Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 30(8): 1630-1641, 2024 Apr 15.
Article in English | MEDLINE | ID: mdl-38319672

ABSTRACT

PURPOSE: The immunocytokine cergutuzumab amunaleukin (CEA-IL2v) showed manageable safety and favorable pharmacodynamics in phase I/Ib trials in patients with advanced/metastatic carcinoembryonic antigen-positive (CEA+) solid tumors, but this was accompanied by a high incidence of anti-drug antibodies (ADA). We examined B-cell depletion with obinutuzumab as a potential mitigation strategy. EXPERIMENTAL DESIGN: Preclinical data comparing B-cell depletion with rituximab versus obinutuzumab are summarized. Substudies of phase I/Ib trials investigated the effect of obinutuzumab pretreatment on ADA development, safety, pharmacodynamics, and antitumor activity of CEA-IL2v ± atezolizumab in patients with advanced/metastatic or unresectable CEA+ solid tumors who had progressed on standard of care. RESULTS: Preclinical data showed superior B-cell depletion with obinutuzumab versus rituximab. In clinical studies, patients received CEA-IL2v monotherapy with (n = 16) or without (n = 6) obinutuzumab pretreatment (monotherapy study), or CEA-IL2v + atezolizumab + obinutuzumab pretreatment (n = 5; combination study). In the monotherapy study, after four cycles (every 2 weeks treatment), 0/15 evaluable patients administered obinutuzumab pretreatment had ADAs versus 4/6 patients without obinutuzumab. Obinutuzumab pretreatment with CEA-IL2v monotherapy showed no new safety signals and pharmacodynamic data suggested minimal impact on T cells and natural killer cells. Conversely, increased liver toxicity was observed in the combination study (CEA-IL2v + atezolizumab + obinutuzumab pretreatment). CONCLUSIONS: These preliminary findings suggest that obinutuzumab pretreatment before CEA-IL2v administration in patients with CEA+ solid tumors may be a feasible and potent ADA mitigation strategy, with an acceptable safety profile, supporting broader investigation of obinutuzumab pretreatment for ADA mitigation in other settings.


Subject(s)
Antibodies, Monoclonal, Humanized , Carcinoembryonic Antigen , Neoplasms , Humans , Rituximab , Neoplasms/drug therapy
2.
Anal Chem ; 92(16): 11135-11144, 2020 08 18.
Article in English | MEDLINE | ID: mdl-32459957

ABSTRACT

Complex biotherapeutic modalities, such as antibody-drug conjugates (ADC), present significant challenges for the comprehensive bioanalytical characterization of their pharmacokinetics (PK) and catabolism in both preclinical and clinical settings. Thus, the bioanalytical strategy for ADCs must be designed to address the specific structural elements of the protein scaffold, linker, and warhead. A typical bioanalytical strategy for ADCs involves quantification of the Total ADC, Total IgG, and Free Warhead concentrations. Herein, we present bioanalytical characterization of the PK and catabolism of a novel ADC. MEDI3726 targets prostate-specific membrane antigen (PMSA) and is comprised of a humanized IgG1 antibody site-specifically conjugated to tesirine (SG3249). The MEDI3726 protein scaffold lacks interchain disulfide bonds and has an average drug to antibody ratio (DAR) of 2. Based on the structural characteristics of MEDI3726, an array of 4 bioanalytical assays detecting 6 different surrogate analyte classes representing at least 14 unique species was developed, validated, and employed in support of a first-in-human clinical trial (NCT02991911). MEDI3726 requires the combination of heavy-light chain structure and conjugated warhead to selectively deliver the warhead to the target cells. Therefore, both heavy-light chain dissociation and the deconjugation of the warhead will affect the activity of MEDI3726. The concentration-time profiles of subjects dosed with MEDI3726 revealed catabolism of the protein scaffold manifested by the more rapid clearance of the Active ADC, while exhibiting minimal deconjugation of the pyrrolobenzodiazepine (PBD) warhead (SG3199).


Subject(s)
Antineoplastic Agents/pharmacokinetics , Benzodiazepines/pharmacokinetics , Immunoconjugates/pharmacokinetics , Immunoglobulin G/metabolism , Pyrroles/pharmacokinetics , Antineoplastic Agents/blood , Antineoplastic Agents/metabolism , Benzodiazepines/blood , Benzodiazepines/metabolism , Humans , Immunoconjugates/blood , Immunoconjugates/metabolism , Immunoglobulin G/blood , Prostate-Specific Antigen/immunology , Pyrroles/blood , Pyrroles/metabolism
3.
Nat Chem Biol ; 12(10): 779-86, 2016 10.
Article in English | MEDLINE | ID: mdl-27479743

ABSTRACT

Metabolic reprogramming in tumors represents a potential therapeutic target. Herein we used shRNA depletion and a novel lactate dehydrogenase (LDHA) inhibitor, GNE-140, to probe the role of LDHA in tumor growth in vitro and in vivo. In MIA PaCa-2 human pancreatic cells, LDHA inhibition rapidly affected global metabolism, although cell death only occurred after 2 d of continuous LDHA inhibition. Pancreatic cell lines that utilize oxidative phosphorylation (OXPHOS) rather than glycolysis were inherently resistant to GNE-140, but could be resensitized to GNE-140 with the OXPHOS inhibitor phenformin. Acquired resistance to GNE-140 was driven by activation of the AMPK-mTOR-S6K signaling pathway, which led to increased OXPHOS, and inhibitors targeting this pathway could prevent resistance. Thus, combining an LDHA inhibitor with compounds targeting the mitochondrial or AMPK-S6K signaling axis may not only broaden the clinical utility of LDHA inhibitors beyond glycolytically dependent tumors but also reduce the emergence of resistance to LDHA inhibition.


Subject(s)
Cell Plasticity/drug effects , Enzyme Inhibitors/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , Pyridones/pharmacology , Thiophenes/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , Humans , L-Lactate Dehydrogenase/metabolism , Models, Molecular , Molecular Structure , Pyridones/chemistry , Structure-Activity Relationship , Thiophenes/chemistry
4.
Cytometry B Clin Cytom ; 90(2): 117-27, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26054054

ABSTRACT

Receptor occupancy (RO) assays are designed to quantify the binding of therapeutics to their targets on the cell surface and are frequently used to generate pharmacodynamic (PD) biomarker data in nonclinical and clinical studies of biopharmaceuticals. When combined with the pharmacokinetic (PK) profile, RO data can establish PKPD relationships, which are crucial for informing dose decisions. RO is commonly measured by flow cytometry on fresh blood specimens and is subject to numerous technical and logistical challenges. To ensure that reliable and high quality results are generated from RO assays, careful assay design, key reagent characterization, data normalization/reporting, and thorough planning for implementation are of critical importance during development. In this article, the authors share their experiences and perspectives in these areas and discuss challenges and potential solutions when developing and implementing a flow cytometry-based RO method in support of biopharmaceutical drug development.


Subject(s)
Biomarkers/analysis , Drug Discovery , Flow Cytometry/methods , Humans , Pharmacokinetics
5.
J Biol Chem ; 289(51): 35182-92, 2014 Dec 19.
Article in English | MEDLINE | ID: mdl-25355314

ABSTRACT

Depletion of the central metabolite NAD in cells results in broad metabolic defects leading to cell death and is a proposed novel therapeutic strategy in oncology. There is, however, a limited understanding of the underlying mechanisms that connect disruption of this central metabolite with cell death. Here we utilize GNE-617, a small molecule inhibitor of NAMPT, a rate-limiting enzyme required for NAD generation, to probe the pathways leading to cell death following NAD depletion. In all cell lines examined, NAD was rapidly depleted (average t½ of 8.1 h) following NAMPT inhibition. Concurrent with NAD depletion, there was a decrease in both cell proliferation and motility, which we attribute to reduced activity of NAD-dependent deacetylases because cells fail to deacetylate α-tubulin-K40 and histone H3-K9. Following depletion of NAD by >95%, cells lose the ability to regenerate ATP. Cell lines with a slower rate of ATP depletion (average t½ of 45 h) activate caspase-3 and show evidence of apoptosis and autophagy, whereas cell lines with rapid depletion ATP (average t½ of 32 h) do not activate caspase-3 or show signs of apoptosis or autophagy. However, the predominant form of cell death in all lines is oncosis, which is driven by the loss of plasma membrane homeostasis once ATP levels are depleted by >20-fold. Thus, our work illustrates the sequence of events that occurs in cells following depletion of a key metabolite and reveals that cell death caused by a loss of NAD is primarily driven by the inability of cells to regenerate ATP.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Heterocyclic Compounds, 2-Ring/pharmacology , NAD/metabolism , Sulfones/pharmacology , Acetylation/drug effects , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Autophagy/drug effects , Blotting, Western , Caspase 3/metabolism , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , HCT116 Cells , Heterocyclic Compounds, 2-Ring/chemistry , Histones/metabolism , Humans , Microscopy, Electron, Transmission , Molecular Structure , Nicotinamide Phosphoribosyltransferase/antagonists & inhibitors , Nicotinamide Phosphoribosyltransferase/metabolism , Sulfones/chemistry , Time Factors , Tubulin/metabolism
6.
Cell Cycle ; 13(2): 303-14, 2014.
Article in English | MEDLINE | ID: mdl-24247149

ABSTRACT

Activation of Checkpoint kinase 1 (Chk1) following DNA damage mediates cell cycle arrest to prevent cells with damaged DNA from entering mitosis. Here we provide a high-resolution analysis of cells as they undergo S- and G2-checkpoint bypass in response to Chk1 inhibition with the selective Chk1 inhibitor GNE-783. Within 4-8 h of Chk1 inhibition following gemcitabine induced DNA damage, cells with both sub-4N and 4N DNA content prematurely enter mitosis. Coincident with premature transition into mitosis, levels of DNA damage dramatically increase and chromosomes condense and attempt to align along the metaphase plate. Despite an attempt to congress at the metaphase plate, chromosomes rapidly fragment and lose connection to the spindle microtubules. Gemcitabine mediated DNA damage promotes the formation of Rad51 foci; however, while Chk1 inhibition does not disrupt Rad51 foci that are formed in response to gemcitabine, these foci are lost as cells progress into mitosis. Premature entry into mitosis requires the Aurora, Cdk1/2 and Plk1 kinases and even though caspase-2 and -3 are activated upon mitotic exit, they are not required for cell death. Interestingly, p53, but not p21, deficiency enables checkpoint bypass and chemo-potentiation. Finally, we uncover a differential role for the Wee-1 checkpoint kinase in response to DNA damage, as Wee-1, but not Chk1, plays a more prominent role in the maintenance of S- and G2-checkpoints in p53 proficient cells.


Subject(s)
Caspases/metabolism , Chromosomes, Human/genetics , DNA Fragmentation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Carbolines/pharmacology , Cell Cycle Proteins/metabolism , Cell Death/drug effects , Cell Line, Tumor , Checkpoint Kinase 1 , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Enzyme Activation , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Mitosis/drug effects , Nuclear Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Rad51 Recombinase/metabolism , S Phase Cell Cycle Checkpoints/drug effects , Tumor Suppressor Protein p53/metabolism , Gemcitabine
7.
PLoS One ; 8(11): e80533, 2013.
Article in English | MEDLINE | ID: mdl-24265828

ABSTRACT

The CD20-specific monoclonal antibody rituximab (MabThera(®), Rituxan(®)) is widely used as the backbone of treatment for patients with hematologic disorders. Intravenous administration of rituximab is associated with infusion times of 4-6 hours, and can be associated with infusion-related reactions. Subcutaneous administration of rituximab may reduce this and facilitate administration without infusion-related reactions. We sought to determine the feasibility of achieving equivalent efficacy (measured by endogenous B-cell depletion) and long-term durability of CD20 target coverage for subcutaneously administered rituximab compared with intravenous dosing. In these preclinical studies, male cynomolgus monkeys were treated with either intravenous rituximab or novel subcutaneous formulation of rituximab containing human recombinant DNA-derived hyaluronidase enzyme. Peripheral blood samples were analyzed for serum rituximab concentrations, peripheral B-cell depletion, and CD20 target coverage, including subset analysis according to CD21+ status. Distal lymph node B-cell depletion and CD20 target coverage were also measured. Initial peak serum concentrations of rituximab were significantly higher following intravenous administration than subcutaneous. However, the mean serum rituximab trough concentrations were comparable at 2 and 7 days post-first dose and 9 and 14 days post-second dose. Efficacy of B-cell depletion in both peripheral blood and distal lymph nodes was comparable for both methods. In lymph nodes, 9 days after the second dose with subcutaneous and intravenous rituximab, B-cell levels were decreased by 57% and 42% respectively. Similarly, levels of peripheral blood B cells were depleted by >94% for both subcutaneous and intravenous dosing at all time points. Long-term recovery of free unbound surface CD20 levels was similar, and the duration of B-cell depletion was equally sustained over 2 months for both methods. These results demonstrate that, despite initial peak serum drug level differences, subcutaneous rituximab has similar durability, pharmacodynamics, and efficacy compared with intravenous rituximab.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antineoplastic Agents/administration & dosage , Administration, Intravenous , Animals , Antibodies, Monoclonal, Murine-Derived/pharmacokinetics , Antigens, CD20/metabolism , Antineoplastic Agents/pharmacokinetics , B-Lymphocyte Subsets/metabolism , Immunophenotyping , Infusions, Subcutaneous , Leukocytes, Mononuclear , Lymph Nodes/cytology , Lymphocyte Depletion , Macaca fascicularis , Male , Receptors, Complement 3d/metabolism , Rituximab
8.
Mol Cancer Ther ; 12(11): 2285-95, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24038068

ABSTRACT

Here we report that GNE-783, a novel checkpoint kinase-1 (CHK1) inhibitor, enhances the activity of gemcitabine by disabling the S- and G2 cell-cycle checkpoints following DNA damage. Using a focused library of 51 DNA-damaging agents, we undertook a systematic screen using three different cell lines to determine which chemotherapeutics have their activity enhanced when combined with GNE-783. We found that GNE-783 was most effective at enhancing activity of antimetabolite-based DNA-damaging agents; however, there was a surprisingly wide range of activity within each class of agents. We, next, selected six different therapeutic agents and screened these in combination with GNE-783 across a panel of cell lines. This revealed a preference for enhanced chemopotentiation of select agents within tumor types, as, for instance, GNE-783 preferentially enhanced the activity of temozolomide only in melanoma cell lines. Additionally, although p53 mutant status was important for the overall response to combinations with some agents; our data indicate that this alone was insufficient to predict synergy. We finally compared the ability of a structurally related CHK1 inhibitor, GNE-900, to enhance the in vivo activity of gemcitabine, CPT-11, and temozolomide in xenograft models. GNE-900 significantly enhanced activity of only gemcitabine in vivo, suggesting that strong chemopotentiation in vitro can translate into chemopotentiation in vivo. In conclusion, our results show that selection of an appropriate agent to combine with a CHK1 inhibitor needs to be carefully evaluated in the context of the genetic background and tumor type in which it will be used.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Protein Kinases/metabolism , Animals , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Carbolines/pharmacology , Cell Cycle Checkpoints/drug effects , Checkpoint Kinase 1 , DNA Damage/drug effects , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , HCT116 Cells , HT29 Cells , High-Throughput Nucleotide Sequencing , Humans , Irinotecan , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/pathology , Neoplasms, Experimental , Protein Kinases/genetics , Pyridines/pharmacology , Pyrroles/pharmacology , Temozolomide , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays , Gemcitabine
9.
Blood ; 115(22): 4393-402, 2010 Jun 03.
Article in English | MEDLINE | ID: mdl-20194898

ABSTRACT

CD20 is an important target for the treatment of B-cell malignancies, including non-Hodgkin lymphoma as well as autoimmune disorders. B-cell depletion therapy using monoclonal antibodies against CD20, such as rituximab, has revolutionized the treatment of these disorders, greatly improving overall survival in patients. Here, we report the development of GA101 as the first Fc-engineered, type II humanized IgG1 antibody against CD20. Relative to rituximab, GA101 has increased direct and immune effector cell-mediated cytotoxicity and exhibits superior activity in cellular assays and whole blood B-cell depletion assays. In human lymphoma xenograft models, GA101 exhibits superior antitumor activity, resulting in the induction of complete tumor remission and increased overall survival. In nonhuman primates, GA101 demonstrates superior B cell-depleting activity in lymphoid tissue, including in lymph nodes and spleen. Taken together, these results provide compelling evidence for the development of GA101 as a promising new therapy for the treatment of B-cell disorders.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/pharmacology , Antigens, CD20/immunology , B-Lymphocytes/immunology , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal, Humanized , Antibodies, Monoclonal, Murine-Derived , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Humans , Immunity, Cellular , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Variable Region/genetics , In Vitro Techniques , Lymphocyte Depletion/methods , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/therapy , Macaca fascicularis , Mice , Mice, SCID , Neoplasm Transplantation , Protein Engineering , Receptors, IgG/immunology , Rituximab , Transplantation, Heterologous
10.
Immunity ; 31(5): 749-60, 2009 Nov 20.
Article in English | MEDLINE | ID: mdl-19896393

ABSTRACT

Anergy is a critical physiologic mechanism to sensor self-reactive B cells. However, a biochemical understanding of how anergy is achieved and maintained is lacking. Herein, we investigated the role of the phosphoinositide 3-kinase (PI3K) lipid product PI(3,4,5)P(3) in B cell anergy. We found reduced generation of PI(3,4,5)P(3) in anergic B cells, which was attributable to reduced phosphorylation of the PI3K membrane adaptor CD19, as well as increased expression of the inositol phosphatase PTEN. Sustained production of PI(3,4,5)P(3) in B cells, achieved through conditional deletion of Pten, resulted in failed tolerance induction and abundant autoantibody production. In contrast to wild-type immature B cells, B cell receptor engagement of PTEN-deficient immature B cells resulted in activation and proliferation, indicating a central defect in early B cell responsiveness. These findings establish repression of the PI3K signaling pathway as a necessary condition to avert the generation, activation, and persistence of self-reactive B cells.


Subject(s)
B-Lymphocytes/immunology , Clonal Anergy , Phosphatidylinositol Phosphates/metabolism , Animals , Antigens, CD19/metabolism , Glycosylphosphatidylinositols/metabolism , Mice , Mice, Knockout , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Signal Transduction
11.
J Immunol ; 175(8): 5379-89, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16210644

ABSTRACT

Complement C3 cleavage products mediate the recognition and clearance of toxic or infectious agents. In addition, binding of the C3d fragment to Ag promotes B lymphocyte activation through coengagment of the BCR and complement receptor 2 (CD21). Signal augmentation is thought to be achieved through enhanced recruitment and activation of CD21-associated CD19. In this study we show, using the DBA/1 collagen-induced arthritis (CIA) model, that conjugation of C3d to heterologous type II collagen is sufficient to cause disease in the absence of the mycobacterial components of CFA. Transient depletion of C3 during the inductive phase of CIA delays and lessens the severity of disease, and DBA/1 mice deficient for coreceptor components CD19 or CD21 are not susceptible to CIA. Adoptive transfer experiments revealed that CD21 expression on either B cells or follicular dendritic cells is sufficient to acquire disease susceptibility. Although CD19(-/-) and CD21(-/-) mice produce primary Ab responses to heterologous and autologous type II collagen, they are impaired in the ability to activate T cells, form germinal centers, and produce secondary autoantibody responses. These findings indicate that binding of C3d to self-Ags can promote autoimmunity through enhanced Ag retention and presentation by follicular dendritic cells and B cells, respectively.


Subject(s)
Antigens, CD19/physiology , Arthritis, Experimental/immunology , Complement C3d/physiology , Receptors, Complement 3d/physiology , Animals , Antigens, CD19/genetics , Antigens, CD19/metabolism , Arthritis, Experimental/metabolism , Cattle , Cells, Cultured , Collagen Type II/immunology , Complement C3d/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred DBA , Mice, Knockout , Receptors, Complement 3d/genetics , Receptors, Complement 3d/metabolism , T-Lymphocytes/immunology
12.
Immunol Res ; 31(2): 119-31, 2005.
Article in English | MEDLINE | ID: mdl-15778510

ABSTRACT

Although the B-cell antigen receptor (BCR) factors most prominently in the maintenance and differentiation of mature B cells, it is now appreciated that co-receptor molecules can positively or negatively modulate signals through the BCR. Co-receptors are functionally defined as modifiers of BCR engagement and signal transduction, and are distinct from other accessory molecules that act independently to regulate B-cell growth. The co-receptor CD19 functions to augment signals by the pre-BCR/BCR and in doing so can modulate B-cell fate decisions at multiple stages of development. In mature B cells, CD19 also associates with complement receptor 2 (CR2/CD21) and is pivotal for transducing signals induced by co-recognition of complement C3d-fixed antigens by the BCR and CD21. In this article, we focus on recent progress in the understanding of CD19 function through the characterization of mouse models that relate in vivo function to biochemical properties of CD19.


Subject(s)
Antigens, CD19/physiology , B-Lymphocyte Subsets/cytology , B-Lymphocyte Subsets/immunology , Cell Differentiation/physiology , Animals , B-Lymphocyte Subsets/metabolism , Cell Differentiation/immunology , Humans , Receptors, Antigen, B-Cell/physiology , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...