108 results on '"Matthew V. Lorenzi"'
Search Results
2. JNJ-64041757 (JNJ-757), a Live, Attenuated, Double-Deleted Listeria monocytogenes–Based Immunotherapy in Patients With NSCLC: Results From Two Phase 1 Studies
- Author
-
Julie R. Brahmer, MD, Melissa L. Johnson, MD, Manuel Cobo, MD, PhD, Santiago Viteri, MD, Juan Coves Sarto, MD, Ammar Sukari, MD, Mark M. Awad, MD, PhD, Ravi Salgia, MD, PhD, Vali A. Papadimitrakopoulou, MD, Arun Rajan, MD, Nibedita Bandyopadhyay, PhD, Alicia J. Allred, PhD, Mark Wade, MA, Gary E. Mason, MD, Enrique Zudaire, PhD, Roland E. Knoblauch, MD, PhD, Nicole Stone, PhD, Matthew V. Lorenzi, PhD, and Raffit Hassan, MD
- Subjects
Non–small cell lung cancer ,Mesothelin ,LADD Lm ,JNJ-757 ,Vaccine ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Introduction: JNJ-64041757 (JNJ-757) is a live, attenuated, double-deleted Listeria monocytogenes–based immunotherapy expressing human mesothelin. JNJ-757 was evaluated in patients with advanced NSCLC as monotherapy (phase 1) and in combination with nivolumab (phase 1b/2). Methods: Patients with stage IIIB/IV NSCLC who had received previous therapy were treated with JNJ-757 (1 × 108 or 1 × 109 colony-forming units [CFUs]) alone (NCT02592967) or JNJ-757 (1 × 109 CFU) plus intravenous nivolumab 240 mg (NCT03371381). Study objectives included the assessment of immunogenicity, safety, and efficacy. Results: In the monotherapy study, 18 patients (median age 63.5 y; women 61%) were treated with JNJ-757 (1 × 108 or 1 × 109 CFU) with a median duration of 1.4 months (range: 0–29). The most common adverse events (AEs) were pyrexia (72%) and chills (61%), which were usually mild and resolved within 48 hours. Peripheral proinflammatory cytokines and lymphocyte activation were induced posttreatment with transient mesothelin-specific T-cell responses in 10 of 13 biomarker-evaluable patients. With monotherapy, four of 18 response-evaluable patients had stable disease of 16 or more weeks, including one patient with a reduction in target lesions. In the combination study, 12 patients were enrolled (median age 63.5 y; women 33%). The most common AEs with combination therapy were pyrexia (67%) and chills (58%); six patients had grade 3 AEs or greater, including two cases of treatment-related fatal pneumonitis. The best overall response for the combination was stable disease in four of nine response-evaluable patients. Conclusions: As monotherapy, JNJ-757 was immunogenic and tolerable, with mild infusion-related fever and chills. The limited efficacy of JNJ-757, alone or with nivolumab, did not warrant further investigation of the combination.
- Published
- 2021
- Full Text
- View/download PDF
3. Data from The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor Immunity
- Author
-
Matthew V. Lorenzi, Raluca I. Verona, Kwok-Kin Wong, Christopher Moy, Pasi A. Janne, Kathryn Packman, Paul T. Kirschmeier, Mark A. Bittinger, Sylvie Laquerre, Catherine Sanders, Julie A. Rytlewski, Andrew H. Beck, Aditya Khosla, Jessie M. English, Catherine Ferrante, Elena Ivanova, Samuel N. Regan, Dyane Bailey, Martha R. Gowaski, Kristin DePeaux, Abha Dhaneshwar, Jeffrey Liu, Dennis M. Bonal, Wei Huang, Enrique Zudaire, Sima J. Zacharek, Mari Kuraguchi, and Sangeetha Palakurthi
- Abstract
The success of targeted or immune therapies is often hampered by the emergence of resistance and/or clinical benefit in only a subset of patients. We hypothesized that combining targeted therapy with immune modulation would show enhanced antitumor responses. Here, we explored the combination potential of erdafitinib, a fibroblast growth factor receptor (FGFR) inhibitor under clinical development, with PD-1 blockade in an autochthonous FGFR2K660N/p53mut lung cancer mouse model. Erdafitinib monotherapy treatment resulted in substantial tumor control but no significant survival benefit. Although anti–PD-1 alone was ineffective, the erdafitinib and anti–PD-1 combination induced significant tumor regression and improved survival. For both erdafitinib monotherapy and combination treatments, tumor control was accompanied by tumor-intrinsic, FGFR pathway inhibition, increased T-cell infiltration, decreased regulatory T cells, and downregulation of PD-L1 expression on tumor cells. These effects were not observed in a KRASG12C-mutant genetically engineered mouse model, which is insensitive to FGFR inhibition, indicating that the immune changes mediated by erdafitinib may be initiated as a consequence of tumor cell killing. A decreased fraction of tumor-associated macrophages also occurred but only in combination-treated tumors. Treatment with erdafitinib decreased T-cell receptor (TCR) clonality, reflecting a broadening of the TCR repertoire induced by tumor cell death, whereas combination with anti–PD-1 led to increased TCR clonality, suggesting a more focused antitumor T-cell response. Our results showed that the combination of erdafitinib and anti–PD-1 drives expansion of T-cell clones and immunologic changes in the tumor microenvironment to support enhanced antitumor immunity and survival.
- Published
- 2023
- Full Text
- View/download PDF
4. Supplementary Figure Legends, Tables and Methods from The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor Immunity
- Author
-
Matthew V. Lorenzi, Raluca I. Verona, Kwok-Kin Wong, Christopher Moy, Pasi A. Janne, Kathryn Packman, Paul T. Kirschmeier, Mark A. Bittinger, Sylvie Laquerre, Catherine Sanders, Julie A. Rytlewski, Andrew H. Beck, Aditya Khosla, Jessie M. English, Catherine Ferrante, Elena Ivanova, Samuel N. Regan, Dyane Bailey, Martha R. Gowaski, Kristin DePeaux, Abha Dhaneshwar, Jeffrey Liu, Dennis M. Bonal, Wei Huang, Enrique Zudaire, Sima J. Zacharek, Mari Kuraguchi, and Sangeetha Palakurthi
- Abstract
Supplemental figure legends 1-8, Supplementary tables 1-5, and Supplementary methods
- Published
- 2023
- Full Text
- View/download PDF
5. Supplemental Figures 1-8 from The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor Immunity
- Author
-
Matthew V. Lorenzi, Raluca I. Verona, Kwok-Kin Wong, Christopher Moy, Pasi A. Janne, Kathryn Packman, Paul T. Kirschmeier, Mark A. Bittinger, Sylvie Laquerre, Catherine Sanders, Julie A. Rytlewski, Andrew H. Beck, Aditya Khosla, Jessie M. English, Catherine Ferrante, Elena Ivanova, Samuel N. Regan, Dyane Bailey, Martha R. Gowaski, Kristin DePeaux, Abha Dhaneshwar, Jeffrey Liu, Dennis M. Bonal, Wei Huang, Enrique Zudaire, Sima J. Zacharek, Mari Kuraguchi, and Sangeetha Palakurthi
- Abstract
Supplemental Figures 1-8
- Published
- 2023
- Full Text
- View/download PDF
6. Supplementary Movie M5 from Amivantamab (JNJ-61186372), an Fc Enhanced EGFR/cMet Bispecific Antibody, Induces Receptor Downmodulation and Antitumor Activity by Monocyte/Macrophage Trogocytosis
- Author
-
Sheri L. Moores, Sylvie Laquerre, Matthew V. Lorenzi, Kathryn Packman, Hillary J. Millar, Marilda Beqiri, Jocelyn Sendecki, Ryan Lenhart, Benjamin Henley, Barbara S. Bushey, Kristen Chevalier, Lorraine Lipfert, and Smruthi Vijayaraghavan
- Abstract
Live cell imaging of M2 macrophage mediated trogocytosis in adherent co-culture
- Published
- 2023
- Full Text
- View/download PDF
7. Supplementary Text from Amivantamab (JNJ-61186372), an Fc Enhanced EGFR/cMet Bispecific Antibody, Induces Receptor Downmodulation and Antitumor Activity by Monocyte/Macrophage Trogocytosis
- Author
-
Sheri L. Moores, Sylvie Laquerre, Matthew V. Lorenzi, Kathryn Packman, Hillary J. Millar, Marilda Beqiri, Jocelyn Sendecki, Ryan Lenhart, Benjamin Henley, Barbara S. Bushey, Kristen Chevalier, Lorraine Lipfert, and Smruthi Vijayaraghavan
- Abstract
Contains Supplementary methods, supp figure legends
- Published
- 2023
- Full Text
- View/download PDF
8. Supplemental Figure 1 from Discovery and Pharmacological Characterization of JNJ-42756493 (Erdafitinib), a Functionally Selective Small-Molecule FGFR Family Inhibitor
- Author
-
Matthew V. Lorenzi, Sylvie Laquerre, Patrick Angibaud, Christopher Moy, Jayaprakash D. Karkera, Suso J. Platero, Jennifer Yang, Liang Xie, Na Cheng, David R. Newell, Neil T. Thompson, George Ward, Ron Gilissen, Christopher W. Murray, Martin Page, Gordon Saxty, Matthew Squires, David C. Rees, Eddy Freyne, Peter King, Kelly Van De Ven, Caroline Paulussen, Tinne Verhulst, Desiree De Lange, Jorge Vialard, Laurence Mevellec, Eleonora Jovcheva, and Timothy P.S. Perera
- Abstract
A) Structure of JNJ-42541707, a structurally related compound to JNJ-42756493. B) 72h growth inhibition (IC50) of JNJ-42541707 against 236 cancer cell lines from multiple origins color coded based on FGFR1,2,4 mRNA overexpression and FGFR WT.
- Published
- 2023
- Full Text
- View/download PDF
9. Supplementary Table S1-S2 and Figure S1-S2 from Oncogenic Characterization and Pharmacologic Sensitivity of Activating Fibroblast Growth Factor Receptor (FGFR) Genetic Alterations to the Selective FGFR Inhibitor Erdafitinib
- Author
-
Suso J. Platero, Matthew V. Lorenzi, John D. Alvarez, Feng R. Luo, Rastislav Bahleda, Michael Sharp, Peter King, Christopher H. Moy, Ademi Santiago-Walker, Joseph C. Portale, Dana Gaffney, Katherine Bell, Gabriela Martinez Cardona, and Jayaprakash D. Karkera
- Abstract
Table S1. Frequencies of gene locus amplifications across tumor types; Table S2. Effect of FGFR inhibitors on growth of RK3E expressing FGFR2 and FGFR3 fusion constructs and RT-4 bladder cancer cell line; Figure S1. Chemical structure of JNJ-42756493; Figure S2. Western Blot confirming the overexpression of HA-tagged FGFR2 fusion genes
- Published
- 2023
- Full Text
- View/download PDF
10. Data from Amivantamab (JNJ-61186372), an Fc Enhanced EGFR/cMet Bispecific Antibody, Induces Receptor Downmodulation and Antitumor Activity by Monocyte/Macrophage Trogocytosis
- Author
-
Sheri L. Moores, Sylvie Laquerre, Matthew V. Lorenzi, Kathryn Packman, Hillary J. Millar, Marilda Beqiri, Jocelyn Sendecki, Ryan Lenhart, Benjamin Henley, Barbara S. Bushey, Kristen Chevalier, Lorraine Lipfert, and Smruthi Vijayaraghavan
- Abstract
Small molecule inhibitors targeting mutant EGFR are standard of care in non–small cell lung cancer (NSCLC), but acquired resistance invariably develops through mutations in EGFR or through activation of compensatory pathways such as cMet. Amivantamab (JNJ-61186372) is an anti-EGFR and anti-cMet bispecific low fucose antibody with enhanced Fc function designed to treat tumors driven by activated EGFR and/or cMet signaling. Potent in vivo antitumor efficacy is observed upon amivantamab treatment of human tumor xenograft models driven by mutant activated EGFR, and this activity is associated with receptor downregulation. Despite these robust antitumor responses in vivo, limited antiproliferative effects and EGFR/cMet receptor downregulation by amivantamab were observed in vitro. Interestingly, in vitro addition of isolated human immune cells notably enhanced amivantamab-mediated EGFR and cMet downregulation, leading to antibody dose-dependent cancer cell killing. Through a comprehensive assessment of the Fc-mediated effector functions, we demonstrate that monocytes and/or macrophages, through trogocytosis, are necessary and sufficient for Fc interaction-mediated EGFR/cMet downmodulation and are required for in vivo antitumor efficacy. Collectively, our findings represent a novel Fc-dependent macrophage-mediated antitumor mechanism of amivantamab and highlight trogocytosis as an important mechanism of action to exploit in designing new antibody-based cancer therapies.
- Published
- 2023
- Full Text
- View/download PDF
11. Supplementary Figures from Amivantamab (JNJ-61186372), an Fc Enhanced EGFR/cMet Bispecific Antibody, Induces Receptor Downmodulation and Antitumor Activity by Monocyte/Macrophage Trogocytosis
- Author
-
Sheri L. Moores, Sylvie Laquerre, Matthew V. Lorenzi, Kathryn Packman, Hillary J. Millar, Marilda Beqiri, Jocelyn Sendecki, Ryan Lenhart, Benjamin Henley, Barbara S. Bushey, Kristen Chevalier, Lorraine Lipfert, and Smruthi Vijayaraghavan
- Abstract
Contains supp figures S1-S13 and the corresponding figure legends
- Published
- 2023
- Full Text
- View/download PDF
12. Supplemental Table 1 from Discovery and Pharmacological Characterization of JNJ-42756493 (Erdafitinib), a Functionally Selective Small-Molecule FGFR Family Inhibitor
- Author
-
Matthew V. Lorenzi, Sylvie Laquerre, Patrick Angibaud, Christopher Moy, Jayaprakash D. Karkera, Suso J. Platero, Jennifer Yang, Liang Xie, Na Cheng, David R. Newell, Neil T. Thompson, George Ward, Ron Gilissen, Christopher W. Murray, Martin Page, Gordon Saxty, Matthew Squires, David C. Rees, Eddy Freyne, Peter King, Kelly Van De Ven, Caroline Paulussen, Tinne Verhulst, Desiree De Lange, Jorge Vialard, Laurence Mevellec, Eleonora Jovcheva, and Timothy P.S. Perera
- Abstract
In vitro kinase inhibition by DiscoverX KinomeScan assay. For details see (19)
- Published
- 2023
- Full Text
- View/download PDF
13. Supplemental Table 2 from Discovery and Pharmacological Characterization of JNJ-42756493 (Erdafitinib), a Functionally Selective Small-Molecule FGFR Family Inhibitor
- Author
-
Matthew V. Lorenzi, Sylvie Laquerre, Patrick Angibaud, Christopher Moy, Jayaprakash D. Karkera, Suso J. Platero, Jennifer Yang, Liang Xie, Na Cheng, David R. Newell, Neil T. Thompson, George Ward, Ron Gilissen, Christopher W. Murray, Martin Page, Gordon Saxty, Matthew Squires, David C. Rees, Eddy Freyne, Peter King, Kelly Van De Ven, Caroline Paulussen, Tinne Verhulst, Desiree De Lange, Jorge Vialard, Laurence Mevellec, Eleonora Jovcheva, and Timothy P.S. Perera
- Abstract
Inhibitory activity of Brivanib and JNJ-42756943 in kinase (top) and BaF3 kinase dependent proliferation (bottom) assays and ratio of FGFRs/VEGFR2 activities.
- Published
- 2023
- Full Text
- View/download PDF
14. Supplemental Table 3 from Discovery and Pharmacological Characterization of JNJ-42756493 (Erdafitinib), a Functionally Selective Small-Molecule FGFR Family Inhibitor
- Author
-
Matthew V. Lorenzi, Sylvie Laquerre, Patrick Angibaud, Christopher Moy, Jayaprakash D. Karkera, Suso J. Platero, Jennifer Yang, Liang Xie, Na Cheng, David R. Newell, Neil T. Thompson, George Ward, Ron Gilissen, Christopher W. Murray, Martin Page, Gordon Saxty, Matthew Squires, David C. Rees, Eddy Freyne, Peter King, Kelly Van De Ven, Caroline Paulussen, Tinne Verhulst, Desiree De Lange, Jorge Vialard, Laurence Mevellec, Eleonora Jovcheva, and Timothy P.S. Perera
- Abstract
JNJ-42756493 anti-proliferative activity against cancer cells lines from multiple origins.. Detailed data supporting Figure 2.
- Published
- 2023
- Full Text
- View/download PDF
15. Supplemental Table S1 from A Novel Bispecific Antibody Targeting EGFR and cMet Is Effective against EGFR Inhibitor–Resistant Lung Tumors
- Author
-
G. Mark Anderson, Matthew V. Lorenzi, Sylvie Laquerre, Ricardo M. Attar, Janine Schuurman, Paul W.H.I. Parren, Joost Neijssen, Pauline L. Martin, Sheng-Jiun Wu, Thomas Kelly, Peter Haytko, Randall J. Brezski, Keri Dorn, Leopoldo Luistro, Kristen Chevalier, Barbara S. Bushey, Mark L. Chiu, and Sheri L. Moores
- Abstract
Origins and Genotypes of Lung Cancer Tumor Cell Lines
- Published
- 2023
- Full Text
- View/download PDF
16. Data from A Novel Bispecific Antibody Targeting EGFR and cMet Is Effective against EGFR Inhibitor–Resistant Lung Tumors
- Author
-
G. Mark Anderson, Matthew V. Lorenzi, Sylvie Laquerre, Ricardo M. Attar, Janine Schuurman, Paul W.H.I. Parren, Joost Neijssen, Pauline L. Martin, Sheng-Jiun Wu, Thomas Kelly, Peter Haytko, Randall J. Brezski, Keri Dorn, Leopoldo Luistro, Kristen Chevalier, Barbara S. Bushey, Mark L. Chiu, and Sheri L. Moores
- Abstract
Non–small cell lung cancers (NSCLC) with activating EGFR mutations become resistant to tyrosine kinase inhibitors (TKI), often through second-site mutations in EGFR (T790M) and/or activation of the cMet pathway. We engineered a bispecific EGFR-cMet antibody (JNJ-61186372) with multiple mechanisms of action to inhibit primary/secondary EGFR mutations and the cMet pathway. JNJ-61186372 blocked ligand-induced phosphorylation of EGFR and cMet and inhibited phospho-ERK and phospho-AKT more potently than the combination of single receptor–binding antibodies. In NSCLC tumor models driven by EGFR and/or cMet, JNJ-61186372 treatment resulted in tumor regression through inhibition of signaling/receptor downmodulation and Fc-driven effector interactions. Complete and durable regression of human lung xenograft tumors was observed with the combination of JNJ-61186372 and a third-generation EGFR TKI. Interestingly, treatment of cynomolgus monkeys with JNJ-61186372 resulted in no major toxicities, including absence of skin rash observed with other EGFR-directed agents. These results highlight the differentiated potential of JNJ-61186372 to inhibit the spectrum of mutations driving EGFR TKI resistance in NSCLC. Cancer Res; 76(13); 3942–53. ©2016 AACR.
- Published
- 2023
- Full Text
- View/download PDF
17. Supplemental Figure S5 from A Novel Bispecific Antibody Targeting EGFR and cMet Is Effective against EGFR Inhibitor–Resistant Lung Tumors
- Author
-
G. Mark Anderson, Matthew V. Lorenzi, Sylvie Laquerre, Ricardo M. Attar, Janine Schuurman, Paul W.H.I. Parren, Joost Neijssen, Pauline L. Martin, Sheng-Jiun Wu, Thomas Kelly, Peter Haytko, Randall J. Brezski, Keri Dorn, Leopoldo Luistro, Kristen Chevalier, Barbara S. Bushey, Mark L. Chiu, and Sheri L. Moores
- Abstract
Efficacy of JNJ-61186372 in H292-HGF Xenograft Tumors
- Published
- 2023
- Full Text
- View/download PDF
18. Antitumor Activity of Amivantamab (JNJ-61186372), an EGFR–MET Bispecific Antibody, in Diverse Models of EGFR Exon 20 Insertion–Driven NSCLC
- Author
-
A. Roshak, Chae Won Park, Matthew V. Lorenzi, Seong Gu Heo, Min Hee Hong, Mi Ran Yun, Sun Min Lim, Soo-Hwan Lee, Meena Thayu, Kyoung Ho Pyo, Byoung Chul Cho, Seo Yoon Jeong, Hye Ryun Kim, Seok Young Kim, S.M. Lim, J.C. Curtin, Jiyeon Yun, Jae Hwan Kim, and Roland Elmar Knoblauch
- Subjects
0301 basic medicine ,Antitumor activity ,Bispecific antibody ,Cetuximab ,business.industry ,Poziotinib ,respiratory tract diseases ,Phase i study ,03 medical and health sciences ,Exon ,030104 developmental biology ,0302 clinical medicine ,Oncology ,In vivo ,030220 oncology & carcinogenesis ,Cancer research ,medicine ,Secretion ,business ,medicine.drug - Abstract
EGFR exon 20 insertion driver mutations (Exon20ins) in non–small cell lung cancer (NSCLC) are insensitive to EGFR tyrosine kinase inhibitors (TKI). Amivantamab (JNJ-61186372), a bispecific antibody targeting EGFR–MET, has shown preclinical activity in TKI-sensitive EGFR-mutated NSCLC models and in an ongoing first-in-human study in patients with advanced NSCLC. However, the activity of amivantamab in Exon20ins-driven tumors has not yet been described. Ba/F3 cells and patient-derived cells/organoids/xenograft models harboring diverse Exon20ins were used to characterize the antitumor mechanism of amivantamab. Amivantamab inhibited proliferation by effectively downmodulating EGFR–MET levels and inducing immune-directed antitumor activity with increased IFNγ secretion in various models. Importantly, in vivo efficacy of amivantamab was superior to cetuximab or poziotinib, an experimental Exon20ins-targeted TKI. Amivantamab produced robust tumor responses in two Exon20ins patients, highlighting the important translational nature of this preclinical work. These findings provide mechanistic insight into the activity of amivantamab and support its continued clinical development in Exon20ins patients, an area of high unmet medical need. Significance: Currently, there are no approved targeted therapies for EGFR Exon20ins–driven NSCLC. Preclinical data shown here, together with promising clinical activity in an ongoing phase I study, strongly support further clinical investigation of amivantamab in EGFR Exon20ins–driven NSCLC. This article is highlighted in the In This Issue feature, p. 1079
- Published
- 2020
- Full Text
- View/download PDF
19. Discovery and pharmacological characterization of cetrelimab (JNJ-63723283), an anti-programmed cell death protein-1 (PD-1) antibody, in human cancer models
- Author
-
Nikki DeAngelis, Catherine Ferrante, Gordon Powers, Jocelyn Sendecki, Bethany Mattson, Darlene Pizutti, Kathryn Packman, Weirong Wang, Kevin Trouba, Rupesh Nanjunda, John Wheeler, Ray Brittingham, Sheng-Jiun Wu, Jinquan Luo, Matthew V. Lorenzi, and Raluca I. Verona
- Subjects
Pharmacology ,Cancer Research ,Programmed Cell Death 1 Receptor ,Antibodies, Monoclonal ,Toxicology ,Antibodies, Monoclonal, Humanized ,Disease Models, Animal ,Macaca fascicularis ,Mice ,Oncology ,Neoplasms ,Animals ,Cytokines ,Humans ,Pharmacology (medical) ,Immune Checkpoint Inhibitors - Abstract
Purpose Preclinical characterization of cetrelimab (JNJ-63723283), a fully humanized immunoglobulin G4 kappa monoclonal antibody targeting programmed cell death protein-1 (PD-1), in human cancer models. Methods Cetrelimab was generated by phage panning against human and cynomolgus monkey (cyno) PD-1 extracellular domains (ECDs) and affinity maturation. Binding to primate and rodent PD-1 ECDs, transfected and endogenous cell-surface PD-1, and inhibition of ligand binding were measured. In vitro activity was evaluated using cytomegalovirus recall, mixed lymphocyte reaction, staphylococcal enterotoxin B stimulation, and Jurkat-PD-1 nuclear factor of activated T cell reporter assays. In vivo activity was assessed using human PD-1 knock-in mice implanted with MC38 tumors and a lung patient-derived xenograft (PDX) model (LG1306) using CD34 cord-blood-humanized NSG mice. Pharmacodynamics, toxicokinetics, and safety were assessed in cynos following single and/or repeat intravenous dosing. Results Cetrelimab showed high affinity binding to human (1.72 nM) and cyno (0.90 nM) PD-1 and blocked binding of programmed death-ligand 1 (PD-L1; inhibitory concentration [IC] 111.7 ng/mL) and PD-L2 (IC 138.6 ng/mL). Cetrelimab dose-dependently increased T cell-mediated cytokine production and stimulated cytokine expression. Cetrelimab 10 mg/kg reduced mean MC38 tumor volume in PD-1 knock-in mice at Day 21 (P P Conclusion Cetrelimab potently inhibits PD-1 in vitro and in vivo, supporting its clinical evaluation.
- Published
- 2021
20. The Combined Effect of FGFR Inhibition and PD-1 Blockade Promotes Tumor-Intrinsic Induction of Antitumor Immunity
- Author
-
Paul Kirschmeier, Jeffrey Liu, Andrew H. Beck, Catherine Ferrante, Sangeetha Palakurthi, Christopher Moy, Elena Ivanova, Mark A. Bittinger, Martha R. Gowaski, Abha Dhaneshwar, Dennis M. Bonal, Mari Kuraguchi, Matthew V. Lorenzi, Kwok-Kin Wong, Kristin Depeaux, Samuel N. Regan, Jessie M. English, Catherine Sanders, Raluca I. Verona, Julie A. Rytlewski, Pasi A. Jänne, Kathryn Packman, Dyane Bailey, Enrique Zudaire, Wei Huang, Sima Zacharek, Sylvie Laquerre, and Aditya Khosla
- Subjects
0301 basic medicine ,Cancer Research ,medicine.medical_treatment ,FGFR Inhibition ,Programmed Cell Death 1 Receptor ,Immunology ,Receptors, Antigen, T-Cell ,Mice, Transgenic ,Biology ,Immunophenotyping ,Targeted therapy ,Mice ,03 medical and health sciences ,Antineoplastic Agents, Immunological ,Lymphocytes, Tumor-Infiltrating ,0302 clinical medicine ,Immune system ,Downregulation and upregulation ,Erdafitinib ,T-Lymphocyte Subsets ,Cell Line, Tumor ,Neoplasms ,Quinoxalines ,Tumor Microenvironment ,medicine ,Animals ,Humans ,Tumor microenvironment ,T-cell receptor ,Immunity ,Drug Synergism ,Prognosis ,Receptors, Fibroblast Growth Factor ,Disease Models, Animal ,Treatment Outcome ,030104 developmental biology ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Pyrazoles ,Biomarkers ,Signal Transduction - Abstract
The success of targeted or immune therapies is often hampered by the emergence of resistance and/or clinical benefit in only a subset of patients. We hypothesized that combining targeted therapy with immune modulation would show enhanced antitumor responses. Here, we explored the combination potential of erdafitinib, a fibroblast growth factor receptor (FGFR) inhibitor under clinical development, with PD-1 blockade in an autochthonous FGFR2K660N/p53mut lung cancer mouse model. Erdafitinib monotherapy treatment resulted in substantial tumor control but no significant survival benefit. Although anti–PD-1 alone was ineffective, the erdafitinib and anti–PD-1 combination induced significant tumor regression and improved survival. For both erdafitinib monotherapy and combination treatments, tumor control was accompanied by tumor-intrinsic, FGFR pathway inhibition, increased T-cell infiltration, decreased regulatory T cells, and downregulation of PD-L1 expression on tumor cells. These effects were not observed in a KRASG12C-mutant genetically engineered mouse model, which is insensitive to FGFR inhibition, indicating that the immune changes mediated by erdafitinib may be initiated as a consequence of tumor cell killing. A decreased fraction of tumor-associated macrophages also occurred but only in combination-treated tumors. Treatment with erdafitinib decreased T-cell receptor (TCR) clonality, reflecting a broadening of the TCR repertoire induced by tumor cell death, whereas combination with anti–PD-1 led to increased TCR clonality, suggesting a more focused antitumor T-cell response. Our results showed that the combination of erdafitinib and anti–PD-1 drives expansion of T-cell clones and immunologic changes in the tumor microenvironment to support enhanced antitumor immunity and survival.
- Published
- 2019
- Full Text
- View/download PDF
21. Discovery and Pharmacological Characterization of JNJ-64619178, a Novel Small-Molecule Inhibitor of PRMT5 with Potent Antitumor Activity
- Author
-
Tinne Verhulst, Lieven Meerpoel, Emmanuel Gustin, Weimei Sun, Matthew V. Lorenzi, Junchen Gu, Longen Zhou, Wim Bert Griet Schepens, Hillary Millar, Tongfei Wu, Viellevoye Marcel, Vineet Pande, Petra Vinken, Desiree De Lange, An Boeckx, Christopher Moy, Friederike Pastore, Geert Mannens, Sylvie Laquerre, Ulrike Philippar, Vipul Bhargava, Gaston Stanislas Marcella Diels, Thomas Nys, Kathryn Packman, Jan Willem Thuring, Erika van Heerde, Bie Verbist, Sumit Rai, Lijs Beke, Pegah Safabakhsh, Timothy A. Graubert, Yue Fan, Angelique N Gilbert, Dirk Brehmer, Vikki Keersmaekers, Barbara Morschhäuser, Danilo Fiore, David Walker, Amy J. Johnson, Brehmer, D., Beke, L., Wu, T., Millar, H. J., Moy, C., Sun, W., Mannens, G., Pande, V., Boeckx, A., van Heerde, E., Nys, T., Gustin, E. M., Verbist, B., Zhou, L., Fan, Y., Bhargava, V., Safabakhsh, P., Vinken, P., Verhulst, T., Gilbert, A., Rai, S., Graubert, T. A., Pastore, F., Fiore, D., Gu, J., Johnson, A., Philippar, U., Morschhauser, B., Walker, D., de Lange, D., Keersmaekers, V., Viellevoye, M., Diels, G., Schepens, W., Thuring, J. W., Meerpoel, L., Packman, K., Lorenzi, M. V., and Laquerre, S.
- Subjects
Cancer Research ,Protein-Arginine N-Methyltransferases ,Lung Neoplasms ,PROTEIN ,Splicing factor ,Mice ,In vivo ,REVEALS ,medicine ,Animals ,Humans ,Pyrroles ,Enzyme Inhibitors ,Lung cancer ,VULNERABILITY ,Science & Technology ,business.industry ,Protein arginine methyltransferase 5 ,PRE-MESSENGER-RNA ,METHYLATION ,Myeloid leukemia ,SELECTIVE INHIBITOR ,medicine.disease ,Lymphoma ,Disease Models, Animal ,Pyrimidines ,Oncology ,Cancer research ,ARGININE METHYLTRANSFERASE PRMT5 ,Signal transduction ,business ,Life Sciences & Biomedicine ,Ex vivo - Abstract
The protein arginine methyltransferase 5 (PRMT5) methylates a variety of proteins involved in splicing, multiple signal transduction pathways, epigenetic control of gene expression, and mechanisms leading to protein expression required for cellular proliferation. Dysregulation of PRMT5 is associated with clinical features of several cancers, including lymphomas, lung cancer, and breast cancer. Here, we describe the characterization of JNJ-64619178, a novel, selective, and potent PRMT5 inhibitor, currently in clinical trials for patients with advanced solid tumors, non-Hodgkin's lymphoma, and lower-risk myelodysplastic syndrome. JNJ-64619178 demonstrated a prolonged inhibition of PRMT5 and potent antiproliferative activity in subsets of cancer cell lines derived from various histologies, including lung, breast, pancreatic, and hematological malignancies. In primary acute myelogenous leukemia samples, the presence of splicing factor mutations correlated with a higher ex vivo sensitivity to JNJ-64619178. Furthermore, the potent and unique mechanism of inhibition of JNJ-64619178, combined with highly optimized pharmacological properties, led to efficient tumor growth inhibition and regression in several xenograft models in vivo, with once-daily or intermittent oral-dosing schedules. An increase in splicing burden was observed upon JNJ-64619178 treatment. Overall, these observations support the continued clinical evaluation of JNJ-64619178 in patients with aberrant PRMT5 activity-driven tumors. ispartof: MOLECULAR CANCER THERAPEUTICS vol:20 issue:12 pages:2317-2328 ispartof: location:United States status: published
- Published
- 2021
22. Abstract ND07: JNJ-78306358: A first-in-class bispecific T cell redirecting HLA-G antibody
- Author
-
Nataša Obermajer, Adam Zwolak, Kelly Van De Ven, Shana Versmissen, Aleksandra Brajic, Ted Petley, Dan Weinstock, Jason Aligo, Fang Yi, Stephen Jarantow, Keith Schutsky, Ken Tian, Angelilo Lorraine, Diana Alvarez Arias, Kristel Buyens, Vince Torti, Krista Menard, Katharine Rogers, Brian Geist, Marjolein Van Heerden, Gerald Chu, Bie Verbist, Maté Ongenaert, Julien Hasler, Kathryn Packman, Jacintha Shenton, Dirk Brehmer, Josh Lauring, Regina J. Brown, James Greger, Daphne Salick Ryan, Sanjaya Singh, Matthew V. Lorenzi, Laurie Lenox, and Sylvie Laquerre
- Subjects
Cancer Research ,Oncology - Abstract
JNJ-78306358 is a first-in-class bispecific antibody (bsAb), engineered using the Zymeworks Azymetric™ platform, to treat advanced stage solid tumors. Human leukocyte antigen G (HLA-G) is a non-classical major histocompatibility class I molecule with an immune tolerance role at the maternal-fetal interface. HLA-G has limited normal tissue expression, mainly detected in placenta and pituitary gland. However, HLA-G is expressed in multiple human cancers, with a potential role in cancer immune evasion. Comprehensive immunohistochemistry analysis of patient-derived tumors revealed high prevalence of HLA-G expression in renal cell carcinoma (RCC, 75%), ovarian (61%), colon (64%) and rectal cancers (40%), and moderate HLA-G expression prevalence in lung adenocarcinoma, endometrial, and pancreatic cancer. JNJ-78306358 induces HLA-G-expressing tumor cell killing via T cell redirection. This bsAb features an anti-HLA-G single-chain fragment variable (scFv) domain that binds with high affinity (KD ~ 13 pM) to HLA-G on tumor cells and a Fab domain that binds with weaker affinity (KD ~22 nM) to the epsilon subunit of the cluster of differentiation 3 (CD3ε). The immunoglobulin (Ig)G1 heavy chains feature Fc region mutations that disrupt interaction with Fcγ receptors. JNJ-78306358 demonstrated potent PBMC- and T cell-mediated in vitro cytotoxicity (EC50 10.4 - 442.3 pM) against endogenous membrane HLA-G-expressing tumor cell lines and absence of killing against cancer cells lacking HLA-G membrane expression, highlighting the specificity against antigen-expressing tumor cells. JNJ-78306358 exhibited hallmarks of T cell engagement in vitro, including T cell proliferation and cytokine release. In addition, JNJ-78306358 showed HLA-G-expression-dependent anti-tumor activity in mice (humanized with human donor CD3+ pan-T cells or human umbilical cord-blood-derived CD34+ hematopoietic stem cells [HSCs]) bearing cell line- and patient-derived tumors. In these xenograft models, a dose-dependent increase in CD4+ and CD8+ T cell infiltration into tumors was observed with complete tumor regressions at low doses of JNJ-78306358 (0.03 mg/kg). JNJ-78306358’s safety, tolerability and preliminary anti-tumor activity are currently being evaluated in a first-in-human phase I study in advanced stage solid tumors with high prevalence of HLA-G protein expression (NCT04991740). This antigen-targeting approach may address an unmet medical need in patients with tumors expressing HLA-G. Citation Format: Nataša Obermajer, Adam Zwolak, Kelly Van De Ven, Shana Versmissen, Aleksandra Brajic, Ted Petley, Dan Weinstock, Jason Aligo, Fang Yi, Stephen Jarantow, Keith Schutsky, Ken Tian, Angelilo Lorraine, Diana Alvarez Arias, Kristel Buyens, Vince Torti, Krista Menard, Katharine Rogers, Brian Geist, Marjolein Van Heerden, Gerald Chu, Bie Verbist, Maté Ongenaert, Julien Hasler, Kathryn Packman, Jacintha Shenton, Dirk Brehmer, Josh Lauring, Regina J. Brown, James Greger, Daphne Salick Ryan, Sanjaya Singh, Matthew V. Lorenzi, Laurie Lenox, Sylvie Laquerre. JNJ-78306358: A first-in-class bispecific T cell redirecting HLA-G antibody [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr ND07.
- Published
- 2022
- Full Text
- View/download PDF
23. JNJ-64041757 (JNJ-757), a Live, Attenuated, Double-Deleted
- Author
-
Julie R, Brahmer, Melissa L, Johnson, Manuel, Cobo, Santiago, Viteri, Juan Coves, Sarto, Ammar, Sukari, Mark M, Awad, Ravi, Salgia, Vali A, Papadimitrakopoulou, Arun, Rajan, Nibedita, Bandyopadhyay, Alicia J, Allred, Mark, Wade, Gary E, Mason, Enrique, Zudaire, Roland E, Knoblauch, Nicole, Stone, Matthew V, Lorenzi, and Raffit, Hassan
- Subjects
Mesothelin ,LADD Lm ,Non–small cell lung cancer ,JNJ-757 ,Original Article ,Vaccine - Abstract
Introduction JNJ-64041757 (JNJ-757) is a live, attenuated, double-deleted Listeria monocytogenes–based immunotherapy expressing human mesothelin. JNJ-757 was evaluated in patients with advanced NSCLC as monotherapy (phase 1) and in combination with nivolumab (phase 1b/2). Methods Patients with stage IIIB/IV NSCLC who had received previous therapy were treated with JNJ-757 (1 × 108 or 1 × 109 colony-forming units [CFUs]) alone (NCT02592967) or JNJ-757 (1 × 109 CFU) plus intravenous nivolumab 240 mg (NCT03371381). Study objectives included the assessment of immunogenicity, safety, and efficacy. Results In the monotherapy study, 18 patients (median age 63.5 y; women 61%) were treated with JNJ-757 (1 × 108 or 1 × 109 CFU) with a median duration of 1.4 months (range: 0–29). The most common adverse events (AEs) were pyrexia (72%) and chills (61%), which were usually mild and resolved within 48 hours. Peripheral proinflammatory cytokines and lymphocyte activation were induced posttreatment with transient mesothelin-specific T-cell responses in 10 of 13 biomarker-evaluable patients. With monotherapy, four of 18 response-evaluable patients had stable disease of 16 or more weeks, including one patient with a reduction in target lesions. In the combination study, 12 patients were enrolled (median age 63.5 y; women 33%). The most common AEs with combination therapy were pyrexia (67%) and chills (58%); six patients had grade 3 AEs or greater, including two cases of treatment-related fatal pneumonitis. The best overall response for the combination was stable disease in four of nine response-evaluable patients. Conclusions As monotherapy, JNJ-757 was immunogenic and tolerable, with mild infusion-related fever and chills. The limited efficacy of JNJ-757, alone or with nivolumab, did not warrant further investigation of the combination.
- Published
- 2020
24. Discovery and Pharmacological Characterization of JNJ-42756493 (Erdafitinib), a Functionally Selective Small-Molecule FGFR Family Inhibitor
- Author
-
Peter King, Suso Platero, Matthew V. Lorenzi, Kelly Van De Ven, Caroline Paulussen, Liang Xie, Jennifer Yang, Jorge Vialard, Christopher Moy, Eleonora Jovcheva, Timothy Perera, David R. Newell, Jayaprakash Karkera, Sylvie Laquerre, Martin Page, Ron Gilissen, David C. Rees, Neil T. Thompson, George Ward, Desiree De Lange, Laurence Anne Mevellec, Patrick Angibaud, Matthew S Squires, Tinne Verhulst, Na Cheng, Eddy Jean Edgard Freyne, Christopher William Murray, and Gordon Saxty
- Subjects
Male ,0301 basic medicine ,Cancer Research ,Angiogenesis ,Antineoplastic Agents ,Biology ,Fibroblast growth factor ,Metastasis ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Erdafitinib ,Cell Line, Tumor ,Quinoxalines ,Drug Discovery ,medicine ,Animals ,Humans ,Molecular Targeted Therapy ,Phosphorylation ,Receptor ,Protein Kinase Inhibitors ,Cell Proliferation ,Dose-Response Relationship, Drug ,Kinase ,medicine.disease ,Receptors, Fibroblast Growth Factor ,Xenograft Model Antitumor Assays ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,Oncology ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,Pyrazoles ,Lysosomes ,Tyrosine kinase ,Protein Binding ,Signal Transduction - Abstract
Fibroblast growth factor (FGF) signaling plays critical roles in key biological processes ranging from embryogenesis to wound healing and has strong links to several hallmarks of cancer. Genetic alterations in FGF receptor (FGFR) family members are associated with increased tumor growth, metastasis, angiogenesis, and decreased survival. JNJ-42756493, erdafitinib, is an orally active small molecule with potent tyrosine kinase inhibitory activity against all four FGFR family members and selectivity versus other highly related kinases. JNJ-42756493 shows rapid uptake into the lysosomal compartment of cells in culture, which is associated with prolonged inhibition of FGFR signaling, possibly due to sustained release of the inhibitor. In xenografts from human tumor cell lines or patient-derived tumor tissue with activating FGFR alterations, JNJ-42756493 administration results in potent and dose-dependent antitumor activity accompanied by pharmacodynamic modulation of phospho-FGFR and phospho-ERK in tumors. The results of the current study provide a strong rationale for the clinical investigation of JNJ-42756493 in patients with tumors harboring FGFR pathway alterations. Mol Cancer Ther; 16(6); 1010–20. ©2017 AACR.
- Published
- 2017
- Full Text
- View/download PDF
25. B03 JNJ-61186372, an Fc Effector Enhanced EGFR/cMet Bispecific Antibody, Induces EGFR/cMet Downmodulation and Efficacy Through Monocyte and Macrophage Trogocytosis
- Author
-
Kristen M. Chevalier, Jocelyn Sendecki, Hillary Millar, Kathryn Packman, Barbara Bushey, Lorraine Lipfert, Sylvie Laquerre, Ryan Lenhart, Marilda Beqiri, Benjamin Henley, Smruthi Vijayaraghavan, Matthew V. Lorenzi, and Sheri Moores
- Subjects
Pulmonary and Respiratory Medicine ,Bispecific antibody ,medicine.anatomical_structure ,Oncology ,Trogocytosis ,business.industry ,Effector ,Monocyte ,medicine ,Cancer research ,Macrophage ,business - Published
- 2020
- Full Text
- View/download PDF
26. Antitumor Activity of Amivantamab (JNJ-61186372), an EGFR-MET Bispecific Antibody, in Diverse Models of
- Author
-
Jiyeon, Yun, Soo-Hwan, Lee, Seok-Young, Kim, Seo-Yoon, Jeong, Jae-Hwan, Kim, Kyoung-Ho, Pyo, Chae-Won, Park, Seong Gu, Heo, Mi Ran, Yun, Sangbin, Lim, Sun Min, Lim, Min Hee, Hong, Hye Ryun, Kim, Meena, Thayu, Joshua C, Curtin, Roland E, Knoblauch, Matthew V, Lorenzi, Amy, Roshak, and Byoung Chul, Cho
- Subjects
Mice, Inbred BALB C ,Lung Neoplasms ,Mice, Nude ,Exons ,Proto-Oncogene Proteins c-met ,ErbB Receptors ,Antineoplastic Agents, Immunological ,Carcinoma, Non-Small-Cell Lung ,Cell Line, Tumor ,Antibodies, Bispecific ,Mutation ,Animals ,Humans ,Female - Published
- 2020
27. Amivantamab (JNJ-61186372), an Fc Enhanced EGFR/cMet Bispecific Antibody, Induces Receptor Downmodulation and Antitumor Activity by Monocyte/Macrophage Trogocytosis
- Author
-
Hillary Millar, Ryan Lenhart, Kathryn Packman, Benjamin Henley, Sylvie Laquerre, Smruthi Vijayaraghavan, Matthew V. Lorenzi, Lorraine Lipfert, Jocelyn Sendecki, Kristen M. Chevalier, Barbara Bushey, Sheri Moores, and Marilda Beqiri
- Subjects
0301 basic medicine ,Cancer Research ,Trogocytosis ,Monocytes ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Downregulation and upregulation ,In vivo ,Antibodies, Bispecific ,medicine ,Humans ,Receptor ,biology ,Chemistry ,Macrophages ,Cancer ,medicine.disease ,030104 developmental biology ,Oncology ,Mechanism of action ,030220 oncology & carcinogenesis ,Cancer cell ,biology.protein ,Cancer research ,Antibody ,medicine.symptom - Abstract
Small molecule inhibitors targeting mutant EGFR are standard of care in non–small cell lung cancer (NSCLC), but acquired resistance invariably develops through mutations in EGFR or through activation of compensatory pathways such as cMet. Amivantamab (JNJ-61186372) is an anti-EGFR and anti-cMet bispecific low fucose antibody with enhanced Fc function designed to treat tumors driven by activated EGFR and/or cMet signaling. Potent in vivo antitumor efficacy is observed upon amivantamab treatment of human tumor xenograft models driven by mutant activated EGFR, and this activity is associated with receptor downregulation. Despite these robust antitumor responses in vivo, limited antiproliferative effects and EGFR/cMet receptor downregulation by amivantamab were observed in vitro. Interestingly, in vitro addition of isolated human immune cells notably enhanced amivantamab-mediated EGFR and cMet downregulation, leading to antibody dose-dependent cancer cell killing. Through a comprehensive assessment of the Fc-mediated effector functions, we demonstrate that monocytes and/or macrophages, through trogocytosis, are necessary and sufficient for Fc interaction-mediated EGFR/cMet downmodulation and are required for in vivo antitumor efficacy. Collectively, our findings represent a novel Fc-dependent macrophage-mediated antitumor mechanism of amivantamab and highlight trogocytosis as an important mechanism of action to exploit in designing new antibody-based cancer therapies.
- Published
- 2020
28. Gene signatures of tumor inflammation and epithelial-to-mesenchymal transition (EMT) predict responses to immune checkpoint blockade in lung cancer with high accuracy
- Author
-
Seth Jeffries, Wei-Ting Hwang, Steven M. Albelda, Matthew V. Lorenzi, Charuhas Deshpande, Vinod Krishna, Raluca Verona, Jeffrey C. Thompson, Christiana Davis, Denis Smirnov, Corey J. Langer, and Yashoda Rajpurohit
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Oncology ,Male ,Cancer Research ,medicine.medical_specialty ,Epithelial-Mesenchymal Transition ,Lung Neoplasms ,medicine.medical_treatment ,Inflammation ,Adenocarcinoma of Lung ,Article ,03 medical and health sciences ,0302 clinical medicine ,Antineoplastic Agents, Immunological ,Internal medicine ,Carcinoma, Non-Small-Cell Lung ,medicine ,Biomarkers, Tumor ,Humans ,Epithelial–mesenchymal transition ,Lung cancer ,Immune Checkpoint Inhibitors ,Retrospective Studies ,business.industry ,Retrospective cohort study ,Immunotherapy ,Middle Aged ,medicine.disease ,Prognosis ,Immune checkpoint ,Blockade ,Gene Expression Regulation, Neoplastic ,Survival Rate ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer biomarkers ,Female ,medicine.symptom ,business ,Follow-Up Studies - Abstract
Objectives Treatment of non-small cell lung cancer (NSCLC) with immune checkpoint blockade (ICB) has resulted in striking clinical responses, but only in a subset of patients. The goal of this study was to evaluate transcriptional signatures previously reported in the literature in an independent cohort of NSCLC patients receiving ICB. Materials and methods This retrospective study analyzed transcriptional profiles from pre-treatment tumor samples of 52 chemotherapy-refractory advanced NSCLC patients treated with anti-PD1/PD-L1 therapy. Gene signatures based on published reports were created and examined for their association with response to therapy and progression-free and overall survival (PFS, OS). Results Two signatures predicting response and outcomes were identified. One reflected the degree of immune infiltration and upregulation of interferon-gamma-induced genes. A second reflected the EMT status. Compared to those not responding to therapy, patients whose tumors responded to ICB had higher scores in an inflammatory gene signature (6.0 ± 2.9 vs -5.5 ± 3.4, p = 0.014) or a more epithelial phenotype (-1.7 ± 1.0 vs 2.1 ± 1.2, p = 0.016). Both signatures demonstrated a satisfactory predictive accuracy for response: AUC of 0.69 (95% CI: 0.54, 0.84) for the inflammatory and 0.70 (95% CI: 0.55, 0.85) for EMT signatures, respectively. A weighted score combining EMT and inflammatory signatures showed increased predictive value with AUC of 0.92 (95% CI: 0.85, 0.99). Kaplan-Meier curves for patients above and below the median combined score showed a significant separation for PFS and OS (all p Conclusions The EMT/Inflammation signature score may be useful in directing checkpoint inhibitor therapy in lung cancer and suggests that reversal of EMT might augment efficacy of ICB.
- Published
- 2019
29. Abstract 953: Efficacy of amivantamab, a bispecific EGFR/MET antibody, correlates with EGFR expression and signaling in NSCLC models with wild-type EGFR
- Author
-
Benjamin Henley, Kristen M. Chevalier, Sylvie Laquerre, Sheri Moores, Matthew V. Lorenzi, Eric B. Haura, Barbara Bushey, Smruthi Vijayaraghaven, Gerald Chu, Matthew Smith, Nataša Obermajer, J.C. Curtin, and Kathryn Packman
- Subjects
Cancer Research ,biology ,business.industry ,Wild type ,Cancer ,Tumor-associated macrophage ,medicine.disease ,Immune system ,Oncology ,Cancer research ,biology.protein ,Medicine ,Immunohistochemistry ,Antibody ,business ,Receptor ,EGFR inhibitors - Abstract
Small molecule EGFR inhibitors have proven effective in treatment of Non-Small Cell Lung Cancer (NSCLC) with activating EGFR mutations, however there is minimal to no efficacy in wild-type EGFR NSCLC. Amivantamab, an EGFR/MET bispecific antibody, has demonstrated clinical activity across a diverse range of EGFR activating mutations in an ongoing Phase I trial and differentiates from anti-EGFR antibodies tested previously in NSCLC clinical trials due to dual targeting and enhanced interaction with Fc receptors on immune cells. As candidate biomarkers to predict response in wt EGFR disease, EGFR and MET expression (immunohistochemistry (IHC)) and signaling (proximity ligation assays (PLA)), as well as tumor associated macrophage (TAM) content, were assessed in 39 NSCLC wt EGFR patient-derived xenograft (PDX) models. A strong correlation (EGFR r=0.63; p Citation Format: Benjamin Henley, Kristen Chevalier, Matthew Smith, Smruthi Vijayaraghaven, Barbara Bushey, Gerald Chu, Joshua Curtin, Nataša Obermajer, Kathryn Packman, Sylvie Laquerre, Matthew Lorenzi, Eric Haura, Sheri Moores. Efficacy of amivantamab, a bispecific EGFR/MET antibody, correlates with EGFR expression and signaling in NSCLC models with wild-type EGFR [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 953.
- Published
- 2021
- Full Text
- View/download PDF
30. JNJ-64041757 (JNJ-757), a Live, Attenuated, Double-Deleted Listeria monocytogenes–Based Immunotherapy in Patients With NSCLC: Results From Two Phase 1 Studies
- Author
-
Roland Elmar Knoblauch, Raffit Hassan, Nicole L. Stone, Ammar Sukari, Enrique Zudaire, Mark Wade, Arun Rajan, Alicia J. Allred, Vali A. Papadimitrakopoulou, Matthew V. Lorenzi, M. Cobo, Ravi Salgia, Melissa Lynne Johnson, Julie R. Brahmer, Gary Mason, Juan Coves Sarto, Nibedita Bandyopadhyay, Mark M. Awad, and Santiago Viteri
- Subjects
Pulmonary and Respiratory Medicine ,medicine.medical_specialty ,Combination therapy ,medicine.medical_treatment ,JNJ-757 ,lcsh:RC254-282 ,Gastroenterology ,Internal medicine ,Non–small cell lung cancer ,Medicine ,Mesothelin ,Adverse effect ,Pneumonitis ,biology ,business.industry ,Immunogenicity ,Immunotherapy ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,Oncology ,LADD Lm ,biology.protein ,Chills ,Nivolumab ,medicine.symptom ,business ,Vaccine - Abstract
Introduction JNJ-64041757 (JNJ-757) is a live, attenuated, double-deleted Listeria monocytogenes–based immunotherapy expressing human mesothelin. JNJ-757 was evaluated in patients with advanced NSCLC as monotherapy (phase 1) and in combination with nivolumab (phase 1b/2). Methods Patients with stage IIIB/IV NSCLC who had received previous therapy were treated with JNJ-757 (1 × 108 or 1 × 109 colony-forming units [CFUs]) alone (NCT02592967) or JNJ-757 (1 × 109 CFU) plus intravenous nivolumab 240 mg (NCT03371381). Study objectives included the assessment of immunogenicity, safety, and efficacy. Results In the monotherapy study, 18 patients (median age 63.5 y; women 61%) were treated with JNJ-757 (1 × 108 or 1 × 109 CFU) with a median duration of 1.4 months (range: 0–29). The most common adverse events (AEs) were pyrexia (72%) and chills (61%), which were usually mild and resolved within 48 hours. Peripheral proinflammatory cytokines and lymphocyte activation were induced posttreatment with transient mesothelin-specific T-cell responses in 10 of 13 biomarker-evaluable patients. With monotherapy, four of 18 response-evaluable patients had stable disease of 16 or more weeks, including one patient with a reduction in target lesions. In the combination study, 12 patients were enrolled (median age 63.5 y; women 33%). The most common AEs with combination therapy were pyrexia (67%) and chills (58%); six patients had grade 3 AEs or greater, including two cases of treatment-related fatal pneumonitis. The best overall response for the combination was stable disease in four of nine response-evaluable patients. Conclusions As monotherapy, JNJ-757 was immunogenic and tolerable, with mild infusion-related fever and chills. The limited efficacy of JNJ-757, alone or with nivolumab, did not warrant further investigation of the combination.
- Published
- 2021
- Full Text
- View/download PDF
31. Abstract DDT02-04: A novel PRMT5 inhibitor with potent in vitro and in vivo activity in preclinical lung cancer models
- Author
-
Dushen Chetty, Matthew V. Lorenzi, Sylvie Laquerre, Gaston Stanislas Marcella Diels, Edward C. Lawson, Carol Yanovich, Italo Poggesi, Ivan Somers, Dana Gaffney, Wim Bert Griet Schepens, Hillary Millar, Weimei Sun, Tongfei Wu, Lijs Beke, Viellevoye Marcel, Marc Du Jardin, Petra Vinken, Erika van Heerde, Tinne Verhulst, James P. Edwards, Dirk Brehmer, Barbara Herkert, Geert Mannens, Vineet Pande, Alain Philippe Poncelet, Christopher Moy, An Boeckx, Marc Parade, Thomas Nys, Jan-Willem Thuring, Lieven Meerpoel, and Joannes T. M. Linders
- Subjects
0301 basic medicine ,Cancer Research ,Methyltransferase ,business.industry ,Protein arginine methyltransferase 5 ,Methylation ,medicine.disease_cause ,medicine.disease ,In vitro ,03 medical and health sciences ,030104 developmental biology ,Oncology ,In vivo ,Cancer research ,medicine ,Cytotoxic T cell ,Carcinogenesis ,business ,Lung cancer - Abstract
PRMT5 is a type II methyltransferase that specifically adds methyl groups to arginine as a long-lasting post-translational modification. The PRMT5/MEP50 complex regulates a plethora of cellular processes, such as epigenetics and splicing, which are notable events involved in tumorigenesis. Although not frequently mutated or amplified in tumors, elevated PRMT5 protein levels in lung and hematologic cancers are correlated with poorer survival. The PRMT5 inhibitor JNJ-64619178 has been selected as a clinical candidate based on its high selectivity and potency (subnanomolar range) under different in vitro and cellular conditions, paired with favorable pharmacokinetics and safety properties. JNJ-64619178 binds into the SAM binding pocket and reaches the substrate binding pocket to inhibit PRMT5/MEP50 function in a time-dependent manner. Broad cell line panel profiling of JNJ-64619178 revealed a wide range of sensitivity, which is indicative of a genomic dependency instead of a general cytotoxic on-target consequence of PRMT5 inhibition. Further investigations indicate a synthetic lethal correlation between PRMT5 inhibition and key cancer driver pathways. JNJ-64619178, dosed orally (10 mg/kg, every day), showed selective and efficient blockage of the methylation of SMD1/3 proteins, which are crucial components of the spliceosome and substrates of PRMT5/MEP50. JNJ-64619178 also demonstrated tumor regression in a biomarker-driven human small cell lung cancer xenograft model (NCI-H1048) and prolonged tumor growth inhibition after dosing cessation. In rodent and nonrodent toxicology studies, a tolerated dose of JNJ-64619178 has been identified, with the observed toxicity consistent with on-target activity. In summary, JNJ-64619178 has a favorable preclinical package that supports clinical testing in patients diagnosed with lung cancer and hematologic malignancies. Citation Format: Dirk Brehmer, Tongfei Wu, Geert Mannens, Lijs Beke, Petra Vinken, Dana Gaffney, Weimei Sun, Vineet Pande, Jan-Willem Thuring, Hillary Millar, Italo Poggesi, Ivan Somers, An Boeckx, Marc Parade, Erika van Heerde, Thomas Nys, Carol Yanovich, Barbara Herkert, Tinne Verhulst, Marc Du Jardin, Lieven Meerpoel, Christopher Moy, Gaston Diels, Marcel Viellevoye, Wim Schepens, Alain Poncelet, Joannes T. Linders, Edward C. Lawson, James P. Edwards, Dushen Chetty, Sylvie Laquerre, Matthew V. Lorenzi. A novel PRMT5 inhibitor with potent in vitro and in vivo activity in preclinical lung cancer models [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 DDT02-04. doi:10.1158/1538-7445.AM2017-DDT02-04
- Published
- 2017
- Full Text
- View/download PDF
32. 537MO First-in-human study of JNJ-64619178, a protein arginine methyltransferase 5 (PRMT5) inhibitor, in patients with advanced cancers
- Author
-
A. Maes, Manish R. Patel, Irene Brana, Judy S. Wang, Anna Spreafico, Hong Xie, Anthony J. Johnson, Pankaj Mistry, Nahor Haddish-Berhane, Matthew V. Lorenzi, Josh Lauring, T. Hernandez, J. Haslam, Jeffrey R. Infante, Jaydeep Mehta, Victor Moreno, Laurie Lenox, Albiruni Ryan Abdul Razak, M. Vieito Villar, and A. Kalota
- Subjects
Oncology ,business.industry ,Protein arginine methyltransferase 5 ,A protein ,Medicine ,In patient ,Hematology ,First in human ,Pharmacology ,Arginine methyltransferase ,business - Published
- 2020
- Full Text
- View/download PDF
33. Abstract 5199: JNJ-61186372, an EGFR-cMet bispecific antibody, in EGFR Exon 20 insertion-driven advanced non-small cell lung cancer (NSCLC)
- Author
-
Matthew V. Lorenzi, Hye Ryun Kim, Meena Thayu, Ji Min Lee, Seo-Yoon Jeong, Jiyeon Yun, Min Hee Hong, Han Na Kang, Jae Hwan Kim, Soo-Hwan Lee, Roland Elmar Knoblauch, Kyoung Ho Pyo, Chae Won Park, J.C. Curtin, Byoung Chul Cho, and Seok-Young Kim
- Subjects
0301 basic medicine ,Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,Cetuximab ,business.industry ,Cancer ,non-small cell lung cancer (NSCLC) ,Cell cycle ,medicine.disease ,EGFR Antibody ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Gefitinib ,Oncology ,030220 oncology & carcinogenesis ,medicine ,Cancer research ,Osimertinib ,business ,medicine.drug - Abstract
PURPOSE: Although EGFR exon20 insertion (ex20ins) mutations account for 4~12 % of EGFR mutant NSCLC patients, there is no effective and selectable anticancer drugs targeting ex20ins mutations so far due to various variant mutations in ex20ins mutations. Moreover, most EGFR ex20ins mutants show primary resistance to EGFR TKIs. JNJ-61186372 (JNJ-372), a bispecific antibody that targets the EGFR and cMet receptors, is currently being explored in a first-in-human study in patients with NSCLC. To better understand the mechanism of JNJ-372 activity in this patient population, we conducted preclinical studies exploring the activity of JNJ-372 in different EGFR ex20ins models. METHODS: To elucidate whether JNJ-372 has antitumor effect in EGFR ex20ins mutants via EGFR and c-MET inhibition, cell viability, western blot, cell cycle, colony formation assay, FACS analysis were performed in JNJ-372 treated BaF3 cells, PDCs, PDOs, and PDX expressing EGFR ex20ins mutation. For mouse tumor models, JNJ-372 was administered i.p. twice a week at 10 mg/kg or 30 mg/kg. Antibody dependent cellular cytotoxicity (ADCC) assay was assessed to figure out whether JNJ-372 had ADCC effects. Referenced patients with ex20ins disease were administered 1050 mg JNJ-372 i.v. weekly for the first 4-week cycle, then biweekly for each subsequent cycle RESULTS: JNJ-372 inhibited the growth of BaF3 cells, PDCs, and a PDO harboring a range of ex20ins, which were resistant to osimertinib and gefitinib. Mechanistic assays revealed the reduction of EGFR and cMet receptor levels and decreases in phospho-EGFR and c-Met, as well as inhibition of their downstream signaling pathways. Cleaved caspase-3 and BIMEL were upregulated at anti-proliferative doses, suggesting caspase-mediated cell death. JNJ-372 demonstrated corresponding antitumor activity in PDC and PDX models harboring different ex20ins; inhibition of signaling and engagement of the apoptotic pathway was confirmed in tumors of JNJ-372-treated mice. In PDCs with EGFR ex20ins mutation, we verified that JNJ-372 had a significant ADCC effect compared to EGFR antibody drug cetuximab. In the first-in-human trial, CT scans from two patients treated with JNJ-372 revealed reductions in tumor burden. A 58-year patient harboring H773delinsNPY showed -63% tumor reduction with progression-free survival of > 20 months and a 48-year patient harboring S768_D770dup showed -38.9% tumor reduction. CONCLUSION: JNJ-372 drives antitumor activity in preclinical models of EGFR ex20ins, which have no therapeutic options in clinic, by decreasing EGFR and cMet receptor levels, inhibiting downstream signaling cascades, activating apoptotic signaling as well as ADCC. These results provide a promising therapeutic option to patients with EGFR ex20ins mutations and an understanding of the activity of JNJ-372 being observed in the first-in-human study. Citation Format: Jiyeon Yun, Han Na Kang, Soo-Hwan Lee, Seo-Yoon Jeong, Chae Won Park, Jae-Hwan Kim, Kyoung-Ho Pyo, Ji Min Lee, Seok-Young Kim, Min Hee Hong, Hye Ryun Kim, Meena Thayu, Joshua Curtin, Roland Knoblauch, Matthew Lorenzi, Byoung Chul Cho. JNJ-61186372, an EGFR-cMet bispecific antibody, in EGFR Exon 20 insertion-driven advanced non-small cell lung cancer (NSCLC) [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5199.
- Published
- 2020
- Full Text
- View/download PDF
34. Abstract 5651: JNJ-61186372, an Fc enhanced EGFR/cMet bispecific antibody, mediates EGFR and cMet downmodulation and therapeutic efficacy preclinically through monocyte / macrophage mediated trogocytosis
- Author
-
Sheri Moores, Barbara Bushey, Benjamin Henley, Kathryn Packman, Matthew V. Lorenzi, Hillary Millar, Sylvie Laquerre, Smruthi Vijayaraghavan, Marilda Beqiri, Lorraine Lipfert, Kristen M. Chevalier, and Ryan Lenhart
- Subjects
Cancer Research ,Trogocytosis ,business.industry ,Monocyte ,Cell ,Cancer ,medicine.disease ,T790M ,medicine.anatomical_structure ,Immune system ,Oncology ,Downregulation and upregulation ,medicine ,Cancer research ,business ,Receptor - Abstract
Small molecule inhibitors targeting EGFR are now standard of care in NSCLC patients harboring EGFR mutations, but acquired resistance invariably develops through secondary mutations within EGFR and/or through activation of compensatory pathways such as cMet. JNJ-61186372 (JNJ-372) is an anti-EGFR and cMet bispecific antibody with enhanced binding to immune cell Fcγ receptors, designed to target tumors with activated EGFR and cMet signaling through a distinct mechanism of action. Ongoing first-in-human study in patients with advanced, treatment refractory EGFR mutant NSCLC revealed JNJ-372 to have clinical activity in patients with diverse EGFR-mutated NSCLC, including Exon 20 mutations, TKI resistance mutations (T790M, C797S), and resistance due to MET amplification. However preclinically, despite potent anti-tumor activity in NSCLC xenograft models, only modest anti-proliferative effects were observed with JNJ-372 in cell lines in vitro. Interestingly, the addition of isolated human immune cells (PBMCs) to the in vitro assays enhanced JNJ-372-mediated EGFR and cMet downregulation, and dose-dependent tumor cell killing. Through depletion or enrichment of individual immune cell types, we demonstrated that monocytes and/or macrophages are necessary for JNJ-372 Fc interaction-mediated EGFR/cMet downmodulation. Depletion of macrophages in mice showed that they are required for JNJ-372 anti-tumor efficacy. Finally, we showed that the down-modulation of EGFR and cMet receptors occurs through monocyte or macrophage-mediated trogocytosis. Collectively, these results demonstrate a novel Fc-dependent mechanism of action for JNJ-372 and support its continued clinical development in patients with aberrant EGFR and cMet signaling. Citation Format: Smruthi Vijayaraghavan, Lorraine Lipfert, Barbara Bushey, Kristen Chevalier, Benjamin Henley, Ryan Lenhart, Marilda Beqiri, Hillary J. Millar, Kathryn Packman, Matthew V. Lorenzi, Sylvie Laquerre, Sheri Moores. JNJ-61186372, an Fc enhanced EGFR/cMet bispecific antibody, mediates EGFR and cMet downmodulation and therapeutic efficacy preclinically through monocyte / macrophage mediated trogocytosis [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5651.
- Published
- 2020
- Full Text
- View/download PDF
35. Erratum: Drug Discovery Approaches to Target Wnt Signaling in Cancer Stem Cells
- Author
-
Joshua C. Curtin and Matthew V. Lorenzi
- Subjects
0301 basic medicine ,Drug discovery ,business.industry ,Wnt signaling pathway ,Antineoplastic Agents ,Biology ,Wnt Proteins ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Text mining ,Oncology ,Cancer stem cell ,030220 oncology & carcinogenesis ,Neoplasms ,Drug Discovery ,Cancer research ,Neoplastic Stem Cells ,Animals ,Humans ,Molecular Targeted Therapy ,Erratum ,business ,Signal Transduction - Abstract
Cancer stem cells (CSCs) represent a unique subset of cells within a tumor that possess self-renewal capacity and pluripotency, and can drive tumor initiation and maintenance. First identified in hematological malignancies, CSCs are now thought to play an important role in a wide variety of solid tumors such as NSCLC, breast and colorectal cancer. The role of CSCs in driving tumor formation illustrates the dysregulation of differentiation in tumorigenesis. The Wnt, Notch and Hedgehog (HH) pathways are developmental pathways that are commonly activated in many types of cancer. While substantial progress has been made in developing therapeutics targeting Notch and HH, the Wnt pathway has remained an elusive therapeutic target. This review will focus on the clinical relevance of the Wnt pathway in CSCs and tumor cell biology, as well as points of therapeutic intervention and recent advances in targeting Wnt/β-catenin signaling.
- Published
- 2018
36. P1.01-94 JNJ-61186372, an EGFR-cMet Bispecific Antibody, in EGFR Exon 20 Insertion-Driven Advanced NSCLC
- Author
-
Han Na Kang, Minsun Hong, A. Roshak, Seo-Yoon Jeong, J.C. Curtin, Jiyeon Yun, Roland Elmar Knoblauch, S.J. Lee, Matthew V. Lorenzi, Meena Thayu, H.R. Kim, Chae Won Park, and Byoung Chul Cho
- Subjects
Pulmonary and Respiratory Medicine ,Exon ,Bispecific antibody ,Oncology ,business.industry ,Cancer research ,Medicine ,business - Published
- 2019
- Full Text
- View/download PDF
37. Structure-Based Design of Selective Janus Kinase 2 Imidazo[4,5-d]pyrrolo[2,3-b]pyridine Inhibitors
- Author
-
Hart Amy C, Javed Khan, Grebinski James W, James Kempson, Jonathan Lippy, Gretchen M. Schroeder, Theresa M. McDevitt, Ashok V. Purandare, Ragini Vuppugalla, John S. Tokarski, George L. Trainor, Junqing Guo, Jennifer Inghrim, Matthew V. Lorenzi, Dan You, Honghe Wan, John S. Sack, William J. Pitts, Yueping Zhang, and Louis J. Lombardo
- Subjects
Janus kinase 2 ,biology ,Kinase ,Organic Chemistry ,Hit to lead ,Biochemistry ,Combinatorial chemistry ,chemistry.chemical_compound ,chemistry ,Drug Discovery ,Pyridine ,biology.protein ,Side chain ,Structure based ,Selectivity ,Thiazole - Abstract
Early hit to lead work on a pyrrolopyridine chemotype provided access to compounds with biochemical and cellular potency against Janus kinase 2 (JAK2). Structure-based drug design along the extended hinge region of JAK2 led to the identification of an important H-bond interaction with the side chain of Tyr 931, which improved JAK family selectivity. The 4,5-dimethyl thiazole analogue 18 demonstrated high levels of JAK family selectivity and was identified as a promising lead for the program.
- Published
- 2015
- Full Text
- View/download PDF
38. Selective JAK2/ABL dual inhibition therapy effectively eliminates TKI-insensitive CML stem/progenitor cells
- Author
-
Katharina Rothe, Matthew V. Lorenzi, Xiaoyan Jiang, Adrian Woolfson, Min Chen, and Hanyang Lin
- Subjects
Time Factors ,CD34 ,Dasatinib ,Antigens, CD34 ,Apoptosis ,Mice, SCID ,leukemic stem cells ,Tyrosine-kinase inhibitor ,Piperazines ,Mice, Inbred NOD ,hemic and lymphatic diseases ,Antineoplastic Combined Chemotherapy Protocols ,STAT5 Transcription Factor ,Tumor Cells, Cultured ,Molecular Targeted Therapy ,Proto-Oncogene Proteins c-abl ,CML ,BCR-ABL ,Nuclear Proteins ,3. Good health ,Leukemia ,Haematopoiesis ,Oncology ,JAK2 ,Benzamides ,Imatinib Mesylate ,Neoplastic Stem Cells ,Stem cell ,Heterocyclic Compounds, 3-Ring ,medicine.drug ,Research Paper ,Signal Transduction ,medicine.drug_class ,Biology ,BMS-911543 ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,medicine ,Animals ,Humans ,Progenitor cell ,Protein Kinase Inhibitors ,Adaptor Proteins, Signal Transducing ,Cell Proliferation ,TKI resistance ,Janus Kinase 2 ,medicine.disease ,Xenograft Model Antitumor Assays ,respiratory tract diseases ,Thiazoles ,Imatinib mesylate ,Pyrimidines ,Drug Resistance, Neoplasm ,Cancer research ,K562 Cells - Abstract
Imatinib Mesylate (IM) and other tyrosine kinase inhibitor (TKI) therapies have had a major impact on the treatment of chronic myeloid leukemia (CML). However, TKI monotherapy is not curative, with relapse and persistence of leukemic stem cells (LSCs) remaining a challenge. We have recently identified an AHI-1-BCR-ABL-JAK2 protein complex that contributes to the transforming activity of BCR-ABL and IM-resistance in CML stem/progenitor cells. JAK2 thus emerges as an attractive target for improved therapies, but off-target effects of newly developed JAK2 inhibitors on normal hematopoietic cells remain a concern. We have examined the biological effects of a highly selective, orally bioavailable JAK2 inhibitor, BMS-911543, in combination with TKIs on CD34+ treatment-naive IM-nonresponder cells. Combination therapy reduces JAK2/STAT5 and CRKL activities, induces apoptosis, inhibits proliferation and colony growth, and eliminates CML LSCs in vitro. Importantly, BMS-911543 selectively targets CML stem/progenitor cells while sparing healthy stem/progenitor cells. Oral BMS-911543 combined with the potent TKI dasatinib more effectively eliminates infiltrated leukemic cells in hematopoietic tissues than TKI monotherapy and enhances survival of leukemic mice. Dual targeting BCR-ABL and JAK2 activities in CML stem/progenitor cells may consequently lead to more effective disease eradication, especially in patients at high risk of TKI resistance and disease progression.
- Published
- 2014
39. OA10.06 A First-in-Human Phase 1 Trial of the EGFR-cMET Bispecific Antibody JNJ-61186372 in Patients with Advanced Non-Small Cell Lung Cancer (NSCLC)
- Author
-
Matthew V. Lorenzi, M-J. Ahn, H.R. Kim, S.J. Lee, Roland Elmar Knoblauch, Z. Aguilar, Sylvie Laquerre, Byoung Chul Cho, S. Triantos, Nahor Haddish-Berhane, K. Park, Minsun Hong, M. Curtis, Dawn Millington, and S. Chaplan
- Subjects
0301 basic medicine ,Pulmonary and Respiratory Medicine ,Bispecific antibody ,business.industry ,non-small cell lung cancer (NSCLC) ,First in human ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Cancer research ,Medicine ,In patient ,business - Published
- 2018
- Full Text
- View/download PDF
40. Abstract 4818: Fc-mediated mechanism of action for the novel EGFR-cMET bispecific antibody (JNJ-61186372) in non-small cell lung cancer
- Author
-
Smruthi Vijayaraghavan, Barbara Bushey, Lorriane Lipfert, Rupesh Nanjunda, Eilyn R. Lacy, Peter Buckley, Sylvie Laquerre, Matthew V. Lorenzi, and Sheri Moores
- Subjects
Cancer Research ,Oncology - Abstract
JNJ-61186372 (JNJ-372) is an anti-EGFR and cMet bispecific antibody with an active Fc backbone (IgG1) designed to treat non-small cell lung cancer (NSCLC) disease. A first-in-human study is currently being conducted to assess the safety and preliminary efficacy of JNJ-372 in patients with advanced NSCLC. Early data suggests that JNJ-372 can induce partial responses in subjects with diverse populations of EGFR-mutated NSCLC, including Exon 20ins as well as TKI resistance mutations. Our previous pre-clinical in vivo studies showed that the Fc inactive version (IgG2sigma) of the EGFR/cMet antibody was significantly impaired in its ability to inhibit tumor growth compared to the Fc active JNJ-372. The IgG2sigma variant also reduced the ability of the bispecific antibody to mediate downregulation of EGFR, cMet and downstream signaling components. This suggested that the interaction of the Fc arm with the Fc receptors on the innate immune cells play a crucial role in the mechanism of action of JNJ-372. While JNJ-372 has demonstrated antibody dependent cellular cytotoxicity (ADCC) and antibody dependent cellular phagocytosis (ADCP) in vitro, the potential role of Fc interactions in downregulation of EGFR and cMet was not well understood. We explored the different Fc-mediated immune effector functions of JNJ-372 and interrogated how they contribute to EGFR and cMet downregulation and overall efficacy. In NSCLC cell lines, JNJ-372 induced Fc-mediated dose-dependent ADCC and ADCP but not complement-dependent cytotoxicity. Further, the presence of isolated human immune cells (PBMC) significantly enhanced JNJ-372 mediated EGFR and cMet downregulation and dose-dependent tumor cell killing. Studies are in progress to better understand which immune cells and which Fc receptor interactions are essential for the drug efficacy through immune cell depletion and FcR blocking studies; such studies may help guide clinical biomarker development. This work elucidates a novel Fc-dependent mechanism of action for the EGFR-cMet bispecific antibody. Citation Format: Smruthi Vijayaraghavan, Barbara Bushey, Lorriane Lipfert, Rupesh Nanjunda, Eilyn R. Lacy, Peter Buckley, Sylvie Laquerre, Matthew V. Lorenzi, Sheri Moores. Fc-mediated mechanism of action for the novel EGFR-cMET bispecific antibody (JNJ-61186372) in non-small cell lung cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4818.
- Published
- 2019
- Full Text
- View/download PDF
41. JNJ-61186372 (JNJ-372), an EGFR-cMet bispecific antibody, in EGFR-driven advanced non-small cell lung cancer (NSCLC)
- Author
-
Jong Seok Lee, Ji-Youn Han, Nahor Haddish-Berhane, Eun Kyung Cho, Roland Elmar Knoblauch, Karen L. Reckamp, Joshua K. Sabari, Rachel E. Sanborn, Matthew V. Lorenzi, Keunchil Park, Laurie Sherman, Ki Hyeong Lee, Byoung Chul Cho, Kyounghwa Bae, Eric B. Haura, Joshua Bauml, Meena Thayu, Sang-We Kim, Ramaswamy Govindan, and J.C. Curtin
- Subjects
Cancer Research ,Bispecific antibody ,business.industry ,non-small cell lung cancer (NSCLC) ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,030220 oncology & carcinogenesis ,Block (telecommunications) ,Cancer research ,Medicine ,Cell-mediated cytotoxicity ,business ,Receptor degradation ,030215 immunology - Abstract
9009 Background: JNJ-372 binds EGFR and cMet to block ligand binding, promote receptor degradation, and trigger antibody-dependent cellular cytotoxicity in models of EGFR-mutated (EGFRm) NSCLC. Here we describe the ongoing phase 1 safety, pharmacokinetics (PK), and activity of JNJ-372 in patients (pts) with NSCLC, including 3rd generation tyrosine kinase inhibitor (3GTKI)-relapsed EGFRm NSCLC and EGFR Exon20ins disease. Methods: Pts received JNJ-372 (140–1400 mg) IV weekly for the first 28-day cycle and biweekly thereafter. 1050–1400 mg doses are being explored in dose expansion. Blood samples were collected for PK analyses. Efficacy by investigator per RECIST v1.1 in pts with EGFRm NSCLC treated at ≥700 mg is presented. Tumors were characterized by next-generation sequencing of circulating tumor (ct)DNA and/or tumor tissue. Results: As of 17 Jan 2019, 116 enrolled pts with NSCLC were treated. Median age was 63 years, 38% were male, 77% were Asian, and 97% had EGFR mutations. Mean duration of treatment was 3.8 months, longest exposure was 20 cycles. The PK data set included pts from Korea (77%) and the US (23%). At the 1050 mg dose, 72% of pts achieved average concentrations above the EC90 based on preclinical models. Adverse events (AEs; ≥20%) were rash (59%), infusion related reaction (58%), paronychia (28%), and constipation (22%). Additional EGFR/cMet-related AEs include stomatitis (17%), pruritis (15%), peripheral edema (11%), and diarrhea (7%). Grade ≥3 AEs were reported in 34% (8% treatment-related) with dyspnea (6%) and pneumonia (3%) most frequently observed. Among response-evaluable pts, 25/88 (28%) achieved best timepoint response of partial response (PR). 10/47 pts with prior 3GTKI therapy had best timepoint response of PR (6 confirmed), including 4 with C797S, 1 with cMet amplification, and 5 without identifiable EGFR/cMet-dependent resistance. 6/20 pts with Exon20ins had best timepoint response of PR (3 confirmed). Conclusions: JNJ-372 has a manageable safety profile consistent with EGFR and cMet inhibition. Preliminary responses were achieved in 3GTKI-relapsed disease, including C797S and cMet amplification, and Exon20ins disease; enrollment in dose expansion is ongoing. Clinical trial information: NCT02609776.
- Published
- 2019
- Full Text
- View/download PDF
42. Preliminary immunogenicity, safety, and efficacy of JNJ-64041757 (JNJ-757) in non-small cell lung cancer (NSCLC): Results from two phase 1 studies
- Author
-
Roland Elmar Knoblauch, J. Coves, Julie R. Brahmer, Santiago Viteri Ramirez, Alicia J. Allred, Mark Wade, Melissa Lynne Johnson, Nicole L. Stone, Enrique Zudaire, Gary Mason, Vassiliki A. Papadimitrakopoulou, Raffit Hassan, Matthew V. Lorenzi, Mark M. Awad, Manuel Cobo Dols, Ammar Sukari, Arun Rajan, and Ravi Salgia
- Subjects
Cancer Research ,biology ,business.industry ,medicine.medical_treatment ,Immunogenicity ,non-small cell lung cancer (NSCLC) ,Immunotherapy ,medicine.disease ,medicine.disease_cause ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Listeria monocytogenes ,030220 oncology & carcinogenesis ,medicine ,biology.protein ,Cancer research ,Mesothelin ,business ,030215 immunology - Abstract
9093 Background: The immunogenicity, safety, and efficacy of JNJ-757, a live attenuated, double-deleted Listeria monocytogenes-based immunotherapy expressing human mesothelin (MSLN), were evaluated in patients (pts) with advanced NSCLC (adenocarcinoma) as monotherapy (phase 1) and in combination with nivolumab (phase 1b/2). Methods: Adult pts with Stage IIIB/IV NSCLC who had received prior systemic therapy (including 1 platinum-based chemotherapy, prior PD-1/PD-L1 therapy allowed) were included. Dose-limiting toxicities, adverse events (AEs), tumor response, T cell response, and JNJ-757 bacterial shedding profile were evaluated in pts treated with JNJ-757 (108 or 109 colony forming units [CFU]) alone or JNJ-757 (109 CFU)+nivolumab 240 mg combination therapy until progression. Results: In the monotherapy trial, 18 pts (median age 63.5 years; women 61%) were treated with JNJ-757 108 or 109 CFU with a median duration of 1.4 months (range 0-29). Most common AEs were pyrexia (72%) and chills (61%), which were usually mild and resolved within 48 hours, suggesting transient activation of an innate immune response to JNJ-757. Treatment-related grade ≥3 AEs were infrequent (4 [22%]). Induction of peripheral proinflammatory cytokines and lymphocyte activation was observed post-treatment with transient MSLN-specific T cell responses in 10/13 evaluable pts, consistent with the mechanism of action of JNJ-757. With monotherapy, 4/18 response-evaluable pts had stable disease (SD) ≥16 weeks, including 1 pt with a 53% reduction in target lesions. In the combination therapy study, 12 pts were enrolled (median age 63.5 years; women 33%). The most common AEs were pyrexia (67%) and chills (58%); 6 pts had grade ≥3 AEs including 2 cases of treatment-related fatal pneumonitis. Best overall response for the combination was SD in 4/9 evaluable pts. JNJ-757 in combination with nivolumab suggested increased risk of pneumonitis. Conclusions: As monotherapy, JNJ-757 was tolerable with mild infusion-related fever and chills supporting the initiation of the combination therapy study. However, the risk-benefit profile of JNJ-757+nivolumab, did not support proceeding to phase 2. Clinical trial information: NCT02592967, NCT03371381.
- Published
- 2019
- Full Text
- View/download PDF
43. Analysis of FGFR alterations from circulating tumor DNA (ctDNA) and Tissue in a phase II trial of erdafitinib in urothelial carcinoma (UC)
- Author
-
Ian McCaffery, Peter De Porre, Christopher Moy, Anjali Narayan Avadhani, Matthew V. Lorenzi, Arlene O. Siefker-Radtke, Clifford Motley, Yohann Loriot, Yauheniya Cherkas, Ademi E. Santiago-Walker, and Anne OHagan
- Subjects
musculoskeletal diseases ,Cancer Research ,animal structures ,Plasma samples ,business.industry ,Phases of clinical research ,03 medical and health sciences ,0302 clinical medicine ,Oncology ,Erdafitinib ,Circulating tumor DNA ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,Phase (matter) ,embryonic structures ,Cancer research ,Medicine ,biological phenomena, cell phenomena, and immunity ,business ,030215 immunology ,Urothelial carcinoma - Abstract
420 Background: Plasma samples from a Phase 2 study of the pan-FGFR inhibitor erdafitinib in advanced UC patients (pts) with FGFR mutations (mut) or fusions, were tested using next generation sequencing (NGS) for circulating tumor DNA (ctDNA), and results compared to central FGFR status determination from tissue. Methods: FGFR alterations were measured in archival tissue using an RNA-based RT-PCR test and compared with FGFR alterations identified in pre-treatment plasma specimens using the Guardant360 ctDNA test. Sensitivity of the ctDNA test to detect the FGFR alterations identified by RT-PCR from tissue, the proportion of pts with a study-eligible FGFR alteration in ctDNA, and the ability to detect a tissue FGFR alteration in ctDNA in relation to clinical response were assessed. Results: Samples from 155 pts with detectable baseline ctDNA were included. Overall, concordance between ctDNA and tissue-based FGFR results was 56% (87/155): 63% (77/122) for tissue mut-positive pts vs 24% (7/29) in fusion-positive pts. 61% of pts (94/155) were positive for a study-eligible FGFR alteration from blood-based testing in the tissue-positive population. The response rate was 47% (36/77) for patients for which FGFR mutations could be detected in blood (ctDNA- FGFR-positive) compared with 32% (14/44) in patients for which mutations were not detected in blood (ctDNA- FGFR-negative), with an estimated odds ratio of 1.882 (95% CI: 0.866; 4.090) Conclusions: The 63% concordance rate for detecting FGFR mut in temporally unmatched blood and tissue supports potential for patient selection with blood-based testing, while ctDNA FGFR fusion detection may require further optimization. Although differences in clinical response rate were observed between ctDNA- FGFR-positive and negative patients, the results were not statistically significant. Importantly, a proportion of ctDNA-negative pts responded to erdafitinib, indicating that tissue-based testing may be warranted for pts negative for FGFR alterations via blood-based testing. (BLC2001, NCT02365597). Clinical trial information: NCT02365597.
- Published
- 2019
- Full Text
- View/download PDF
44. Oncogenic Characterization and Pharmacologic Sensitivity of Activating Fibroblast Growth Factor Receptor (FGFR) Genetic Alterations to the Selective FGFR Inhibitor Erdafitinib
- Author
-
Gabriela Martinez Cardona, John Alvarez, Matthew V. Lorenzi, Dana Gaffney, Jayaprakash Karkera, Suso Platero, Michael Sharp, Katherine Bell, Feng R. Luo, Joseph Portale, Rastislav Bahleda, Ademi E. Santiago-Walker, Christopher Moy, and Peter King
- Subjects
musculoskeletal diseases ,0301 basic medicine ,Male ,Cancer Research ,animal structures ,Oncogene Proteins, Fusion ,FGFR Inhibition ,Biology ,03 medical and health sciences ,Rats, Nude ,0302 clinical medicine ,Erdafitinib ,In vivo ,Quinoxalines ,medicine ,Biomarkers, Tumor ,Animals ,Receptor ,Melanoma ,Cancer ,Oncogenes ,medicine.disease ,Molecular biology ,Receptors, Fibroblast Growth Factor ,030104 developmental biology ,Oncology ,Urinary Bladder Neoplasms ,Fibroblast growth factor receptor ,030220 oncology & carcinogenesis ,embryonic structures ,Pyrazoles ,biological phenomena, cell phenomena, and immunity ,Signal transduction ,Tomography, X-Ray Computed - Abstract
Fibroblast growth factor receptor (FGFR) genetic alterations are frequently observed in cancer, suggesting that FGFR inhibition may be a promising therapy in patients harboring these lesions. Identification of predictive and pharmacodynamic biomarkers to select and monitor patients most likely to respond to FGFR inhibition will be the key to clinical development of this class of agents. Sensitivity to FGFR inhibition and correlation with FGFR pathway activation status were determined in molecularly annotated panels of cancer cell lines and xenograft models. Pathway inhibition in response to FGFR inhibitor treatment was assessed in cell lines (both in vitro and in vivo) and in samples from patients treated with the FGFR inhibitor JNJ-42756493 (erdafitinib). Frequency of FGFR aberrations was assessed in a panel of NSCLC, breast, prostate, ovarian, colorectal, and melanoma human tumor tissue samples. FGFR translocations and gene amplifications present in clinical specimens were shown to display potent transforming activity associated with constitutive pathway activation. Tumor cells expressing these FGFR activating mutants displayed sensitivity to the selective FGFR inhibitor erdafitinib and resulted in suppression of FGFR phosphorylation and downstream signal transduction. Clinically, patients receiving erdafitinib showed decreased Erk phosphorylation in tumor biopsies and elevation of serum phosphate. In a phase I study, a heavily pretreated bladder cancer patient with an FGFR3–TACC3 translocation experienced a partial response when treated with erdafitinib. This preclinical study confirmed pharmacodynamics and identified new predictive biomarkers to FGFR inhibition with erdafitinib and supports further clinical evaluation of this compound in patients with FGFR genetic alterations. Mol Cancer Ther; 16(8); 1717–26. ©2017 AACR.
- Published
- 2016
45. Characterization of BMS-911543, a functionally selective small-molecule inhibitor of JAK2
- Author
-
Matthew V. Lorenzi, Xia Han, Honghe Wan, Kim W. McIntyre, Yifan Zhang, E Michaud, A Tefferi, Stuart Emanuel, Ragini Vuppugalla, Becky Penhallow, George L. Trainor, Y Blat, Animesh Pardanani, Tracy L. Taylor, Elizabeth Fitzpatrick, Petra Ross-Macdonald, Frank Lee, Louis J. Lombardo, Theresa M. McDevitt, Ashok V. Purandare, Marco M. Gottardis, Adrian Woolfson, H de Silva, Terra L. Lasho, Dan You, Stefan Ruepp, Jennifer Hosbach, Donna L. Pedicord, and C Mulligan
- Subjects
Cancer Research ,medicine.drug_class ,Blotting, Western ,Antineoplastic Agents ,Pharmacology ,Biology ,Tyrosine-kinase inhibitor ,In vivo ,medicine ,Humans ,STAT1 ,Progenitor cell ,Protein Kinase Inhibitors ,Cell Proliferation ,Myeloproliferative Disorders ,Gene Expression Profiling ,Hematology ,Janus Kinase 2 ,Small molecule ,In vitro ,Gene expression profiling ,Oncology ,biology.protein ,Cancer research ,Janus kinase ,Heterocyclic Compounds, 3-Ring - Abstract
We report the characterization of BMS-911543, a potent and selective small-molecule inhibitor of the Janus kinase (JAK) family member, JAK2. Functionally, BMS-911543 displayed potent anti-proliferative and pharmacodynamic (PD) effects in cell lines dependent upon JAK2 signaling, and had little activity in cell types dependent upon other pathways, such as JAK1 and JAK3. BMS-911543 also displayed anti-proliferative responses in colony growth assays using primary progenitor cells isolated from patients with JAK2(V617F)-positive myeloproliferative neoplasms (MPNs). Similar to these in vitro observations, BMS-911543 was also highly active in in vivo models of JAK2 signaling, with sustained pathway suppression being observed after a single oral dose. At low dose levels active in JAK2-dependent PD models, no effects were observed in an in vivo model of immunosuppression monitoring antigen-induced IgG and IgM production. Expression profiling of JAK2(V617F)-expressing cells treated with diverse JAK2 inhibitors revealed a shared set of transcriptional changes underlying pharmacological effects of JAK2 inhibition, including many STAT1-regulated genes and STAT1 itself. Collectively, our results highlight BMS-911543 as a functionally selective JAK2 inhibitor and support the therapeutic rationale for its further characterization in patients with MPN or in other disorders characterized by constitutively active JAK2 signaling.
- Published
- 2011
- Full Text
- View/download PDF
46. Abstract DDT01-03: Discovery and preclinical pharmacology of JNJ-61186372: A novel bispecific antibody targeting EGFR and cMET
- Author
-
Matthew V. Lorenzi, Janine Schuurman, Paul W. H. I. Parren, Peter Haytko, Ricardo M. Attar, Mark L. Chiu, Sheri Moores, Kristen M. Chevalier, Robert A. Kramer, Barbara Bushey, Joost J. Neijssen, and Mark R. Anderson
- Subjects
Antibody-dependent cell-mediated cytotoxicity ,Cancer Research ,business.industry ,Cancer ,Pharmacology ,medicine.disease ,Gefitinib ,Oncology ,Mechanism of action ,medicine ,Phosphorylation ,Erlotinib ,medicine.symptom ,Receptor ,business ,medicine.drug ,EGFR inhibitors - Abstract
NSCLC with activating mutations in the EGFR gene are associated with high response rates to EGFR tyrosine kinase inhibitors (TKIs), such as erlotinib and gefitinib, but invariably acquired resistance emerges over time. A primary cause of resistance is the emergence of secondary mutations in EGFR which neutralize the effectiveness of TKIs. In addition, the cMet pathway is often activated, either through MET gene amplification, overexpression of cMet protein, or an increase in the ligand HGF, to provide a compensatory survival pathway conferring resistance to EGFR TKIs. We have designed a bispecific EGFR-cMet antibody (JNJ-61186372) with a unique set of mechanisms of action resulting in anti-tumor activity in the EGFR mutant setting, with or without cMet pathway activation. We have demonstrated three mechanisms of action that contribute to the activity of JNJ-61186372: 1) inhibition of ligand-induced phosphorylation of both EGFR and cMet, 2) receptor degradation in vivo, and 3) enhanced ADCC activity. JNJ-61186372 inhibited EGF-induced phosphorylation of EGFR in cell lines with either wild-type (WT) EGFR or activating mutations in EGFR. In the same cell lines, JNJ-61186372 inhibited HGF-induced phosphorylation of cMet. JNJ-61186372 also blocked pERK and pAkt with similar IC50 values in EGFR-WT and EGFR mutant cell lines, indicating that downstream signaling pathways were inhibited. Total protein levels of both EGFR and cMet were decreased in xenograft tumor models following treatment with JNJ-61186372 compared to tumors from mice treated with PBS control suggesting that one mechanism by which JNJ-61186372 suppresses EGFR and cMet activity in vivo is through degradation of both receptors. The third mechanism of action is directing immune cells to kill tumor cells. JNJ-61186372 is produced with low levels of fucosylation, which translates to an enhanced antibody-dependent cellular cytotoxicity (ADCC). These three mechanisms of action of JNJ-61186372 provide a distinct preclinical profile for targeting both EGFR and cMET in a single bispecific antibody. JNJ-61186372 demonstrated efficacy in multiple in vivo tumor models with EGFR mutations, including both cell line and patient-derived xenografts. Importantly, JNJ-61186372 effectively inhibited tumor growth in models with mutant EGFR and cMet activation, whereas single agent EGFR inhibitors were less effective. The preclinical data support the clinical development of JNJ-61186372 in patients with lung cancer and other malignancies associated with aberrant EGFR and cMET signaling. Citation Format: Sheri L. Moores, Mark Chiu, Barbara Bushey, Kristen Chevalier, Peter Haytko, Joost Neijssen, Paul Parren, Janine Schuurman, Mark Anderson, Ricardo Attar, Robert Kramer, Matthew V. Lorenzi. Discovery and preclinical pharmacology of JNJ-61186372: A novel bispecific antibody targeting EGFR and cMET. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr DDT01-03. doi:10.1158/1538-7445.AM2014-DDT01-03
- Published
- 2014
- Full Text
- View/download PDF
47. JNJ-61186372 (JNJ-372), an EGFR-cMET bispecific antibody, in advanced non-small cell lung cancer (NSCLC): An update on phase I results
- Author
-
E. Attiyeh, Matthew V. Lorenzi, Kyounghwa Bae, Laurie Sherman, Rachel E. Sanborn, K.H. Lee, Jung-Gon Lee, K. Park, Joshua Bauml, Roland Elmar Knoblauch, Nahor Haddish-Berhane, M. Curtis, Byoung Chul Cho, E.K. Cho, E.B. Haura, and J-Y. Han
- Subjects
0301 basic medicine ,03 medical and health sciences ,Bispecific antibody ,030104 developmental biology ,Oncology ,business.industry ,Cancer research ,Medicine ,non-small cell lung cancer (NSCLC) ,Hematology ,business ,medicine.disease - Published
- 2018
- Full Text
- View/download PDF
48. Abstract 4859: JNJ-64619178, a selective and pseudo-irreversible PRMT5 inhibitor with potent in vitro and in vivo activity, demonstrated in several lung cancer models
- Author
-
Ivan Sommers, Matthew V. Lorenzi, Dirk Brehmer, Mark Salvati, Dana Gaffney, Geert Mannens, Vineet Pande, Sylvie Laquerre, Wu Tongfei, Junguo Zhou, Lijs Beke, Petra Vinken, Christopher Moy, Jan-Willem Thuring, Hillary Millar, Weimei Sun, and Nahor Haddish-Berhane
- Subjects
0301 basic medicine ,Cancer Research ,Methyltransferase ,Cell growth ,Chemistry ,Protein arginine methyltransferase 5 ,Alternative splicing ,Cancer ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,Oncology ,In vivo ,Cancer research ,medicine ,Lung cancer ,Methylosome - Abstract
PRMT5 is a type II methyltransferase that symmetrically di-methylates arginine residues on proteins involved in signal transduction and cellular transcription. For example, PRMT5 acts as the enzymatic machinery of the methylosome complex, crucial for spliceosome assembly and activity. Although not frequently mutated or amplified in tumors, an elevated PRMT5 protein level that leads to higher methylosome activity and promotes epithelial–mesenchymal transition, has recently been correlated with a poor survival of cancer patients. The PRMT5 inhibitor JNJ-64619178 has been selected as a clinical candidate based on its high selectivity and potency, paired with favorable oral pharmacokinetics and safety properties. JNJ-64619178 binds simultaneously to the SAM- and protein substrate- binding pockets of the PRMT5/MEP50 complex with a pseudo-irreversible mode-of-action. Chemical proteomics, methylomics and RNA-sequencing analyses of PRMT5 inhibitor treated cell line samples support the current biological understanding of PRMT5 as a regulator of alternative splicing events. JNJ-64619178 showed potent and broad inhibition of cellular growth, observed in several cell line panels that represent diverse cancer histologies. Ongoing investigations will explore the potential synthetic lethal correlation between PRMT5 inhibition and cancer driver pathways, including those addicted to altered splicing. Oral administration of JNJ-64619178 resulted in efficient inhibition of di-methylation of SMD1/3 proteins, components of the splicing machinery and direct substrates of the methylosome, in several human NSCLC and SCLC cancer mouse xenograft models. JNJ-64619178 demonstrated dose-dependent tumor growth inhibition and regression in several human NSCLC and SCLC cancer mouse xenograft models with sustained blockage of tumor re-growth after dosing cessation. In summary, JNJ-64619178 has a favorable pre-clinical profile supporting clinical testing in patients with lung cancer and other malignancies. Citation Format: Tongfei Wu, Hillary Millar, Dana Gaffney, Lijs Beke, Geert Mannens, Petra Vinken, Ivan Sommers, Jan-Willem Thuring, Weimei Sun, Christopher Moy, Vineet Pande, Junguo Zhou, Nahor Haddish-Berhane, Mark Salvati, Sylvie Laquerre, Matthew V. Lorenzi, Dirk Brehmer. JNJ-64619178, a selective and pseudo-irreversible PRMT5 inhibitor with potent in vitro and in vivo activity, demonstrated in several lung cancer models [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 4859.
- Published
- 2018
- Full Text
- View/download PDF
49. Abstract 929: Discovery of novel covalent KRASG12C inhibitors that display high potency and selectivity in vitro and in vivo
- Author
-
Carol Thach, Jeff Kucharski, Jingchuan Zhang, Yvonne Yao, Shuangwei Li, Anjali Babbar, Dashyant Dhanak, Rasmus Hansen, Yuan Liu, Yuching Chen, Pingda Ren, Ulf Peters, Yi Liu, Shisheng Li, Ke Yu, Yi Wang, Patrick Parvis Zarrinkar, Xiaohu Deng, Matthew R. Janes, Sarah J. Firdaus, Ata Zarieh, Lian-Sheng Li, Tao Wu, Xin Guo, Jun Feng, Matthew V. Lorenzi, Dana D. Hu-Lowe, and Jeffrey H. Chen
- Subjects
Cancer Research ,Mutation ,Chemistry ,Cell ,Mutant ,Allosteric regulation ,Cancer ,medicine.disease ,medicine.disease_cause ,digestive system diseases ,medicine.anatomical_structure ,Oncology ,In vivo ,medicine ,Cancer research ,Adenocarcinoma ,KRAS - Abstract
Activating mutations in KRAS have a high prevalence in human cancer. The codon 12 glycine to cysteine missense mutation (KRASG12C) is among the most common KRAS mutations, present in non-small cell lung adenocarcinoma (~15 %), colorectal adenocarcinoma (~3 %), and pancreatic adenocarcinoma (~1 %). KRASG12C was previously identified as potentially druggable by allele-specific covalent targeting of cysteine 12 near the allosteric switch II pocket (S-IIP). Building on this early work, we recently described the ARS-853 series of S-IIP KRASG12C inhibitors that covalently react with the GDP-bound state of KRASG12C, trapping KRASG12C in this inactive state. In cells, ARS-853 series compounds profoundly deplete the signaling competent GTP-bound state of KRASG12C, thereby inhibiting downstream RAS signaling. However, this series of covalent KRASG12C inhibitors exhibited modest cellular potency and/or poor pharmacokinetic properties, making them unsuitable for further evaluation of covalent KRASG12C inhibition in animal models. We now describe in further detail the discovery and characterization of a new series of structurally distinct quinazoline based S-IIP KRASG12C inhibitors with substantially improved potency and pharmacologic properties that overcome limitations of the ARS-853 series. Through structure-guided medicinal chemistry optimization we identified compound ARS-1620, a potent, orally bioavailable covalent inhibitor of KRASG12C. The co-crystal structure of ARS-1620 covalently bound to KRASG12C reveals a distinct binding mode and additional interactions, compared to ARS-853. ARS-1620 rapidly engages KRASG12C, depletes KRASG12C-GTP in tumor cell lines, and inhibits downstream RAS signaling in a dose-dependent manner. The compound potently inhibits the growth of cell lines harboring the KRASG12C mutation with little or no effect on control cell lines. ARS-1620 demonstrates robust dose-dependent efficacy with once daily oral administration across a panel of KRASG12C-positive mouse cell line (CDX) and patient-derived (PDX) tumor xenograft models, with no response observed at all doses tested in KRASG12C-negative tumor models. The anti-tumor activity of ARS-1620 correlates with target engagement in the tumors as well as with inhibition of downstream RAS signaling. The in vivo efficacy and mutant selectivity observed with ARS-1620 across a wide range of KRASG12C mouse tumor models provides the first in vivo evidence that the S-IIP targeted approach may be a promising therapeutic strategy for patients with KRASG12C mutant cancers. Citation Format: Liansheng Li, Matthew R. Janes, Jingchuan Zhang, Rasmus Hansen, Ulf Peters, Xin Guo, Yuching Chen, Anjali Babbar, Sarah J. Firdaus, Jun Feng, Jeffrey H. Chen, Shuangwei Li, Shisheng Li, Carol Thach, Yuan Liu, Ata Zarieh, Jeff M. Kucharski, Tao Wu, Ke Yu, Yi Wang, Yvonne Yao, Xiaohu Deng, Patrick P. Zarrinkar, Dashyant Dhanak, Matthew V. Lorenzi, Dana Hu-Lowe, Pingda Ren, Yi Liu. Discovery of novel covalent KRASG12C inhibitors that display high potency and selectivity in vitro and in vivo [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 929.
- Published
- 2018
- Full Text
- View/download PDF
50. Optimization of pyrazole inhibitors of Coactivator Associated Arginine Methyltransferase 1 (CARM1)
- Author
-
Matthew V. Lorenzi, Wayne Vaccaro, Marco M. Gottardis, Simona Bindi, Michael A. Poss, Sandrine Thieffine, George L. Trainor, Lata Jayaraman, Paola Vianello, Fulvia Roletto, Ashok V. Purandare, Suhong Pang, Zhong Chen, Tram N. Huynh, Arturo Galvani, Jieping Geng, and Tiziano Bandiera
- Subjects
Protein-Arginine N-Methyltransferases ,Methyltransferase ,CARM1 ,Clinical Biochemistry ,Pharmaceutical Science ,Pyrazole ,Biochemistry ,Chemical synthesis ,Structure-Activity Relationship ,chemistry.chemical_compound ,Drug Discovery ,Coactivator ,Humans ,Structure–activity relationship ,Enzyme Inhibitors ,skin and connective tissue diseases ,Molecular Biology ,chemistry.chemical_classification ,biology ,fungi ,Organic Chemistry ,body regions ,Enzyme ,chemistry ,Enzyme inhibitor ,Drug Design ,biology.protein ,Pyrazoles ,Molecular Medicine - Abstract
Design, synthesis, and SAR development led to the identification of the potent, novel, and selective pyrazole based inhibitor (7f) of Coactivator Associated Arginine Methyltransferase (CARM1).
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.