Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Cancer Immunol Res ; 11(12): 1589-1597, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37871333

ABSTRACT

Transgenic T-cell receptor (TCR) T cell-based adoptive cell therapies for solid tumors are associated with dramatic initial response rates, but there remain many instances of treatment failure and disease relapse. The association of infusion product cytokine profiles with clinical response has not been explored in the context of TCR T-cell therapy products. Single-cell antigen-dependent secretomic and proteomic analysis of preinfusion clinical TCR T-cell therapy products revealed that TNFα cytokine functionality of CD8+ T cells and phospho-STAT3 signaling in these cells were both associated with superior clinical responsiveness to therapy. By contrast, CD4+ T-helper 2 cell cytokine profiles were associated with inferior clinical responses. In parallel, preinfusion levels of IL15, Flt3-L, and CX3CL1 were all found to be associated with clinical response to therapy. These results have implications for the development of therapeutic biomarkers and identify potential targets for enrichment in the design of transgenic TCR T-cell therapies for solid tumors.


Subject(s)
Neoplasms , Tumor Necrosis Factor-alpha , Animals , Humans , Mice , Proteomics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Cytokines , Animals, Genetically Modified , Cell- and Tissue-Based Therapy , Mice, Transgenic , STAT3 Transcription Factor
2.
J Immunother Cancer ; 11(5)2023 05.
Article in English | MEDLINE | ID: mdl-37156551

ABSTRACT

BACKGROUND: The tumor antigen NY-ESO-1 has been shown to be an effective target for transgenic adoptive cell therapy (ACT) for the treatment of sarcoma and melanoma. However, despite frequent early clinical responses, many patients ultimately develop progressive disease. Understanding the mechanisms underlying treatment resistance is crucial to improve future ACT protocols. Here, we describe a novel mechanism of treatment resistance in sarcoma involving loss of expression of NY-ESO-1 in response to transgenic ACT with dendritic cell (DC) vaccination and programmed cell death protein-1 (PD-1) blockade. METHODS: A HLA-A*02:01-positive patient with an NY-ESO-1-positive undifferentiated pleomorphic sarcoma was treated with autologous NY-ESO-1-specific T-cell receptor (TCR) transgenic lymphocytes, NY-ESO-1 peptide-pulsed DC vaccination, and nivolumab-mediated PD-1 blockade. RESULTS: Peripheral blood reconstitution with NY-ESO-1-specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. There was initial tumor regression, and immunophenotyping of the peripheral transgenic T cells showed a predominantly effector memory phenotype over time. Tracking of transgenic T cells to the tumor sites was demonstrated in on-treatment biopsy via both TCR sequencing-based and RNA sequencing-based immune reconstitution, and nivolumab binding to PD-1 on transgenic T cells was confirmed at the tumor site. At the time of disease progression, the promoter region of NY-ESO-1 was found to be extensively methylated, and tumor NY-ESO-1 expression was completely lost as measured by RNA sequencing and immunohistochemistry. CONCLUSIONS: ACT of NY-ESO-1 transgenic T cells given with DC vaccination and anti-PD-1 therapy resulted in transient antitumor activity. NY-ESO-1 expression was lost in the post-treatment sample in the setting of extensive methylation of the NY-ESO-1 promoter region. BIOLOGICAL/CLINICAL INSIGHT: Antigen loss represents a novel mechanism of immune escape in sarcoma and a new point of improvement in cellular therapy approaches. TRIAL REGISTRATION NUMBER: NCT02775292.


Subject(s)
Melanoma , Sarcoma , Humans , Nivolumab , Immunotherapy/methods , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism
3.
J Immunother ; 43(9): 273-282, 2020.
Article in English | MEDLINE | ID: mdl-32925563

ABSTRACT

Expression of carbonic-anhydrase IX (CAIX) in clear cell renal cell carcinoma (RCC) makes it an attractive vaccine target. We developed a fusion-gene construct, granulocyte-macrophage (GM) colony-stimulating factor+CAIX, delivered by an adenoviral vector (Ad) into autologous dendritic cells (DCs) in this phase 1 study. The injected immature DCs were expected to stimulate an antigen-specific immune response against CAIX expressing RCC. Three dose-escalation cohorts (5, 15, and 50×10 cells/administration) were injected intradermally q2wk×3 doses based on a 3+3 design. The primary objective was the safety of the injections. Secondary objectives were immune responses using enzyme-linked immunosorbent spot, a serum biomarker panel, and clinical response. Fifteen patients with metastatic RCC were enrolled, and 9 patients received all 3 doses. No serious adverse events were seen. There were 3 (33%) patients with grade 1 fatigue, 1 of whom subsequently experienced grade 2 fatigue. One patient (11%) experienced grade 1-2 leukopenia. Only 1 patient (11%) experienced grade 2 flu-like symptoms. Of the 9 patients who received treatment, 1 expired of progressive disease, 2 patients were lost to follow-up and 6 patients are alive. Of the 6 patients, 5 have progressive disease, and 1 has completed treatment with stable disease at 27 months follow-up. Immune response measurements appeared more robust in higher dose cohorts, which appeared to be related to patients with stable disease at 3 months. These early data show that autologous immature DC-AdGMCAIX can be safely given to metastatic RCC patients without any serious adverse events with CAIX-specific immune response elicited by the treatment. These preliminary data support further study of Ad-GMCAIX, particularly with combination therapies that may enhance clinical activity.


Subject(s)
Antigens, Neoplasm/genetics , Cancer Vaccines/administration & dosage , Carbonic Anhydrase IX/genetics , Carcinoma, Renal Cell/therapy , Dendritic Cells/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Kidney Neoplasms/therapy , Antigens, Neoplasm/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/genetics , Cancer Vaccines/metabolism , Carbonic Anhydrase IX/immunology , Carcinoma, Renal Cell/immunology , Carcinoma, Renal Cell/pathology , Dendritic Cells/metabolism , Disease Management , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Kidney Neoplasms/immunology , Kidney Neoplasms/pathology , Treatment Outcome
4.
Cell Stem Cell ; 25(4): 542-557.e9, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31495780

ABSTRACT

Invariant natural killer T (iNKT) cells are potent immune cells for targeting cancer; however, their clinical application has been hindered by their low numbers in cancer patients. Here, we developed a proof-of-concept for hematopoietic stem cell-engineered iNKT (HSC-iNKT) cell therapy with the potential to provide therapeutic levels of iNKT cells for a patient's lifetime. Using a human HSC engrafted mouse model and a human iNKT TCR gene engineering approach, we demonstrated the efficient and long-term generation of HSC-iNKT cells in vivo. These HSC-iNKT cells closely resembled endogenous human iNKT cells, could deploy multiple mechanisms to attack tumor cells, and effectively suppressed tumor growth in vivo in multiple human tumor xenograft mouse models. Preclinical safety studies showed no toxicity or tumorigenicity of the HSC-iNKT cell therapy. Collectively, these results demonstrated the feasibility, safety, and cancer therapy potential of the proposed HSC-iNKT cell therapy and laid a foundation for future clinical development.


Subject(s)
Hematopoietic Stem Cells/physiology , Immunotherapy, Adoptive/methods , Natural Killer T-Cells/physiology , Neoplasms/therapy , Animals , Cells, Cultured , Genetic Engineering , Humans , Mice , Mice, SCID , Natural Killer T-Cells/transplantation , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 25(3): 1000-1011, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30409823

ABSTRACT

PURPOSE: To improve persistence of adoptively transferred T-cell receptor (TCR)-engineered T cells and durable clinical responses, we designed a clinical trial to transplant genetically-modified hematopoietic stem cells (HSCs) together with adoptive cell transfer of T cells both engineered to express an NY-ESO-1 TCR. Here, we report the preclinical studies performed to enable an investigational new drug (IND) application. EXPERIMENTAL DESIGN: HSCs transduced with a lentiviral vector expressing NY-ESO-1 TCR and the PET reporter/suicide gene HSV1-sr39TK and T cells transduced with a retroviral vector expressing NY-ESO-1 TCR were coadministered to myelodepleted HLA-A2/Kb mice within a formal Good Laboratory Practice (GLP)-compliant study to demonstrate safety, persistence, and HSC differentiation into all blood lineages. Non-GLP experiments included assessment of transgene immunogenicity and in vitro viral insertion safety studies. Furthermore, Good Manufacturing Practice (GMP)-compliant cell production qualification runs were performed to establish the manufacturing protocols for clinical use. RESULTS: TCR genetically modified and ex vivo-cultured HSCs differentiated into all blood subsets in vivo after HSC transplantation, and coadministration of TCR-transduced T cells did not result in increased toxicity. The expression of NY-ESO-1 TCR and sr39TK transgenes did not have a detrimental effect on gene-modified HSC's differentiation to all blood cell lineages. There was no evidence of genotoxicity induced by the lentiviral vector. GMP batches of clinical-grade transgenic cells produced during qualification runs had adequate stability and functionality. CONCLUSIONS: Coadministration of HSCs and T cells expressing an NY-ESO-1 TCR is safe in preclinical models. The results presented in this article led to the FDA approval of IND 17471.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cells/immunology , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes/immunology , Animals , Antigens, Neoplasm/genetics , Cells, Cultured , Clinical Trials as Topic , Drugs, Investigational/therapeutic use , HLA-A2 Antigen/genetics , Hematopoietic Stem Cells/metabolism , Humans , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/genetics , Neoplasms/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism
6.
Clin Cancer Res ; 25(7): 2096-2108, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30573690

ABSTRACT

PURPOSE: Transgenic adoptive cell therapy (ACT) targeting the tumor antigen NY-ESO-1 can be effective for the treatment of sarcoma and melanoma. Preclinical models have shown that this therapy can be improved with the addition of dendritic cell (DC) vaccination and immune checkpoint blockade. We studied the safety, feasibility, and antitumor efficacy of transgenic ACT with DC vaccination, with and without CTLA-4 blockade with ipilimumab. PATIENTS AND METHODS: Freshly prepared autologous NY-ESO-1-specific T-cell receptor (TCR) transgenic lymphocytes were adoptively transferred together with NY-ESO-1 peptide-pulsed DC vaccination in HLA-A2.1-positive subjects alone (ESO, NCT02070406) or with ipilimumab (INY, NCT01697527) in patients with advanced sarcoma or melanoma. RESULTS: Six patients were enrolled in the ESO cohort, and four were enrolled in the INY cohort. Four out of six patients treated per ESO (66%), and two out of four patients treated per INY (50%) displayed evidence of tumor regression. Peripheral blood reconstitution with NY-ESO-1-specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. Tracking of transgenic T cells to the tumor sites was demonstrated in on-treatment biopsies via TCR sequencing. Multiparametric mass cytometry of transgenic cells demonstrated shifting of transgenic cells from memory phenotypes to more terminally differentiated effector phenotypes over time. CONCLUSIONS: ACT of fresh NY-ESO-1 transgenic T cells prepared via a short ex vivo protocol and given with DC vaccination, with or without ipilimumab, is feasible and results in transient antitumor activity, with no apparent clinical benefit of the addition of ipilimumab. Improvements are needed to maintain tumor responses.


Subject(s)
Adoptive Transfer , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Ipilimumab/pharmacology , Neoplasms/immunology , Neoplasms/therapy , Adoptive Transfer/methods , Adult , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/metabolism , Female , Gene Knock-In Techniques , Humans , Immunotherapy , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Middle Aged , Molecular Targeted Therapy , Neoplasms/pathology , Phenotype , Pilot Projects , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Young Adult
7.
J Immunother ; 41(5): 248-259, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29470191

ABSTRACT

Adoptive cell therapy (ACT) consisting of genetically engineered T cells expressing tumor antigen-specific T-cell receptors displays robust initial antitumor activity, followed by loss of T-cell activity/persistence and frequent disease relapse. We characterized baseline and longitudinal T-cell phenotype variations resulting from different manufacturing and administration protocols in patients who received ACT. Patients with melanoma who enrolled in the F5-MART-1 clinical trial (NCT00910650) received infusions of MART-1 T-cell receptors transgenic T cells with MART-1 peptide-pulsed dendritic cell vaccination. Patients were divided into cohorts based on several manufacturing changes in the generation and administration of the transgenic T cells: decreasing ex vivo stimulation/expansion time, increased cell dose, and receiving fresh instead of cryopreserved cells. T-cell phenotypes were analyzed by flow cytometry at baseline and longitudinally in peripheral blood. Transgenic T cells with shorter ex vivo culture/expansion periods displayed significantly increased expression of markers associated with less differentiated naive/memory populations, as well as significantly decreased expression of the inhibitory receptor programmed death 1 (PD1). Patients receiving fresh infusions of transgenic cells demonstrated expansion of central memory T cells and delayed acquisition of PD1 expression compared with patients who received cryopreserved products. Freshly infused transgenic T cells showed persistence and expansion of naive and memory T-cell populations and delayed acquisition of PD1 expression, which correlated with this cohort's superior persistence of transgenic cells and response to dendritic cell vaccines. These results may be useful in designing future ACT protocols.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Melanoma/therapy , Skin Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Cells, Cultured , Cryopreservation , Female , Humans , Immunologic Memory , Immunophenotyping , Lymphocyte Activation , MART-1 Antigen/metabolism , Male , Melanoma/immunology , Middle Aged , Phenotype , Skin Neoplasms/immunology
8.
Mol Cancer Ther ; 15(7): 1568-79, 2016 07.
Article in English | MEDLINE | ID: mdl-27196782

ABSTRACT

The MET receptor tyrosine kinase is involved in cell growth, survival, and invasion. Clinical studies with small molecule MET inhibitors have shown the role of biomarkers in identifying patients most likely to benefit from MET-targeted therapy. AMG 337 is an oral, small molecule, ATP-competitive, highly selective inhibitor of the MET receptor. Herein, we describe AMG 337 preclinical activity and mechanism of action in MET-dependent tumor models. These studies suggest MET is the only therapeutic target for AMG 337. In an unbiased tumor cell line proliferation screen (260 cell lines), a closely related analogue of AMG 337, Compound 5, exhibited activity in 2 of 260 cell lines; both were MET-amplified. Additional studies examining the effects of AMG 337 on the proliferation of a limited panel of cell lines with varying MET copy numbers revealed that high-level focal MET amplification (>12 copies) was required to confer MET oncogene addiction and AMG 337 sensitivity. One MET-amplified cell line, H1573 (>12 copies), was AMG 337 insensitive, possibly because of a downstream G12A KRAS mutation. Mechanism-of-action studies in sensitive MET-amplified cell lines demonstrated that AMG 337 inhibited MET and adaptor protein Gab-1 phosphorylation, subsequently blocking the downstream PI3K and MAPK pathways. AMG 337 exhibited potency in pharmacodynamic assays evaluating MET signaling in tumor xenograft models; >90% inhibition of Gab-1 phosphorylation was observed at 0.75 mg/kg. These findings describe the preclinical activity and mechanism of action of AMG 337 in MET-dependent tumor models and indicate its potential as a novel therapeutic for the treatment of MET-dependent tumors. Mol Cancer Ther; 15(7); 1568-79. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Female , Gene Amplification , Humans , MAP Kinase Signaling System/drug effects , Mice , Necrosis , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Cancer Immunol Res ; 4(3): 194-203, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26787823

ABSTRACT

Tumor responses to programmed cell death protein 1 (PD-1) blockade therapy are mediated by T cells, which we characterized in 102 tumor biopsies obtained from 53 patients treated with pembrolizumab, an antibody to PD-1. Biopsies were dissociated, and single-cell infiltrates were analyzed by multicolor flow cytometry using two computational approaches to resolve the leukocyte phenotypes at the single-cell level. There was a statistically significant increase in the frequency of T cells in patients who responded to therapy. The frequency of intratumoral B cells and monocytic myeloid-derived suppressor cells significantly increased in patients' biopsies taken on treatment. The percentage of cells with a regulatory T-cell phenotype, monocytes, and natural killer cells did not change while on PD-1 blockade therapy. CD8(+) memory T cells were the most prominent phenotype that expanded intratumorally on therapy. However, the frequency of CD4(+) effector memory T cells significantly decreased on treatment, whereas CD4(+) effector T cells significantly increased in nonresponding tumors on therapy. In peripheral blood, an unusual population of blood cells expressing CD56 was detected in two patients with regressing melanoma. In conclusion, PD-1 blockade increases the frequency of T cells, B cells, and myeloid-derived suppressor cells in tumors, with the CD8(+) effector memory T-cell subset being the major T-cell phenotype expanded in patients with a response to therapy.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , T-Lymphocyte Subsets/immunology , Aged , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents/pharmacology , Female , Humans , Male , Melanoma/immunology , Melanoma/secondary , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome
10.
Lab Invest ; 94(11): 1296-308, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25243900

ABSTRACT

Sustained angiogenesis is essential for tumor growth as it provides the tumor with a network of blood vessels that supply both oxygen and essential nutrients. Limiting tumor-associated angiogenesis is a proven strategy for the treatment of human cancer. To date, the rapid detection and quantitation of tumor-associated endothelial cell (TAEC) proliferation has been challenging, largely due to the low frequency of endothelial cells (ECs) within the tumor microenvironment. In this report, we address this problem using a new multiparametric flow cytometry method capable of rapid and precise quantitation of proliferation by measuring bromodeoxyuridine (BrdUrd) uptake in mouse TAECs from established human tumor xenografts. We determined the basal proliferation labeling index of TAECs in two human tumor xenografts representing two distinct histologies, COLO 205 (colorectal cancer) and U-87 (glioblastoma). We then investigated the effects of two large-molecule antiangiogenic agents targeting different biochemical pathways. Blocking angiopoietin-Tie2 signaling with the peptide-Fc fusion protein, trebananib (AMG 386), inhibited proliferation of TAECs, whereas blocking Dll4-Notch signaling with an anti-Dll4-specific antibody induced hyperproliferation of TAECs. These pharmacodynamic studies highlight the sensitivity and utility of this flow cytometry-based method and demonstrate the value of this assay to rapidly assess the in vivo proliferative effects of angiogenesis-targeted agents on both the tumor and the associated vasculature.


Subject(s)
Antibodies, Neutralizing/pharmacology , Endothelial Cells/drug effects , Flow Cytometry/methods , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Receptor, TIE-2/antagonists & inhibitors , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Neutralizing/therapeutic use , Bromodeoxyuridine , Cell Proliferation/drug effects , Colorectal Neoplasms/pathology , Female , Glioblastoma/pathology , Humans , Mice, Nude , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/pathology , Recombinant Fusion Proteins/therapeutic use , Xenograft Model Antitumor Assays
11.
Clin Cancer Res ; 20(9): 2457-65, 2014 May 01.
Article in English | MEDLINE | ID: mdl-24634374

ABSTRACT

PURPOSE: It has been demonstrated that large numbers of tumor-specific T cells for adoptive cell transfer (ACT) can be manufactured by retroviral genetic engineering of autologous peripheral blood lymphocytes and expanding them over several weeks. In mouse models, this therapy is optimized when administered with dendritic cell (DC) vaccination. We developed a short 1-week manufacture protocol to determine the feasibility, safety, and antitumor efficacy of this double cell therapy. EXPERIMENTAL DESIGN: A clinical trial (NCT00910650) adoptively transferring MART-1 T-cell receptor (TCR) transgenic lymphocytes together with MART-1 peptide-pulsed DC vaccination in HLA-A2.1 patients with metastatic melanoma. Autologous TCR transgenic cells were manufactured in 6 to 7 days using retroviral vector gene transfer, and reinfused with (n = 10) or without (n = 3) prior cryopreservation. RESULTS: A total of 14 patients with metastatic melanoma were enrolled and 9 of 13 treated patients (69%) showed evidence of tumor regression. Peripheral blood reconstitution with MART-1-specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. Administration of freshly manufactured TCR transgenic T cells resulted in a higher persistence of MART-1-specific T cells in the blood as compared with cryopreserved. Evidence that DC vaccination could cause further in vivo expansion was only observed with ACT using noncryopreserved T cells. CONCLUSION: Double cell therapy with ACT of TCR-engineered T cells with a very short ex vivo manipulation and DC vaccines is feasible and results in antitumor activity, but improvements are needed to maintain tumor responses.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive , MART-1 Antigen/genetics , Melanoma/immunology , Melanoma/therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes/immunology , Adult , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Dendritic Cells/metabolism , Female , Humans , Immunotherapy, Adoptive/adverse effects , MART-1 Antigen/immunology , Male , Melanoma/diagnosis , Melanoma/mortality , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Peptide Fragments/genetics , Peptide Fragments/immunology , Positron-Emission Tomography , T-Lymphocytes/metabolism , Tomography, X-Ray Computed , Transduction, Genetic , Treatment Outcome , Vaccination
12.
J Nucl Med ; 52(6): 942-9, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21571804

ABSTRACT

UNLABELLED: Conatumumab is a fully human monoclonal antibody that binds to and activates human death receptor 5 (DR5; also known as TRAIL receptor 2). The purpose of this study was to characterize (64)Cu-labeled conatumumab as a PET tracer for imaging DR5 in tumors. METHODS: DOTA-conatumumab was synthesized by incubating conatumumab with 2,2',2″-(10-(2-(2,5-dioxopyrrolidin-1-yloxy)-2-oxoethyl)-1,4,7,10-tetraazacyclododecane-1,4,7-triyl)triacetic acid (DOTA-NHS). The absolute numbers of DOTA molecules per conatumumab molecules were determined by matrix-assisted laser desorption ionization mass spectrometry and electrospray ionization quadrupole time-of-flight mass spectrometry. (64)Cu-DOTA-conatumumab was prepared by incubating (64)CuCl(2) (33-222 MBq) with DOTA-conatumumab at 37°C for 1 h. Binding of conatumumab and DOTA-conatumumab to Fc-coupled human DR5 (huTR2-Fc) was tested in a kinetic analysis assay, and the biologic activity of copper-DOTA-conatumumab was measured using a caspase-3/7 luminescent assay. In vivo evaluation of DOTA-conatumumab and copper-DOTA-conatumumab was done in severe combined immunodeficiency mice bearing Colo205 xenografts: tissue uptake was determined with biodistribution studies, and small-animal PET and autoradiography were used to determine the uptake of (64)Cu-DOTA conatumumab into tumors and other tissues. RESULTS: DOTA-conatumumab was prepared with an average of 5 DOTA molecules per conatumumab molecule. The in vitro median effective concentration required to induce a 50% effect of DOTA-conatumumab and conatumumab from the assay were 389 and 320 pM, respectively. The median effective dose (±SD) of DOTA-conatumumab and conatumumab via the caspase assay was 135 ± 31 and 128 ± 30 pM, respectively. In female CB17 severe combined immunodeficiency mice bearing Colo205 xenografts, DOTA-conatumumab and conatumumab inhibited tumor growth to the same extent. Small-animal PET studies showed tumor uptake at 24 h after injection of the tracer, with a mean standardized uptake value of 3.16 (n = 2). Tumor uptake was decreased by the coadministration of 400 µg of unlabeled conatumumab (mean standardized uptake value, 1.55; n = 2), suggesting saturable uptake. Tissue uptake determined by biodistribution studies was in agreement with the small-animal PET findings. CONCLUSION: These results suggest that (64)Cu-DOTA-conatumumab is a potential PET tracer for imaging DR5 in tumors and may be useful for measuring on-target occupancy by conatumumab.


Subject(s)
Antibodies, Monoclonal , Organometallic Compounds , Radiopharmaceuticals , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Autoradiography , Caspase 3/metabolism , Caspase 7/metabolism , Dose-Response Relationship, Drug , Female , Half-Life , Isotope Labeling/methods , Mice , Mice, SCID , Neoplasms/diagnostic imaging , Organometallic Compounds/chemical synthesis , Organometallic Compounds/pharmacology , Positron-Emission Tomography , Radiopharmaceuticals/chemical synthesis , Radiopharmaceuticals/pharmacology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tissue Distribution
13.
Cytometry A ; 77(9): 849-60, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20623688

ABSTRACT

Conatumumab is a monoclonal antibody specific for death receptor 5 (DR5) that activates caspases leading to DNA fragmentation and tumor-cell death. Like other Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) receptor therapies, conatumumab is currently being evaluated in clinical trials across a variety of tumor types. However, molecular evidence of on-target drug activity in tumors is often an elusive goal for clinical investigation. Here we evaluated a translational approach using a relevant biopsy method, fine needle aspirates (FNAs), to study the on-target pharmacodynamics of conatumumab pre-clinically. As detected by laser scanning cytometry, drug-induced caspase-3 activation in FNA biopsies of Colo205 xenografts correlated well with activated caspase-3 in conventional section-based samples. Furthermore, in tumor-bearing mice, surrogate assays of serum caspase-3/7 activity and serum drug exposure correlated with in situ caspase-3 activation. We found that one advantage of FNA sampling over other sampling techniques was the ability to measure caspase activity on a per cell basis using DNA content information. To adapt the utility of FNAs for measuring pharmacodynamic markers in humans, detection of activated caspase-3 was multiplexed with EpCAM to characterize mock and clinical FNAs from colorectal and nonsmall cell lung cancer patients. These data suggest that FNA sampling is a practical method to cytometrically evaluate tumors for pharmacological impact in a clinical setting.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis , Caspase 3/analysis , Colorectal Neoplasms/pathology , Flow Cytometry/methods , Lung Neoplasms/pathology , Receptors, TNF-Related Apoptosis-Inducing Ligand/antagonists & inhibitors , Animals , Biopsy, Fine-Needle , Cell Line, Tumor , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/enzymology , Female , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Mice , Xenograft Model Antitumor Assays
14.
Cancer Biol Ther ; 9(8): 618-31, 2010 Apr 15.
Article in English | MEDLINE | ID: mdl-20150762

ABSTRACT

Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) binds to death receptors 4 and 5 (DR4, DR5) to transduce apoptotic signals. Conatumumab (AMG 655) is an investigational, fully human monoclonal agonist antibody (IgG(1)) to human DR5, which induces apoptosis via caspase activation. In this study, we demonstrate that conatumumab binds to DR5, activating intracellular caspases in vitro in the presence of a cross-linker. We also show that conatumumab has activity in vivo and inhibits tumor growth in colon (Colo205 and HCT-15), lung (H2122) and pancreatic (MiaPaCa2/T2) xenograft models. Conatumumab also enhances the antitumor activity of chemotherapeutics in vivo. Caspase activation in Colo205 tumors is dose-dependent and correlated with serum concentrations of conatumumab. We demonstrate for the first time that increases in serum caspase-3/7 activity and levels of M30 (neoepitope of caspase-cleaved cytokeratin-18) are linked to activation of the extrinsic apoptotic pathway using conatumumab in a preclinical model. These data suggest that conatumumab has potential as a therapeutic agent for treating patients with multiple tumor types, and that serum caspase-3/7 and M30 levels may serve as biomarkers of conatumumab activity.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Caspase 3/metabolism , Caspase 7/metabolism , Neoplasms/drug therapy , Receptors, TNF-Related Apoptosis-Inducing Ligand/agonists , Animals , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Mice , Neoplasms/enzymology , Neoplasms/pathology , Xenograft Model Antitumor Assays
15.
Clin Cancer Res ; 16(2): 699-710, 2010 Jan 15.
Article in English | MEDLINE | ID: mdl-20068101

ABSTRACT

PURPOSE: The aims were to assess the safety, pharmacokinetics, maximum tolerated dose, and antitumor activity of AMG 102, a fully human hepatocyte growth factor/scatter factor (HGF/SF)-neutralizing monoclonal antibody, in patients with solid tumors. EXPERIMENTAL DESIGN: Patients (N = 40) with refractory advanced solid tumors were enrolled into six sequential dose-escalation cohorts (0.5, 1, 3, 5, 10, or 20 mg/kg AMG 102 i.v. every 2 weeks) and a dose-expansion cohort (20 mg/kg AMG 102 every 2 weeks). Safety, anti-AMG 102 antibody formation, pharmacokinetics, tumor response, and exploratory biomarkers were assessed. RESULTS: AMG 102 was well tolerated up to the planned maximum dose of 20 mg/kg, and the maximum tolerated dose was not reached. Treatment-related adverse events were generally mild and included fatigue (13%), constipation (8%), nausea (8%), vomiting (5%), anorexia (5%), myalgia (5%), and hypertension (5%). Two patients experienced dose-limiting toxicities: one patient (0.5 mg/kg cohort) experienced grade 3 hypoxia and grade 3 dyspnea and one patient (1 mg/kg cohort) experienced grade 3 upper gastrointestinal hemorrhage. No anti-AMG 102 antibodies were detected, and AMG 102 had linear pharmacokinetics within the dose range investigated. Sixteen of 23 (70%) evaluable patients had a best response of stable disease with progression-free survival ranging from 7.9 to 40 weeks. Circulating levels of the biomarker HGF/SF (bound and unbound) increased in a dose-dependent manner, whereas soluble c-Met concentrations were generally similar across doses. CONCLUSIONS: AMG 102 is safe and well tolerated, has a favorable pharmacokinetic profile, and will be further investigated as a monotherapy and in combination with other agents.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Neoplasms/drug therapy , Adult , Aged , Animals , Antibodies, Monoclonal, Humanized , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Disease Progression , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Female , Hepatocyte Growth Factor/immunology , Humans , Male , Maximum Tolerated Dose , Mice , Middle Aged , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Young Adult
16.
Bioorg Med Chem Lett ; 19(22): 6307-12, 2009 Nov 15.
Article in English | MEDLINE | ID: mdl-19819693

ABSTRACT

Deregulation of the receptor tyrosine kinase c-Met has been implicated in several human cancers and is an attractive target for small molecule drug discovery. We previously showed that O-linked triazolopyridazines can be potent inhibitors of c-Met. Herein, we report the discovery of a related series of N-linked triazolopyridazines which demonstrate nanomolar inhibition of c-Met kinase activity and display improved pharmacodynamic profiles. Specifically, the potent time-dependent inhibition of cytochrome P450 associated with the O-linked triazolopyridazines has been eliminated within this novel series of inhibitors. N-linked triazolopyridazine 24 exhibited favorable pharmacokinetics and displayed potent inhibition of HGF-mediated c-Met phosphorylation in a mouse liver PD model. Once-daily oral administration of 24 for 22days showed significant tumor growth inhibition in an NIH-3T3/TPR-Met xenograft mouse efficacy model.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis/physiology , Neovascularization, Physiologic/physiology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Animals , Cell Survival , Humans , Mice , Mice, Nude , Phosphorylation , Xenograft Model Antitumor Assays
17.
J Med Chem ; 51(18): 5766-79, 2008 Sep 25.
Article in English | MEDLINE | ID: mdl-18763753

ABSTRACT

c-Met is a receptor tyrosine kinase that plays a key role in several cellular processes but has also been found to be overexpressed and mutated in different human cancers. Consequently, targeting this enzyme has become an area of intense research in drug discovery. Our studies began with the design and synthesis of novel pyrimidone 7, which was found to be a potent c-Met inhibitor. Subsequent SAR studies identified 22 as a more potent analog, whereas an X-ray crystal structure of 7 bound to c-Met revealed an unexpected binding conformation. This latter finding led to the development of a new series that featured compounds that were more potent both in vitro and in vivo than 22 and also exhibited different binding conformations to c-Met. Novel c-Met inhibitors have been designed, developed, and found to be potent in vitro and in vivo.


Subject(s)
Protein Kinase Inhibitors/chemistry , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Cell Line, Tumor , Crystallography, X-Ray , Drug Evaluation, Preclinical , Humans , Magnetic Resonance Spectroscopy , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Spectrometry, Mass, Electrospray Ionization , Structure-Activity Relationship
18.
Cancer Res ; 68(16): 6680-7, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18701492

ABSTRACT

Recepteur d'origine nantais (RON) is a receptor tyrosine kinase closely related to c-Met. Both receptors are involved in cell proliferation, migration, and invasion, and there is evidence that both are deregulated in cancer. Receptor overexpression has been most frequently described, but other mechanisms can lead to the oncogenic activation of RON and c-Met. They include activating mutations or gene amplification for c-Met and constitutively active splicing variants for RON. We identified a novel inhibitor of RON and c-Met, compound I, and characterized its in vitro and in vivo activities. Compound I selectively and potently inhibited the kinase activity of RON and c-Met with IC(50)s of 9 and 4 nmol/L, respectively. Compound I inhibited hepatocyte growth factor-mediated and macrophage-stimulating protein-mediated signaling and cell migration in a dose-dependent manner. Compound I was tested in vivo in xenograft models that either were dependent on c-Met or expressed a constitutively active form of RON (RONDelta160 in HT-29). Compound I caused complete tumor growth inhibition in NIH3T3 TPR-Met and U-87 MG xenografts but showed only partial inhibition in HT-29 xenografts. The effect of compound I in HT-29 xenografts is consistent with the expression of the activating b-Raf V600E mutation, which activates the mitogen-activated protein kinase pathway downstream of RON. Importantly, tumor growth inhibition correlated with the inhibition of c-Met-dependent and RON-dependent signaling in tumors. Taken together, our results suggest that a small-molecule dual inhibitor of RON/c-Met has the potential to inhibit tumor growth and could therefore be useful for the treatment of patients with cancers where RON and/or c-Met are activated.


Subject(s)
Colonic Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazoles/pharmacology , Quinolines/pharmacology , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Animals , Blotting, Western , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Immunoprecipitation , Mice , Mice, Nude , Molecular Structure , NIH 3T3 Cells , Phosphorylation , Protein Kinase Inhibitors/chemical synthesis , Proto-Oncogene Proteins c-met/metabolism , Pyrazoles/chemical synthesis , Quinolines/chemical synthesis , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Xenograft Model Antitumor Assays
19.
J Med Chem ; 51(13): 3688-91, 2008 Jul 10.
Article in English | MEDLINE | ID: mdl-18553959

ABSTRACT

Deregulation of the receptor tyrosine kinase c-Met has been implicated in human cancers. Pyrazolones with N-1 bearing a pendent hydroxyalkyl side chain showed selective inhibition of c-Met over VEGFR2. However, studies revealed the generation of active, nonselective metabolites. Blocking this metabolic hot spot led to the discovery of 17 (AMG 458). When dosed orally, 17 significantly inhibited tumor growth in the NIH3T3/TPR-Met and U-87 MG xenograft models with no adverse effect on body weight.


Subject(s)
Aminopyridines/administration & dosage , Aminopyridines/chemistry , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/chemistry , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Pyrazoles/administration & dosage , Pyrazoles/chemistry , Administration, Oral , Aminopyridines/chemical synthesis , Aminopyridines/pharmacokinetics , Animals , Cell Survival/drug effects , Cells, Cultured , Drug Design , Humans , Mice , Mice, Inbred BALB C , Molecular Structure , Mutation/genetics , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Pyrazoles/chemical synthesis , Pyrazoles/pharmacokinetics , Structure-Activity Relationship
20.
Cancer Res ; 68(9): 3495-504, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451178

ABSTRACT

Insulin-like growth factor-I (IGF-I) is a polypeptide hormone that can influence growth, differentiation, and survival of cells expressing the cognate type 1 receptor (IGF-IR). To better understand cell autonomous IGF-IR signaling in the epithelial compartment of the prostate gland, we generated a conditional (Cre/loxP) prostate-specific IGF-IR knockout mouse model. In contrast to epidemiologic studies that established a correlation between elevated serum IGF-I and the risk of developing prostate cancer, we show that abrogation of IGF-IR expression in the dorsal and lateral prostate could activate extracellular signal-regulated kinase 1/2 signaling and cause cell autonomous proliferation and hyperplasia. Moreover, persistent loss of IGF-IR expression in dorsal and ventral lobes induced p53-regulated apoptosis and cellular senescence rescue programs, predicting that titration of IGF-IR signaling might facilitate growth of tumors with compromised p53 activity. Therefore, we crossed the mice carrying the prostate-specific IGF-IR knockout alleles into the transgenic adenocarcinoma of the mouse prostate model that is driven, in part, by T antigen-mediated functional inactivation of p53. Consistent with our prediction, prostate epithelial-specific deletion of IGF-IR accelerated the emergence of aggressive prostate cancer when p53 activity was compromised. Collectively, these data support a critical role for IGF-IR signaling in prostate tumorigenesis and identify an important IGF-IR-dependent growth control mechanism.


Subject(s)
Adenocarcinoma/genetics , Epithelium/metabolism , Gene Deletion , Prostate/metabolism , Prostatic Neoplasms/genetics , Receptor, IGF Type 1/genetics , Adenocarcinoma/metabolism , Age Factors , Animals , Apoptosis/genetics , Cell Proliferation , Cellular Senescence/genetics , Gene Expression Regulation, Neoplastic , Gene Targeting , Integrases/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Organ Specificity/genetics , Prostatic Hyperplasia/genetics , Prostatic Hyperplasia/pathology , Prostatic Neoplasms/metabolism , Receptor, IGF Type 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...