Tumor Necrosis Factor-alpha ,Cell Biology ,Fas receptor ,Cell biology ,Receptors, TNF-Related Apoptosis-Inducing Ligand ,Mutation ,Signal transduction ,Apoptosis Regulatory Proteins ,Signal Transduction - Abstract
Apoptosis-inducing ligand 2 (Apo2L), also called tumor necrosis factor-related apoptosis-inducing ligand (TRAIL), triggers programmed cell death in various types of cancer cells but not in most normal cells. Apo2L/TRAIL is a homotrimeric protein that interacts with five receptors: death receptor 4 (DR4) and DR5 mediate apoptosis activation, whereas decoy receptor 1 (DcR1), DcR2, and osteoprotegerin counteract this function. Many cancer cell lines express both DR4 and DR5, and each of these receptors can initiate apoptosis independently of the other. However, the relative contribution of DR4 and DR5 to ligand-induced apoptosis is unknown. To investigate this question, we generated death receptor-selective Apo2L/TRAIL variants using a novel approach that enables phage display of mutated trimeric proteins. Selective binding to DR4 or DR5 was achieved with three to six-ligand amino acid substitutions. The DR4-selective Apo2L/TRAIL variants examined in this study showed a markedly reduced ability to trigger apoptosis, whereas the DR5-selective variants had minimally decreased or slightly increased apoptosis-inducing activity. These results suggest that DR5 may contribute more than DR4 to Apo2L/TRAIL-induced apoptosis in cancer cells that express both death receptors.
- Published
- 2005
46. Enhanced tumor killing by Apo2L/TRAIL and CPT-11 co-treatment is associated with p21 cleavage and differential regulation of Apo2L/TRAIL ligand and its receptors
- Author
-
Judith A. Fox, John B. Lowe, Mina Aikawa, Enrique Escandon, Kelly DuPree, Klara Totpal, Dominick Sinicropi, and Hong Xiang
- Subjects
Cyclin-Dependent Kinase Inhibitor p21 ,Cancer Research ,Receptor expression ,Apoptosis ,Biology ,Irinotecan ,medicine.disease_cause ,Receptors, Tumor Necrosis Factor ,TNF-Related Apoptosis-Inducing Ligand ,Downregulation and upregulation ,Cyclins ,Neoplasms ,Tumor Cells, Cultured ,Genetics ,medicine ,Humans ,RNA, Neoplasm ,Decoy receptors ,Receptor ,Molecular Biology ,Cells, Cultured ,Membrane Glycoproteins ,Tumor Necrosis Factor-alpha ,Kinase ,Carcinoma ,Cell Cycle ,Drug Synergism ,Cell cycle ,Antineoplastic Agents, Phytogenic ,Kinetics ,Biochemistry ,Caspases ,Colonic Neoplasms ,Cancer research ,Camptothecin ,Apoptosis Regulatory Proteins ,Carcinogenesis - Abstract
Apo2L/TRAIL exhibits enhanced apoptotic activity in tumor xenograft models when used in combination with the topoisomerase 1 inhibitor CPT-11. To investigate the cellular mechanisms involved in this increased tumor-killing activity, a series of in vitro experiments were conducted using the human colon carcinoma cell line (HCT116). Apo2L/TRAIL induced a transient upregulation of DR5 mRNA, while CPT-11 increased both death and decoy receptor expression. Upregulation of decoy receptors by CPT-11 was partially inhibited by co-administration of Apo2L/TRAIL. CPT-11 treatment resulted in accumulation of cells at G(2)M-phase and correlated with a substantial increase in the protein levels of the cyclin-dependent kinase inhibitor p21. However, cells co-treated with CPT-11 and Apo2L/TRAIL, or pretreated with CPT-11 for up to 24 h followed by 2 h Apo2L/TRAIL, resulted in a caspase-dependent degradation of p21, reversal of G(2)-M phase arrest with a concomitant increase in apoptosis. The sequential treatment produced the greatest induction of DR5 and DR4, caspase-3-like cleavage/activation and p21 degradation, as well as increased apoptosis. These data indicate that the up-regulation of Apo2L/TRAIL ligand and its death receptors as well as cleavage of p21 protein in the Apo2L/TRAIL plus CPT-11 treatment contributes to the positive cooperation between these agents in enhancing tumor cell apoptosis.
- Published
- 2002
47. Tumor-cell resistance to death receptor–induced apoptosis through mutational inactivation of the proapoptotic Bcl-2 homolog Bax
- Author
-
John Morlan, Klara Totpal, Avi Ashkenazi, Eugene Varfolomeev, David A. Lawrence, Ralph H. Schwall, Sharon Fong, Heidi LeBlanc, Peter Schow, and Dominick Sinicropi
- Subjects
Fas-Associated Death Domain Protein ,Poly ADP ribose polymerase ,Transplantation, Heterologous ,Poly (ADP-Ribose) Polymerase-1 ,Mice, Nude ,Apoptosis ,Biology ,Caspase 8 ,Receptors, Tumor Necrosis Factor ,General Biochemistry, Genetics and Molecular Biology ,Frameshift mutation ,TNF-Related Apoptosis-Inducing Ligand ,Mice ,Random Allocation ,Bcl-2-associated X protein ,Proto-Oncogene Proteins ,Tumor Cells, Cultured ,Animals ,Humans ,Receptor ,Adaptor Proteins, Signal Transducing ,Etoposide ,bcl-2-Associated X Protein ,Membrane Glycoproteins ,Tumor Necrosis Factor-alpha ,Proteins ,General Medicine ,Flow Cytometry ,Antineoplastic Agents, Phytogenic ,Molecular biology ,Caspase 9 ,Mitochondria ,Enzyme Activation ,Proto-Oncogene Proteins c-bcl-2 ,Caspases ,Colonic Neoplasms ,Mutation ,Cancer cell ,biology.protein ,Camptothecin ,Female ,Tumor necrosis factor alpha ,Poly(ADP-ribose) Polymerases ,Apoptosis Regulatory Proteins ,Carrier Proteins ,BH3 Interacting Domain Death Agonist Protein ,Signal Transduction - Abstract
The importance of Bax for induction of tumor apoptosis through death receptors remains unclear. Here we show that Bax can be essential for death receptor--mediated apoptosis in cancer cells. Bax-deficient human colon carcinoma cells were resistant to death-receptor ligands, whereas Bax-expressing sister clones were sensitive. Bax was dispensable for apical death-receptor signaling events including caspase-8 activation, but crucial for mitochondrial changes and downstream caspase activation. Treatment of colon tumor cells deficient in DNA mismatch repair with the death-receptor ligand apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) selected in vitro or in vivo for refractory subclones with Bax frameshift mutations including deletions at a novel site. Chemotherapeutic agents upregulated expression of the Apo2L/TRAIL receptor DR5 and the Bax homolog Bak in Baxminus sign/minus sign cells, and restored Apo2L/TRAIL sensitivity in vitro and in vivo. Thus, Bax mutation in mismatch repair--deficient tumors can cause resistance to death receptor--targeted therapy, but pre-exposure to chemotherapy rescues tumor sensitivity.
- Published
- 2002
48. Abstract 3628: T cell-dependent bispecific antibody anti-CD79b/CD3 as a potential therapy for B-cell malignancies
- Author
-
Mark S. Dennis, Maria Hristopoulos, Jessica Li, Tanja S. Zabka, Liping Laura Sun, Cecile Chalouni, Allen J. Ebens, Yvonne Chen, Robert Jinze Li, Eric Stefanich, Christoph Spiess, Mary Mathieu, Siddharth Sukumaran, Stephen E. Gould, Andrew Polson, Klara Totpal, Robyn Clark, Diego Ellerman, Jeffrey Wallin, and Peiyin Wang
- Subjects
Cancer Research ,biology ,business.industry ,medicine.medical_treatment ,T cell ,CD3 ,chemistry.chemical_compound ,Cytokine ,medicine.anatomical_structure ,Oncology ,chemistry ,Antigen ,Ibrutinib ,medicine ,biology.protein ,Cancer research ,Cytotoxic T cell ,Antibody ,business ,B cell - Abstract
T-cell recruiting bispecific antibodies and antibody fragments have been used to harness the cytotoxic potential of T cells for cancer treatment. We have adopted a knobs-into-holes antibody format and produced T-cell dependent bispecific antibodies (TDB), as full length, humanized IgG1 antibodies with natural antibody architecture, which allow one arm to target various B cell antigens while the other arm recruits T cells by binding to the CD3ϵ subunit of the T-cell receptor. One B cell antigen targeted is CD79b, a component of the B cell receptor complex, which has been clinically validated by an anti-CD79b antibody-drug conjugate (ADC) as a safe and effective therapeutic target for B cell malignancies (Leukemia 2015 Pfeifer et.al). In the present work, we show that anti-CD79b/CD3 TDB is active against lymphoma cells with a wide range of CD79b antigen levels in vitro. In addition, anti-CD79b/CD3 TDB appears to be insensitive to the status of cellular signaling pathways in lymphoma cells as it is active against cell lines that are resistant to the BTK inhibitor ibrutinib or an anti-CD79b ADC with a cleavable tubulin inhibitor as the payload. In vivo, anti-CD79b/CD3 TDB administration inhibited tumor growth in B-cell lymphoma xenograft models and resulted in potent B-cell depletion in the blood and spleens in a humanized murine model. To assess the safety of targeting CD79b with a T-cell recruiting bispecific antibody in non-human primates, a surrogate anti-cynoCD79b/CD3 TDB with comparable in vitro potency was produced with a target arm that recognizes cynomolgus monkey CD79b and the same anti-CD3 arm. In a single dose pharmacokinetic/pharmacodynamics/safety study, anti-cynoCD79b/CD3 TDB administration at 1mg/kg resulted in potent B cell depletion, as well as T cell activation and proliferation, which was assessed by flow cytometry on blood and lymphoid tissue and immunohistochemistry on lymphoid tissue. The pharmacokinetic properties of anti-cynoCD79b/CD3 TDB resembled that of IgG antibodies; though with a faster clearance likely due to CD79b antigen internalization and enhanced binding to CD3 when compared to previously described anti-CD20/CD3 TDB. Transient cytokine release was observed as elevated levels of IL-2, IL-6, IFN-gamma and TNF-alpha were detected within 24 hours following administration. Anti-cynoCD79b/CD3 TDB was well-tolerated in the majority of dosed animals without toxicologically significant findings. Collectively, these preclinical data suggest anti-CD79b/CD3 TDB can be a potential therapeutic agent in B cell malignancies. Citation Format: Peiyin Wang, Maria Hristopoulos, Robyn Clark, Yvonne Chen, Diego Ellerman, Mary Mathieu, Christoph Spiess, Jessica Li, Cecile Chalouni, Siddharth Sukumaran, Eric Stefanich, Jeffrey Wallin, Robert Li, Tanja Zabka, Klara Totpal, Mark Dennis, Allen Ebens, Stephen Gould, Andrew Polson, Liping Laura Sun. T cell-dependent bispecific antibody anti-CD79b/CD3 as a potential therapy for B-cell 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 3628. doi:10.1158/1538-7445.AM2017-3628
- Published
- 2017
49. Antitumor efficacy of a bispecific antibody that targets HER2 and activates T cells
- Author
-
Guangmin Li, Bryan Irving, Ji Li, Melissa R. Junttila, Teemu T. Junttila, Elizabeth Luis, Robyn Clark, Mary Mathieu, Maria Hristopoulos, Eric Stefanich, Judy Young, Bu Er Wang, Klara Totpal, Allen J. Ebens, Yijin Li, Mark S. Dennis, Robert F. Kelley, Andrew Polson, Jennifer Johnston, Diego Ellerman, Gail Lewis Phillips, Justin Scheer, and Christoph Spiess
- Subjects
Cancer Research ,Receptor, ErbB-2 ,T-Lymphocytes ,Breast Neoplasms ,Biology ,Pharmacology ,Antibodies, Monoclonal, Humanized ,Lymphocyte Activation ,Rats, Sprague-Dawley ,Immunity ,Trastuzumab ,Cell Line, Tumor ,Antibodies, Bispecific ,medicine ,Animals ,Humans ,skin and connective tissue diseases ,neoplasms ,Cancer ,medicine.disease ,Rats ,Oncology ,Mechanism of action ,Cell culture ,Monoclonal ,Cancer cell ,biology.protein ,Female ,medicine.symptom ,Antibody ,medicine.drug - Abstract
Clinical results from the latest strategies for T-cell activation in cancer have fired interest in combination immunotherapies that can fully engage T-cell immunity. In this study, we describe a trastuzumab-based bispecific antibody, HER2-TDB, which targets HER2 and conditionally activates T cells. HER2-TDB specifically killed HER2-expressing cancer cells at low picomolar concentrations. Because of its unique mechanism of action, which is independent of HER2 signaling or chemotherapeutic sensitivity, HER2-TDB eliminated cells refractory to currently approved HER2 therapies. HER2-TDB exhibited potent antitumor activity in four preclinical model systems, including MMTV-huHER2 and huCD3 transgenic mice. PD-L1 expression in tumors limited HER2-TDB activity, but this resistance could be reversed by anti–PD-L1 treatment. Thus, combining HER2-TDB with anti–PD-L1 yielded a combination immunotherapy that enhanced tumor growth inhibition, increasing the rates and durability of therapeutic response. Cancer Res; 74(19); 5561–71. ©2014 AACR.
- Published
- 2014
50. PAK1 mediates pancreatic cancer cell migration and resistance to MET inhibition
- Author
-
Wei, Zhou, Adrian M, Jubb, Karen, Lyle, Qian, Xiao, Christy C, Ong, Rupal, Desai, Ling, Fu, Florian, Gnad, Qinghua, Song, Peter M, Haverty, Daniela, Aust, Robert, Grützmann, Mally, Romero, Klara, Totpal, Richard M, Neve, Yibing, Yan, William F, Forrest, Yulei, Wang, Rajiv, Raja, Christian, Pilarsky, Ana, de Jesus-Acosta, Marcia, Belvin, Lori S, Friedman, Mark, Merchant, Elizabeth M, Jaffee, Lei, Zheng, Hartmut, Koeppen, and Klaus P, Hoeflich
- Subjects
Antibodies, Monoclonal ,Adenocarcinoma ,Proto-Oncogene Proteins c-met ,Immunohistochemistry ,Article ,Pancreatic Neoplasms ,Disease Models, Animal ,Mice ,Piperidines ,p21-Activated Kinases ,Cell Movement ,Drug Resistance, Neoplasm ,Antineoplastic Combined Chemotherapy Protocols ,Animals ,Azetidines ,Humans ,Signal Transduction - Abstract
Pancreatic adenocarcinoma (PDAC) is a major unmet medical need and a deeper understanding of molecular drivers is needed to advance therapeutic options for patients. We report here that p21-activated kinase 1 (PAK1) is a central node in PDAC cells downstream of multiple growth factor signalling pathways, including hepatocyte growth factor (HGF) and MET receptor tyrosine kinase. PAK1 inhibition blocks signalling to cytoskeletal effectors and tumour cell motility driven by HGF/MET. MET antagonists, such as onartuzumab and crizotinib, are currently in clinical development. Given that even highly effective therapies have resistance mechanisms, we show that combination with PAK1 inhibition overcomes potential resistance mechanisms mediated either by activation of parallel growth factor pathways or by direct amplification of PAK1. Inhibition of PAK1 attenuated in vivo tumour growth and metastasis in a model of pancreatic adenocarcinoma. In human tissues, PAK1 is highly expressed in a proportion of PDACs (33% IHC score 2 or 3; n = 304) and its expression is significantly associated with MET positivity (p < 0.0001) and linked to a widespread metastatic pattern in patients (p = 0.067). Taken together, our results provide evidence for a functional role of MET/PAK1 signalling in pancreatic adenocarcinoma and support further characterization of therapeutic inhibitors in this indication.
- Published
- 2014
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.