Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Future Oncol ; 18(27): 3011-3020, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35920133

ABSTRACT

Aim: This phase II study investigated safety and efficacy of dilpacimab or bevacizumab plus FOLFIRI in patients with previously treated metastatic colorectal cancer (mCRC). Materials & methods: Overall, 66 patients were treated (n = 34 dilpacimab + FOLFIRI; n = 32 bevacizumab + FOLFIRI). Progression-free survival, overall survival, response rates and tolerability were assessed. Results: Median progression-free survival for dilpacimab + FOLFIRI compared with bevacizumab + FOLFIRI was 3.78 months (95% CI: 2.07-7.20) versus 7.36 months (95% CI: 5.68-10.55) (hazard ratio: 3.57; 95% CI: 1.57-8.11; stratified). Median overall survival: 7.95 months for dilpacimab + FOLFIRI; not reached for bevacizumab + FOLFIRI. Objective response rates: 5.6% for dilpacimab + FOLFIRI and 14.7% for bevacizumab + FOLFIRI. Patients treated with dilpacimab + FOLFIRI experienced serious treatment-related adverse events (n = 4; 11.8%), including one case of intestinal perforation leading to death; none were reported for bevacizumab + FOLFIRI. Conclusion: Treatment with dilpacimab + FOLFIRI was not well tolerated and did not provide clinical benefit to patients with mCRC compared with bevacizumab + FOLFIRI. Clinical Trial Registration: NCT03368859 (Clinicaltrials.gov).


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Colorectal Neoplasms , Rectal Neoplasms , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Camptothecin/therapeutic use , Colonic Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Disease-Free Survival , Fluorouracil , Humans , Leucovorin , Rectal Neoplasms/drug therapy
2.
Invest New Drugs ; 38(6): 1815-1825, 2020 12.
Article in English | MEDLINE | ID: mdl-32524319

ABSTRACT

ABBV-176 is an antibody-drug conjugate composed of the humanized antibody h16f (PR-1594804) conjugated to a highly potent, cytotoxic cross-linking pyrrolobenzodiazepine dimer (PBD; SGD-1882) targeting the prolactin receptor (PRLR), which is overexpressed in several solid tumor types. This phase 1, dose-escalation study (NCT03145909) evaluated the safety, pharmacokinetics, and preliminary activity of ABBV-176 in patients with advanced solid tumors likely to exhibit elevated levels of PRLR. Patients received ABBV-176 once every 3 weeks. Dose escalation was by an exposure-adjusted, continual reassessment method. Dose-limiting toxicities (DLTs) were assessed from the first day of dosing until the next dose of ABBV-176 to determine the maximum tolerated dose (MTD) and recommended phase 2 dose (RP2D). Nineteen patients received ABBV-176 at doses from 2.7-109.35 µg/kg. Patients enrolled had colorectal cancer (n = 11), breast cancer (n = 6), or adrenocortical carcinoma (n = 2). DLTs occurred in 4 patients and included thrombocytopenia (n = 2; both at 99.9-µg/kg dose level), neutropenia (n = 2; 78.3-µg/kg and 99.9-µg/kg dose levels), and pancytopenia (n = 1; 109.35-µg/kg dose level). The most common treatment-emergent adverse events related to ABBV-176 were thrombocytopenia, neutropenia, increased aspartate aminotransferase, nausea, fatigue, and pleural effusions. Effusions and edema were common, and timing of onset suggested possible cumulative ABBV-176 toxicity. Tumor expression of PRLR varied among patients enrolled and analyzed. No patient had an objective response. MTD was not formally determined, as identification of a tolerable dose was confounded by late-onset toxicities. ABBV-176 was associated with significant toxicity in this phase 1, dose-escalation study. Although cytopenias were often dose limiting, effusions and edema were also common and had late onset that suggested cumulative toxicity. No responses were observed, although data were available from a small number of patients with variable tumor PRLR expression. This study was terminated after the dosing of 19 patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Immunoconjugates/administration & dosage , Neoplasms/drug therapy , Receptors, Prolactin/antagonists & inhibitors , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Female , Humans , Immunoconjugates/adverse effects , Immunoconjugates/pharmacokinetics , Male , Middle Aged , Neoplasms/metabolism , Receptors, Prolactin/metabolism , Treatment Outcome
3.
BMC Cancer ; 16: 105, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26879245

ABSTRACT

BACKGROUND: c-Met is the receptor tyrosine kinase for hepatocyte growth factor (HGF) encoded by the MET proto-oncogene. Aberrant activation of c-Met resulting from MET amplification and c-Met overexpression is associated with poor clinical outcome in multiple malignancies underscoring the importance of c-Met signaling in cancer progression. Several c-Met inhibitors have advanced to the clinic; however, the development of inhibitory c-Met-directed therapeutic antibodies has been hampered by inherent agonistic activity. METHOD: We generated and tested a bivalent anti-c-Met monoclonal antibody ABT-700 in vitro for binding potency and antagonistic activity and in vivo for antitumor efficacy in human tumor xenografts. Human cancer cell lines and gastric cancer tissue microarrays were examined for MET amplification by fluorescence in situ hybridization (FISH). RESULTS: ABT-700 exhibits a distinctive ability to block both HGF-independent constitutive c-Met signaling and HGF-dependent activation of c-Met. Cancer cells addicted to the constitutively activated c-Met signaling driven by MET amplification undergo apoptosis upon exposure to ABT-700. ABT-700 induces tumor regression and tumor growth delay in preclinical tumor models of gastric and lung cancers harboring amplified MET. ABT-700 in combination with chemotherapeutics also shows additive antitumor effect. Amplification of MET in human cancer tissues can be identified by FISH. CONCLUSIONS: The preclinical attributes of ABT-700 in blocking c-Met signaling, inducing apoptosis and suppressing tumor growth in cancers with amplified MET provide rationale for examining its potential clinical utility for the treatment of cancers harboring MET amplification.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Proto-Oncogene Proteins c-met/drug effects , Proto-Oncogene Proteins c-met/genetics , Animals , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Gene Amplification , Humans , Male , Mice , Mice, SCID , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Protein Binding , Proto-Oncogene Mas , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Xenograft Model Antitumor Assays
4.
Proc Natl Acad Sci U S A ; 106(11): 4390-5, 2009 Mar 17.
Article in English | MEDLINE | ID: mdl-19251665

ABSTRACT

The expression of ASPP2 (53BP2L), a proapoptotic member of a family of p53-binding proteins, is frequently suppressed in many human cancers. Accumulating evidence suggests that ASPP2 inhibits tumor growth; however, the mechanisms by which ASPP2 suppresses tumor formation remain to be clarified. To study this, we targeted the ASPP2 allele in a mouse by replacing exons 10-17 with a neoR gene. ASPP2(-/-) mice were not viable because of an early embryonic lethal event. Although ASPP2(+/-) mice appeared developmentally normal, they displayed an increased incidence of a variety of spontaneous tumors as they aged. Moreover, gamma-irradiated 6-week-old ASPP2(+/-) mice developed an increased incidence of high-grade T cell lymphomas of thymic origin compared with ASPP2(+/+) mice. Primary thymocytes derived from ASPP2(+/-) mice exhibited an attenuated apoptotic response to gamma-irradiation compared with ASPP2(+/+) thymocytes. Additionally, ASPP2(+/-) primary mouse embryonic fibroblasts demonstrated a defective G(0)/G(1) cell cycle checkpoint after gamma-irradiation. Our results demonstrate that ASPP2 is a haploinsufficient tumor suppressor and, importantly, open new avenues for investigation into the mechanisms by which disruption of ASPP2 pathways could play a role in tumorigenesis and response to therapy.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/radiation effects , Cell Cycle/radiation effects , Gamma Rays , Genetic Predisposition to Disease , Heterozygote , Lymphoma, T-Cell/etiology , Lymphoma, T-Cell/genetics , Mice , Mice, Mutant Strains , Neoplasms/etiology , Neoplasms/genetics , Thymus Gland
5.
Clin Cancer Res ; 27(13): 3556-3566, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33820780

ABSTRACT

PURPOSE: Leucine-rich repeat containing 15 (LRRC15) is expressed on stromal fibroblasts in the tumor microenvironment of multiple solid tumor types and may represent an interesting target for therapy, particularly in patients with sarcomas where LRRC15 is also expressed by malignant cells. ABBV-085 is a monomethyl auristatin-E antibody-drug conjugate that targets LRRC15 and showed antineoplastic efficacy in preclinical experiments. Herein, we report findings of ABBV-085 monotherapy or combination therapy in adult patients with sarcomas and other advanced solid tumors. PATIENTS AND METHODS: This first-in-human phase I study (NCT02565758) assessed ABBV-085 safety, pharmacokinetics/pharmacodynamics, and preliminary antitumor activity. The study consisted of two parts: dose escalation and dose expansion. ABBV-085 was administered by intravenous infusion at 0.3 to 6.0 mg/kg every 14 days. RESULTS: In total, 85 patients were enrolled; 45 patients received the recommended expansion dose of 3.6 mg/kg ABBV-085 monotherapy, including 10 with osteosarcoma and 10 with undifferentiated pleomorphic sarcoma (UPS). Most common treatment-related adverse events were fatigue, nausea, and decreased appetite. The overall response rate for patients with osteosarcoma/UPS treated at 3.6 mg/kg was 20%, including four confirmed partial responses. No monotherapy responses were observed for other advanced cancers treated at 3.6 mg/kg. One patient treated with ABBV-085 plus gemcitabine achieved partial response. CONCLUSIONS: ABBV-085 appeared safe and tolerable at a dose of 3.6 mg/kg every 14 days, with preliminary antitumor activity noted in patients with osteosarcoma and UPS. Given the high unmet need in these orphan malignancies, further investigation into targeting LRRC15 in these sarcomas may be warranted.


Subject(s)
Antineoplastic Agents , Bone Neoplasms , Immunoconjugates , Neoplasms , Sarcoma , Adult , Antineoplastic Agents/adverse effects , Bone Neoplasms/drug therapy , Humans , Immunoconjugates/adverse effects , Membrane Proteins/genetics , Neoplasms/pathology , Sarcoma/drug therapy , Sarcoma/genetics , Tumor Microenvironment
6.
Mol Cancer Ther ; 20(10): 1988-1995, 2021 10.
Article in English | MEDLINE | ID: mdl-34315767

ABSTRACT

Dilpacimab (formerly ABT-165), a novel dual-variable domain immunoglobulin, targets both delta-like ligand 4 (DLL4) and VEGF pathways. Here, we present safety, pharmacokinetic (PK), pharmacodynamic (PD), and preliminary efficacy data from a phase I study (trial registration ID: NCT01946074) of dilpacimab in patients with advanced solid tumors. Eligible patients (≥18 years) received dilpacimab intravenously on days 1 and 15 in 28-day cycles at escalating dose levels (range, 1.25-7.5 mg/kg) until progressive disease or unacceptable toxicity. As of August 2018, 55 patients with solid tumors were enrolled in the dilpacimab monotherapy dose-escalation and dose-expansion cohorts. The most common treatment-related adverse events (TRAE) included hypertension (60.0%), headache (30.9%), and fatigue (21.8%). A TRAE of special interest was gastrointestinal perforation, occurring in 2 patients (3.6%; 1 with ovarian and 1 with prostate cancer) and resulting in 1 death. The PK of dilpacimab showed a half-life ranging from 4.9 to 9.5 days, and biomarker analysis demonstrated that the drug bound to both VEGF and DLL4 targets. The recommended phase II dose for dilpacimab monotherapy was established as 3.75 mg/kg, primarily on the basis of tolerability through multiple cycles. A partial response was achieved in 10.9% of patients (including 4 of 16 patients with ovarian cancer). The remaining patients had either stable disease (52.7%), progressive disease (23.6%), or were deemed unevaluable (12.7%). These results demonstrate that dilpacimab monotherapy has an acceptable safety profile, with clinical activity observed in patients with advanced solid tumors.


Subject(s)
Adaptor Proteins, Signal Transducing/immunology , Antibodies, Bispecific/pharmacology , Antineoplastic Agents/pharmacology , Calcium-Binding Proteins/immunology , Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/immunology , Adaptor Proteins, Signal Transducing/blood , Adult , Aged , Antibodies, Bispecific/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Calcium-Binding Proteins/blood , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/immunology , Neoplasms/pathology , Prognosis , Tissue Distribution , Vascular Endothelial Growth Factor A/blood
7.
Mol Cancer Ther ; 19(5): 1210-1217, 2020 05.
Article in English | MEDLINE | ID: mdl-32127466

ABSTRACT

This first-in-human phase I study evaluated the pharmacokinetics, safety, and preliminary efficacy of telisotuzumab, formerly called ABT-700, an antagonistic antibody directed against c-Met. For dose escalation (3+3 design), 3 to 6 patients with advanced solid tumors were enrolled into four dose cohorts (5-25 mg/kg). In the dose-expansion phase, a subset of patients was prospectively selected for MET amplification (FISH screening). Patients received telisotuzumab intravenously on day 1 every 21 days. For dose expansion, 15 mg/kg was chosen as the dose on the basis of safety, pharmacokinetics, and other data from the escalation cohorts. Forty-five patients were enrolled and received at least one dose of telisotuzumab (dose escalation, n = 15; dose expansion, n = 30). Telisotuzumab showed a linear pharmacokinetics profile; peak plasma concentration was proportional to dose level. There were no acute infusion reactions and no dose-limiting toxicities were observed. The most common treatment-related adverse events included hypoalbuminemia (n = 9, 20.0%) and fatigue (n = 5, 11.1%). By Response Evaluation Criteria In Solid Tumors (RECIST), 4 of 10 (40.0%) patients with MET-amplified tumors had confirmed partial response in target lesions (one ovarian, two gastric, and one esophageal), two (20.0%) had stable disease, three (30.0%) had progressive disease; one patient was unable to be evaluated. Among patients with nonamplified tumors (n = 35), no objective responses were observed; however, 11 patients had stable disease per RECIST criteria. In conclusion, telisotuzumab has an acceptable safety profile with clinical activity observed in patients with MET-amplified advanced solid tumors.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Neoplasms/drug therapy , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Female , Follow-Up Studies , Humans , Male , Maximum Tolerated Dose , Middle Aged , Neoplasms/immunology , Neoplasms/pathology , Prognosis , Prospective Studies , Proto-Oncogene Proteins c-met/immunology , Retrospective Studies , Tissue Distribution
8.
J Clin Oncol ; 36(33): 3298-3306, 2018 11 20.
Article in English | MEDLINE | ID: mdl-30285518

ABSTRACT

PURPOSE: This first-in-human study evaluated telisotuzumab vedotin (Teliso-V), formerly called ABBV-399, an antibody-drug conjugate of the anti-c-Met monoclonal antibody ABT-700 and monomethyl auristatin E. MATERIALS AND METHODS: For dose escalation, three to six patients with advanced solid tumors were enrolled in eight cohorts (0.15 to 3.3 mg/kg). The dose-expansion phase enrolled patients with non-small-cell lung cancer (NSCLC) with c-Met-overexpressing tumors (c-Met positive; immunohistochemistry membrane H-score ≥ 150). Patients received Teliso-V monotherapy intravenously on day 1 once every 3 weeks. Safety, tolerability, pharmacokinetics, and maximum tolerated dose were determined. RESULTS: Forty-eight patients were enrolled (median age, 65 years; 35.4% NSCLC; median four prior therapies). One patient each in the 3.0-mg/kg (n = 9) and 3.3-mg/kg (n = 3) cohorts experienced dose-limiting toxicities. Although the maximum tolerated dose was not formally identified, the recommended phase II dose was defined as 2.7 mg/kg on the basis of overall safety and tolerability. The most frequent treatment-emergent adverse events (any grade) were fatigue (42%), nausea (27%), constipation (27%), decreased appetite (23%), vomiting (21%), dyspnea (21%), diarrhea (19%), peripheral edema (19%), and neuropathy (17%). The most frequent Teliso-V-related grade ≥ 3 adverse events were fatigue, anemia, neutropenia, and hypoalbuminemia (4% each). Teliso-V and total antibody pharmacokinetics were approximately dose proportional, with a mean harmonic half-life of 2 to 4 days each. Prospective screening identified 35 (60%) of 58 patients with c-Met-positive NSCLC. Of 16 patients with c-Met-positive NSCLC who were treated with Teliso-V 2.4 to 3.0 mg/kg, three (18.8%; 95% CI, 4.1% to 45.7%) achieved a partial response (median response duration, 4.8 months; median progression-free survival, 5.7 months; 95% CI, 1.2 months to 15.4 months). No other patients experienced a response. CONCLUSION: Teliso-V monotherapy demonstrated favorable safety and tolerability profiles, with encouraging evidence of antitumor activity in patients with c-Met-positive NSCLC.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/pharmacokinetics , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Dose-Response Relationship, Drug , Female , Humans , Male , Maximum Tolerated Dose , Middle Aged , Proto-Oncogene Proteins c-met/antagonists & inhibitors
9.
Mol Cancer Ther ; 17(5): 1039-1050, 2018 05.
Article in English | MEDLINE | ID: mdl-29592882

ABSTRACT

Antiangiogenic therapy is a clinically validated modality in cancer treatment. To date, all approved antiangiogenic drugs primarily inhibit the VEGF pathway. Delta-like ligand 4 (DLL4) has been identified as a potential drug target in VEGF-independent angiogenesis and tumor-initiating cell (TIC) survival. A dual-specific biologic targeting both VEGF and DLL4 could be an attractive strategy to improve the effectiveness of anti-VEGF therapy. ABT-165 was uniquely engineered using a proprietary dual-variable domain immunoglobulin (DVD-Ig) technology based on its ability to bind and inhibit both DLL4 and VEGF. In vivo, ABT-165 induced significant tumor growth inhibition compared with either parental antibody treatment alone, due, in part, to the disruption of functional tumor vasculature. In combination with chemotherapy agents, ABT-165 also induced greater antitumor response and outperformed anti-VEGF treatment. ABT-165 displayed nonlinear pharmacokinetic profiles in cynomolgus monkeys, with an apparent terminal half-life > 5 days at a target saturation dose. In a GLP monkey toxicity study, ABT-165 was well-tolerated at doses up to 200 mg/kg with non-adverse treatment-related histopathology findings limited to the liver and thymus. In summary, ABT-165 represents a novel antiangiogenic strategy that potently inhibits both DLL4 and VEGF, demonstrating favorable in vivo efficacy, pharmacokinetic, and safety profiles in preclinical models. Given these preclinical attributes, ABT-165 has progressed to a phase I study. Mol Cancer Ther; 17(5); 1039-50. ©2018 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Glioblastoma/drug therapy , Immunoglobulins/pharmacology , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Membrane Proteins/antagonists & inhibitors , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Xenograft Model Antitumor Assays , Animals , Antineoplastic Combined Chemotherapy Protocols/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacokinetics , Cell Line, Tumor , Drug Screening Assays, Antitumor/methods , Glioblastoma/metabolism , Glioblastoma/pathology , HT29 Cells , Humans , Immunoglobulins/metabolism , Immunologic Factors/metabolism , Immunologic Factors/pharmacokinetics , Immunologic Factors/pharmacology , Intracellular Signaling Peptides and Proteins/metabolism , Macaca fascicularis/metabolism , Membrane Proteins/metabolism , Treatment Outcome , Vascular Endothelial Growth Factor A/metabolism
10.
Mol Cancer ; 6: 9, 2007 Jan 19.
Article in English | MEDLINE | ID: mdl-17233922

ABSTRACT

BACKGROUND: Sapphyrin analogues and related porphyrin-like species have attracted attention as anticancer agents due to their selective localization in various cancers, including hematologic malignancies, relative to surrounding tissues. Sapphyrins are electron affinic compounds that generate high yields of singlet oxygen formation. Although initially explored in the context of photodynamic therapy, sapphyrins have intrinsic anticancer activity that is independent of their photosensitizing properties. However, the mechanisms for their anticancer activity have not been fully elucidated. RESULTS: We have prepared a series of hydrophilic sapphyrins and evaluated their effect on proliferation, uptake, and cell death in adherent human lung (A549) and prostate (PC3) cancer cell lines and in an A549 xenograft tumor model. PCI-2050, the sapphyrin derivative with the highest in vitro growth inhibitory activity, significantly lowered 5-bromo-2'-deoxyuridine incorporation in S-phase A549 cells by 60% within eight hours and increased levels of reactive oxygen species within four hours. The growth inhibition pattern of PCI-2050 in the National Cancer Institute 60 cell line screen correlated most closely using the COMPARE algorithm with known transcriptional or translational inhibitors. Gene expression analyses conducted on A549 plateau phase cultures treated with PCI-2050 uncovered wide-spread decreases in mRNA levels, which especially affected short-lived transcripts. Intriguingly, PCI-2050 increased the levels of transcripts involved in RNA processing and trafficking, transcriptional regulation, and chromatin remodeling. We propose that these changes reflect the activation of cellular processes aimed at countering the observed wide-spread reductions in transcript levels. In our A549 xenograft model, the two lead compounds, PCI-2050 and PCI-2022, showed similar tumor distributions despite differences in plasma and kidney level profiles. This provides a possible explanation for the better tolerance of PCI-2022 relative to PCI-2050. CONCLUSION: Hydrophilic sapphyrins were found to display promise as novel agents that localize to tumors, generate oxidative stress, and inhibit gene expression.


Subject(s)
Hydrophobic and Hydrophilic Interactions , Oligonucleotide Array Sequence Analysis , Porphyrins/pharmacokinetics , Porphyrins/toxicity , Cell Line, Tumor , Cell Proliferation , Cluster Analysis , Gene Expression Profiling , Humans , Molecular Structure , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
11.
Cancer Res ; 65(24): 11676-88, 2005 Dec 15.
Article in English | MEDLINE | ID: mdl-16357179

ABSTRACT

There is an emerging appreciation of the importance of zinc in regulating cancer cell growth and proliferation. Recently, we showed that the anticancer agent motexafin gadolinium (MGd) disrupted zinc metabolism in A549 lung cancer cells, leading, in the presence of exogenous zinc, to cell death. Here, we report the effect of MGd and exogenous zinc on intracellular levels of free zinc, oxidative stress, proliferation, and cell death in exponential phase human B-cell lymphoma and other hematologic cell lines. We find that increased levels of oxidative stress and intracellular free zinc precede and correlate with cell cycle arrest and apoptosis. To better understand the molecular basis of these cellular responses, gene expression profiling analyses were conducted on Ramos cell cultures treated with MGd and/or zinc acetate. Cultures treated with MGd or zinc acetate alone elicited transcriptional responses characterized by induction of metal response element-binding transcription factor-1 (MTF-1)-regulated and hypoxia-inducible transcription factor-1 (HIF-1)-regulated genes. Cultures cotreated with MGd and zinc acetate displayed further increases in the levels of MTF-1- and HIF-1-regulated transcripts as well as additional transcripts regulated by NF-E2-related transcription factor 2. These data provide insights into the molecular changes that accompany the disruption of intracellular zinc homeostasis and support a role for MGd in treatment of B-cell hematologic malignancies.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Lymphoma, B-Cell/drug therapy , Metalloporphyrins/pharmacology , Oxidative Stress/drug effects , Zinc/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Profiling , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphoma, B-Cell/metabolism , Lymphoma, B-Cell/pathology , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Oligonucleotide Array Sequence Analysis , Response Elements , S Phase/drug effects , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured , Transcription Factor MTF-1
12.
Mol Cancer Ther ; 5(5): 1176-82, 2006 May.
Article in English | MEDLINE | ID: mdl-16731749

ABSTRACT

Motexafin gadolinium (MGd, Xcytrin) is a tumor-selective expanded porphyrin that targets oxidative stress-related proteins. MGd treatment of the follicular lymphoma-derived cell line HF-1 resulted in growth suppression and apoptosis whereas MGd treatment of the Burkitt's lymphoma-derived cell line Ramos resulted in growth suppression but not apoptosis. Because phosphorylation status of Akt/protein kinase B is regulated by oxidative stress, we monitored total and phosphorylated Akt (pAkt) in MGd-treated HF-1 and Ramos cells. Levels of pAkt increased within 30 minutes after MGd treatment of HF-1 but after 4 hours began to show a progressive decline to below baseline levels before cells underwent apoptosis. In MGd-treated Ramos cells, pAkt increased approximately 2-fold within 4 hours and remained persistently elevated. Because pAkt activates survival pathways, we determined if MGd-induced cell death could be enhanced by inhibiting phosphorylation of Akt. The addition of specific inhibitors of Akt phosphorylation (Akt inhibitor 1 or SH-5) reduced pAkt levels in MGd-treated HF-1 and Ramos cells and synergistically enhanced MGd-induced cell death. MGd was also evaluated in combination with celecoxib, an inhibitor of Akt phosphorylation, or docetaxel, a microtubule inhibitor that can decrease Akt phosphorylation. The combination of MGd/celecoxib or MGd/docetaxel resulted in decreased Akt phosphorylation and in synergistic cytotoxicity compared with either agent alone. These data point to a potential protective role for pAkt in MGd-induced apoptosis and suggest that MGd activity may be enhanced by combining it with agents that inhibit Akt phosphorylation.


Subject(s)
Antineoplastic Agents/pharmacology , Metalloporphyrins/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Celecoxib , Cell Line, Tumor , Cyclooxygenase Inhibitors/metabolism , Cyclooxygenase Inhibitors/pharmacology , Docetaxel , Drug Synergism , Humans , Lymphoma/metabolism , Metalloporphyrins/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrazoles/metabolism , Pyrazoles/pharmacology , Pyrazoles/toxicity , Sulfonamides/metabolism , Sulfonamides/pharmacology , Sulfonamides/toxicity , Taxoids/metabolism , Taxoids/pharmacology
13.
Mol Cancer Ther ; 5(11): 2798-805, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17121926

ABSTRACT

Sapphyrins are pentapyrrolic metal-free expanded porphyrins with potential medical use as anticancer agents. The novel sapphyrin derivative, PCI-2050, functionalized with 2-[2-(2-methoxyethoxy)ethoxy]ethoxy groups to enhance solubility and a modified bipyrrole moiety was found to be more potent in inducing apoptosis than the previously described sapphyrin PCI-2000. Because some sapphyrins may localize to tumors, we took advantage of the intrinsic fluorescence of these compounds to develop a flow cytometry-based assay to track sapphyrin biodistribution in tumor-bearing mice. Ex vivo analysis of sapphyrin-injected animals revealed that PCI-2050 preferentially localized to tumor, whereas PCI-2000 distributed into normal tissues rather than tumor. PCI-2050 uptake in xenograft tumor cells and resultant tumor cell cytotoxicity was dose dependent. To investigate structure-activity relationships, we focused on PCI-2050 and three derivatives that differ by their alkyl substituents on the bipyrrole moiety: PCI-2051, PCI-2052, and PCI-2053. Treatment of Ramos cells in culture or treatment of Ramos xenograft-bearing animals with each of the sapphyrins followed by ex vivo growth of tumor cells revealed the same pattern of cytotoxicity: PCI-2050 > PCI-2052 > PCI-2051 > PCI-2053. Thus, subtle changes in the alkyl substituents on the bipyrrole moiety result in significant changes in antitumor activity. PCI-2050 displayed significant antitumor efficacy in both Ramos and RKO xenograft models without hematologic, hepatic, or renal abnormalities as assessed by complete blood counts and serum chemistries. On the basis of these findings, it is concluded that the sapphyrin PCI-2050 warrants further evaluation as a potential anticancer agent due to its intrinsic proapoptotic activity and tumor localization ability.


Subject(s)
Antineoplastic Agents/pharmacology , Porphyrins/pharmacology , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Cell Line, Tumor , Female , Flow Cytometry , Mice , Neoplasms/metabolism , Porphyrins/chemical synthesis , Porphyrins/chemistry , Porphyrins/pharmacokinetics , Xenograft Model Antitumor Assays
14.
Clin Cancer Res ; 23(4): 992-1000, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27573171

ABSTRACT

Purpose: Despite the importance of the MET oncogene in many malignancies, clinical strategies targeting c-Met have benefitted only small subsets of patients with tumors driven by signaling through the c-Met pathway, thereby necessitating selection of patients with MET amplification and/or c-Met activation most likely to respond. An ADC targeting c-Met could overcome these limitations with potential as a broad-acting therapeutic.Experimental Design: ADC ABBV-399 was generated with the c-Met-targeting antibody, ABT-700. Antitumor activity was evaluated in cancer cells with overexpressed c-Met or amplified MET and in xenografts including patient-derived xenograft (PDX) models and those refractory to other c-Met inhibitors. The correlation between c-Met expression and sensitivity to ABBV-399 in tumor and normal cell lines was assessed to evaluate the risk of on-target toxicity.Results: A threshold level of c-Met expressed by sensitive tumor but not normal cells is required for significant ABBV-399-mediated killing of tumor cells. Activity extends to c-Met or amplified MET cell line and PDX models where significant tumor growth inhibition and regressions are observed. ABBV-399 inhibits growth of xenograft tumors refractory to other c-Met inhibitors and provides significant therapeutic benefit in combination with standard-of-care chemotherapy.Conclusions: ABBV-399 represents a novel therapeutic strategy to deliver a potent cytotoxin to c-Met-overexpressing tumor cells enabling cell killing regardless of reliance on MET signaling. ABBV-399 has progressed to a phase I study where it has been well tolerated and has produced objective responses in c-Met-expressing non-small cell lung cancer (NSCLC) patients. Clin Cancer Res; 23(4); 992-1000. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins c-met/genetics , Animals , Antibodies, Monoclonal/adverse effects , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , MCF-7 Cells , Mice , Neoplasms/immunology , Neoplasms/pathology , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
15.
Mol Cancer Ther ; 4(6): 968-76, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15956254

ABSTRACT

Sapphyrins are pentapyrrolic, metal-free, expanded porphyrins. In the present study, the activity of sapphyrins as anticancer agents in hematopoietic-derived tumor cells was explored. It was found that a dihydroxylated water-soluble sapphyrin derivative (PCI-2000) is a potent inducer of apoptosis in a wide variety of tumor cell lines including lymphoma (Ramos, DHL-4, and HF-1), leukemia (Jurkat and HL-60), and myeloma (8226/S, 1-310, C2E3, and 1-414). PCI-2000 triggers an apoptotic pathway in these tumor cells as shown by release of cytochrome c from mitochondria; activation of caspases 9, 8, and 3; cleavage of the caspase substrate poly(ADP-ribose) polymerase; and Annexin V binding. Apoptosis can be partially inhibited by overexpression of the antiapoptotic protein Bcl-2 or treatment with benzyloxycarbonyl-valine-alanine-aspartic acid-fluoromethylketone, a cell-permeable caspase inhibitor. Both PCI-2000 and PCI-2010, a tetrahydroxy bis-carbamate derivative of PCI-2000, result in increased levels of phosphorylated p38 mitogen-activated protein kinase. Inhibition of p38 mitogen-activated protein kinase phosphorylation resulted in a synergistic increase of PCI-2000 cytotoxicity. PCI-2010 showed less toxicity in mice than PCI-2000 and was active in slowing the growth of Ramos and HL-60 tumor xenografts in nude mice. These results provide preclinical rationale for the further study of sapphyrins for potential use in the treatment of hematopoietic-derived tumors.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Leukemia/pathology , Lymphoma/pathology , Porphyrins/pharmacology , Angiogenesis Inhibitors/chemistry , Animals , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/metabolism , Cytoplasm/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation, Neoplastic , Humans , Leukemia/metabolism , Lymphoma/metabolism , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/metabolism , Molecular Structure , Phosphorylation/drug effects , Porphyrins/chemistry , Porphyrins/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Oncogene ; 21(51): 7901-11, 2002 Nov 07.
Article in English | MEDLINE | ID: mdl-12420228

ABSTRACT

The tumor suppressor protein p53, once activated, can cause either cell cycle arrest or apoptosis through transactivation of target genes with p53 DNA binding sites (DBS). To investigate the role of p53 DBS in the regulation of this profound, yet poorly understood decision of life versus death, we systematically studied all known and potential p53 DBS. We analysed the DBS separated from surrounding promoter regions in yeast and mammalian assays with and without DNA damage. p53 efficiently utilized the DBS of MDM2 and of genes connected to cell cycle arrest, DNA repair and the death receptor pathway of apoptosis. However, p53 was unable to utilize two-thirds of the isolated DBS, a subset that included almost all DBS of apoptosis-related genes. Neither ASPP2, a p53-interacting protein reported to specifically stimulate p53 transcriptional activity on apoptosis-related promoters, nor DNA damage resulted in p53 utilization of isolated DBS of apoptosis-related genes. Thus, a major regulation of p53 activity occurs at the level of p53 DBS themselves by posing additional requirements for the successful utilization of apoptosis-related DBS.


Subject(s)
Binding Sites , DNA/metabolism , Gene Expression Profiling , Tumor Suppressor Protein p53/physiology , Apoptosis/genetics , Cell Cycle/genetics , Consensus Sequence , DNA Damage , DNA Repair/genetics , Fungal Proteins/biosynthesis , Fungal Proteins/genetics , Genes, Reporter , Genes, p53 , Humans , Luciferases/biosynthesis , Luciferases/genetics , Plasmids/genetics , Recombinant Fusion Proteins/biosynthesis , Saccharomyces cerevisiae/metabolism , Transfection
17.
Mol Cancer Res ; 1(7): 508-18, 2003 May.
Article in English | MEDLINE | ID: mdl-12754298

ABSTRACT

We have identified a novel protein, apoptotic regulator in the membrane of the endoplasmic reticulum (ARMER), which protects HT1080 fibrosarcoma cells from apoptosis induced by various stimuli. We demonstrate that ARMER is an endoplasmic reticulum (ER) integral membrane protein with four predicted transmembrane domains and a COOH-terminal KKXX ER retrieval motif. We used an inducible expression system (pIND) to study the effects of regulated ARMER overexpression. Cells in which ARMER was overexpressed exhibited protection from multiple apoptotic inducers including serum starvation, doxorubicin, UV irradiation, tumor necrosis factor alpha, and the ER stressors brefeldin A, tunicamycin, and thapsigargin. Analysis of the caspase proteolytic cascade reveals that ARMER inhibits proteolysis of the caspase-9-specific fluorogenic substrate LEHD-AFC as well as endogenous substrates downstream of caspase-9; however, it does not inhibit cytochrome c release or cleavage of caspase-9 itself. Apoptotic stimuli cause endogenous levels of ARMER protein and RNA to decrease, leading to cell death; however, sustaining ARMER protein levels through exogenous expression inhibits apoptosis. These data suggest that ARMER is a novel ER integral membrane protein which protects cells by inhibiting caspase-9 activity and reveal a possible role for ARMER in cell survival.


Subject(s)
Apoptosis/physiology , Cell Survival/physiology , Membrane Proteins/genetics , Adaptor Proteins, Signal Transducing , Blotting, Northern , Endoplasmic Reticulum/physiology , Fibrosarcoma , Humans , Recombinant Proteins/metabolism , Transfection , Tumor Cells, Cultured
18.
J Interferon Cytokine Res ; 22(2): 237-43, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11911807

ABSTRACT

As a consequence of the activation of the apoptotic pathway, the subcellular localization of many proteins that modulate cell death is altered. Nmi and IFP 35 are homologous interferon (IFN)-inducible proteins that associate into a 300-400-kDa complex in the cytoplasm. Prior to treatment with IFN, subcellular fractionation reveals that the proteins are found primarily in a soluble cytoplasmic fraction, and immunofluorescence shows a diffuse cytoplasmic distribution. Treatment with IFN results in some Nmi and IFP 35 fractionating with membranes and colocalizing into speckles as detected by immunofluorescence. In IFN-treated apoptotic cells, the speckles dissociate and are replaced by a diffuse cytoplasmic distribution of Nmi and IFP 35. In Jurkat cells, dissociation of speckles could not be separated from other markers of apoptosis, including exposure of phosphatidylserine on the cell surface, caspase cleavage of D4-GDI, and nuclear condensation and fragmentation. The caspase inhibitor, zVAD-fmk, was not able to inhibit the dissociation of speckles in vivo or in a permeabilized cell assay in vitro, suggesting that dissociation of speckles is a caspase-independent process. Nmi and IFP 35 proteins are not caspase substrates, as total levels of Nmi and IFP 35 and their association into a cytoplasmic complex do not change in apoptotic cells. Consistent with dissociation of speckles, subcellular fractionation studies show redistribution of Nmi and IFP 35 from the membrane fraction into the S100 fraction of apoptotic cells. Our studies have revealed dissociation of the IFN-induced Nmi/IFP 35 speckles as a newly defined specific event occurring during apoptosis.


Subject(s)
Apoptosis/drug effects , Carrier Proteins/metabolism , Interferons/pharmacology , Intracellular Signaling Peptides and Proteins , Nuclear Proteins/metabolism , Carrier Proteins/immunology , Cell Extracts/chemistry , Cell Fractionation , Cell Membrane/chemistry , Cell Membrane/drug effects , Cytoplasm/chemistry , Cytoplasm/drug effects , Fluorescent Antibody Technique, Indirect , HeLa Cells , Humans , Inhibitor of Differentiation Proteins , Intracellular Fluid/immunology , Intracellular Fluid/metabolism , Jurkat Cells/chemistry , Jurkat Cells/drug effects , Leucine Zippers , Macromolecular Substances , Molecular Weight , Nuclear Proteins/immunology , Protein Structure, Tertiary/drug effects , Tumor Cells, Cultured
19.
Cancer Lett ; 180(2): 195-202, 2002 Jun 28.
Article in English | MEDLINE | ID: mdl-12175552

ABSTRACT

In order to better understand how tumor cells develop resistance to chemotherapy drugs, we screened a human cDNA expression library in Jurkat cells for cDNA's that conferred resistance to doxorubicin-induced cell death. One of the cDNA's isolated in the screen codes for ribosomal protein L35a, a component of the large subunit of the ribosome. Jurkat cells engineered to overexpress L35a protein were more resistant not only to doxorubicin but also to UV-irradiation, anti-Fas antibody, and serum starvation compared to Jurkat cells expressing endogenous levels of L35a. Jurkat cells overexpressing L35a did not have increased levels of the anti-apoptotic proteins Bcl-2 or Bcl-xL, the drug efflux pump P-glycoprotein, nor altered cellular growth kinetics or total protein synthesis. Our results provide new insight into L35a function and suggest that it may have a role in the cellular response to cytotoxic damage. Since L35a RNA is overexpressed in a significant number of glioblastoma multiforme (GBM) brain tumors, our results may stimulate further investigation into the possible role of L35a in the resistance of GBM to cytotoxic therapy.


Subject(s)
Ribosomal Proteins/physiology , Cell Death , Cell Division , DNA, Complementary/isolation & purification , Drug Resistance, Neoplasm , Glioblastoma/drug therapy , Glioblastoma/metabolism , Humans , Jurkat Cells , Protein Biosynthesis , Ribosomal Proteins/genetics
20.
Nutr Clin Pract ; 18(6): 517-20, 2003 Dec.
Article in English | MEDLINE | ID: mdl-16215087

ABSTRACT

BACKGROUND: Both physiologic and psychological reasons for cycling total parenteral nutrition (TPN) have been well established. Despite widespread acceptance of this practice, the only previously published method for calculating TPN cycle rates is inherently flawed. METHODS: A mathematical formula was derived to facilitate reliable calculation of cyclic TPN flow rates as a function of total volume and cycle time. A publicly accessible website was subsequently developed to expedite rapid determination of TPN cycles. RESULTS: A fail-safe method of calculating TPN cycle flow rates can be expressed as F = V/(4T-10), where F is equal to the basal flow rate (mL/h), T is equal to the desired cycle time (hours), and V is equal to the total volume of TPN (mL) to be delivered in 24 hours. The basal flow rate and twice the basal flow rate are used for the first and last 2 hours of the TPN cycle, and the remainder of the cycle runs at 4 times the basal flow rate. TPN cycles may be easily calculated online using this formula at http://peds.stanford.edu/tpn.html. CONCLUSIONS: We have developed a fail-safe method of calculating TPN cycle flow rates that will consistently deliver the desired volume and have made an online implementation of this formula publicly available.

SELECTION OF CITATIONS
SEARCH DETAIL