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










Publication year range
1.
JCI Insight ; 8(21)2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37788104

ABSTRACT

MTORC1 integrates signaling from the immune microenvironment to regulate T cell activation, differentiation, and function. TSC2 in the tuberous sclerosis complex tightly regulates mTORC1 activation. CD8+ T cells lacking TSC2 have constitutively enhanced mTORC1 activity and generate robust effector T cells; however, sustained mTORC1 activation prevents generation of long-lived memory CD8+ T cells. Here we show that manipulating TSC2 at Ser1365 potently regulated activated but not basal mTORC1 signaling in CD8+ T cells. Unlike nonstimulated TSC2-KO cells, CD8+ T cells expressing a phosphosilencing mutant TSC2-S1365A (TSC2-SA) retained normal basal mTORC1 activity. PKC and T cell receptor (TCR) stimulation induced TSC2 S1365 phosphorylation, and preventing this with the SA mutation markedly increased mTORC1 activation and T cell effector function. Consequently, SA CD8+ T cells displayed greater effector responses while retaining their capacity to become long-lived memory T cells. SA CD8+ T cells also displayed enhanced effector function under hypoxic and acidic conditions. In murine and human solid-tumor models, SA CD8+ T cells used as adoptive cell therapy displayed greater antitumor immunity than WT CD8+ T cells. These findings reveal an upstream mechanism to regulate mTORC1 activity in T cells. The TSC2-SA mutation enhanced both T cell effector function and long-term persistence/memory formation, supporting an approach to engineer better CAR-T cells for treating cancer.


Subject(s)
Tuberous Sclerosis , Mice , Humans , Animals , Mechanistic Target of Rapamycin Complex 1 , CD8-Positive T-Lymphocytes , Mutation , Cell Differentiation , Tumor Microenvironment
2.
Mol Cancer Ther ; 21(8): 1326-1336, 2022 08 02.
Article in English | MEDLINE | ID: mdl-35666803

ABSTRACT

Probody therapeutics (Pb-Txs) are conditionally activated antibody-drug conjugates (ADCs) designed to remain inactive until proteolytically activated in the tumor microenvironment, enabling safer targeting of antigens expressed in both tumor and normal tissue. Previous attempts to target CD71, a highly expressed tumor antigen, have failed to establish an acceptable therapeutic window due to widespread normal tissue expression. This study evaluated whether a probody-drug conjugate targeting CD71 can demonstrate a favorable efficacy and tolerability profile in preclinical studies for the treatment of cancer. CX-2029, a Pb-Tx conjugated to maleimido-caproyl-valine-citrulline-p-aminobenzyloxycarbonyl-monomethyl auristatin E, was developed as a novel cancer therapeutic targeting CD71. Preclinical studies were performed to evaluate the efficacy and safety of this anti-CD71 PDC in patient-derived xenograft (PDX) mouse models and cynomolgus monkeys, respectively. CD71 expression was detected at high levels by IHC across a broad range of tumor and normal tissues. In vitro, the masked Pb-Tx form of the anti-CD71 PDC displayed a >50-fold reduced affinity for binding to CD71 on cells compared with protease-activated, unmasked anti-CD71 PDC. Potent in vivo tumor growth inhibition (stasis or regression) was observed in >80% of PDX models (28/34) at 3 or 6 mg/kg. Anti-CD71 PDC remained mostly masked (>80%) in circulation throughout dosing in cynomolgus monkeys at 2, 6, and 12 mg/kg and displayed a 10-fold improvement in tolerability compared with an anti-CD71 ADC, which was lethal. Preclinically, anti-CD71 PDC exhibits a highly efficacious and acceptable safety profile that demonstrates the utility of the Pb-Tx platform to target CD71, an otherwise undruggable target. These data support further clinical development of the anti-CD71 PDC CX-2029 as a novel cancer therapeutic.


Subject(s)
Antineoplastic Agents , Immunoconjugates , Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Humans , Immunoconjugates/pharmacology , Immunoconjugates/therapeutic use , Lead , Macaca fascicularis/metabolism , Mice , Neoplasms/drug therapy , Tumor Microenvironment , Xenograft Model Antitumor Assays
4.
Cancer Immunol Res ; 9(12): 1451-1464, 2021 12.
Article in English | MEDLINE | ID: mdl-34635485

ABSTRACT

Immune-checkpoint blockade has revolutionized cancer treatment. However, most patients do not respond to single-agent therapy. Combining checkpoint inhibitors with other immune-stimulating agents increases both efficacy and toxicity due to systemic T-cell activation. Protease-activatable antibody prodrugs, known as Probody therapeutics (Pb-Tx), localize antibody activity by attenuating capacity to bind antigen until protease activation in the tumor microenvironment. Herein, we show that systemic administration of anti-programmed cell death ligand 1 (anti-PD-L1) and anti-programmed cell death protein 1 (anti-PD-1) Pb-Tx to tumor-bearing mice elicited antitumor activity similar to that of traditional PD-1/PD-L1-targeted antibodies. Pb-Tx exhibited reduced systemic activity and an improved nonclinical safety profile, with markedly reduced target occupancy on peripheral T cells and reduced incidence of early-onset autoimmune diabetes in nonobese diabetic mice. Our results confirm that localized PD-1/PD-L1 inhibition by Pb-Tx can elicit robust antitumor immunity and minimize systemic immune-mediated toxicity. These data provide further preclinical rationale to support the ongoing development of the anti-PD-L1 Pb-Tx CX-072, which is currently in clinical trials.


Subject(s)
Antibodies, Monoclonal/therapeutic use , B7-H1 Antigen/therapeutic use , Immunotherapy/methods , Amino Acid Sequence , Animals , Antibodies, Monoclonal/pharmacology , B7-H1 Antigen/pharmacology , Cell Line, Tumor , Disease Models, Animal , Humans , Mice , Tumor Microenvironment
5.
J Immunother Cancer ; 7(1): 304, 2019 11 14.
Article in English | MEDLINE | ID: mdl-31727131

ABSTRACT

Despite remarkable success in the treatment of hematological malignancies, CAR T-cell therapies for solid tumors have floundered, in large part due to local immune suppression and the effects of prolonged stimulation leading to T-cell dysfunction and exhaustion. One mechanism by which gliomas and other cancers can hamper CAR T cells is through surface expression of inhibitory ligands such as programmed cell death ligand 1 (PD-L1). Using the CRIPSR-Cas9 system, we created universal CAR T cells resistant to PD-1 inhibition through multiplexed gene disruption of endogenous T-cell receptor (TRAC), beta-2 microglobulin (B2M) and PD-1 (PDCD1). Triple gene-edited CAR T cells demonstrated enhanced activity in preclinical glioma models. Prolonged survival in mice bearing intracranial tumors was achieved after intracerebral, but not intravenous administration. CRISPR-Cas9 gene-editing not only provides a potential source of allogeneic, universal donor cells, but also enables simultaneous disruption of checkpoint signaling that otherwise impedes maximal antitumor functionality.


Subject(s)
Brain Neoplasms/therapy , ErbB Receptors , Glioblastoma/therapy , Immunotherapy, Adoptive , Programmed Cell Death 1 Receptor/genetics , Animals , Brain Neoplasms/immunology , CRISPR-Cas Systems , Cell Line, Tumor , Glioblastoma/immunology , Humans , Mice , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
6.
CPT Pharmacometrics Syst Pharmacol ; 8(9): 676-684, 2019 09.
Article in English | MEDLINE | ID: mdl-31250966

ABSTRACT

PROBODY therapeutics (Pb-Tx) are protease-activatable prodrugs of monoclonal antibodies (mAbs) designed to target tumors where protease activity is elevated while avoiding normal tissue. They are composed of a parental mAb, a mask that inhibits antibody binding to target, and a protease-cleavable substrate between the mask and the mAb. We report a quantitative systems pharmacology model for the rational design and clinical translation of Pb-Tx. The model adequately described monkey pharmacokinetic data following the administration of six anti-CD166 Pb-Tx of varying mask strength and substrate cleavability and captured the trend of decreasing Pb-Tx systemic clearance with increasing mask strength. Projections to humans suggested both higher levels of Pb-Tx in tumor relative to parental mAb and an optimal mask strength for maximizing tumor receptor-mediated uptake. Simulations further suggested the majority of circulating species in humans would be intact/masked Pb-Tx, with no significant flux of cleaved/activated species from tumor to the systemic compartment.


Subject(s)
Antineoplastic Agents, Immunological/pharmacokinetics , Neoplasms/drug therapy , Prodrugs/pharmacokinetics , Animals , Antineoplastic Agents, Immunological/chemistry , Cell Line, Tumor , Humans , Macaca fascicularis , Mice , Models, Biological , Prodrugs/chemistry , Systems Biology , Tissue Distribution , Xenograft Model Antitumor Assays
7.
Clin Cancer Res ; 22(1): 175-86, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26350262

ABSTRACT

PURPOSE: Breast cancer is the second leading cause of cancer mortality among women worldwide. The major problem with current treatments is tumor resistance, recurrence, and disease progression. ErbB-2-positive breast tumors are aggressive and frequently become resistant to trastuzumab or lapatinib. We showed previously that Notch-1 is required for trastuzumab resistance in ErbB-2-positive breast cancer. EXPERIMENTAL DESIGN: Here, we sought to elucidate mechanisms by which ErbB-2 attenuates Notch signaling and how this is reversed by trastuzumab or lapatinib. RESULTS: The current study elucidates a novel Notch inhibitory mechanism by which PKCα downstream of ErbB-2 (i) restricts the availability of Jagged-1 at the cell surface to transactivate Notch, (ii) restricts the critical interaction between Jagged-1 and Mindbomb-1, an E3 ligase that is required for Jagged-1 ubiquitinylation and subsequent Notch activation, (iii) reverses trastuzumab resistance in vivo, and (iv) predicts better outcome in women with ErbB-2-positive breast cancer. CONCLUSIONS: The clinical impact of these studies is PKCα is potentially a good prognostic marker for low Notch activity and increased trastuzumab sensitivity in ErbB-2-positive breast cancer. Moreover, women with ErbB-2-positive breast tumors expressing high Notch activation and low PKCα expression could be the best candidates for anti-Notch therapy.


Subject(s)
Breast Neoplasms/metabolism , Calcium-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Protein Kinase C-alpha/metabolism , Receptor, ErbB-2/metabolism , Receptors, Notch/metabolism , Signal Transduction/drug effects , Trastuzumab/pharmacology , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Models, Animal , Female , Humans , Jagged-1 Protein , Neoplasm Recurrence, Local , Prognosis , Protein Binding , Protein Transport , Serrate-Jagged Proteins , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Xenograft Model Antitumor Assays
8.
Sci Transl Med ; 5(207): 207ra144, 2013 Oct 16.
Article in English | MEDLINE | ID: mdl-24132639

ABSTRACT

Target-mediated toxicity constitutes a major limitation for the development of therapeutic antibodies. To redirect the activity of antibodies recognizing widely distributed targets to the site of disease, we have applied a prodrug strategy to create an epidermal growth factor receptor (EGFR)-directed Probody therapeutic-an antibody that remains masked against antigen binding until activated locally by proteases commonly active in the tumor microenvironment. In vitro, the masked Probody showed diminished antigen binding and cell-based activities, but when activated by appropriate proteases, it regained full activity compared to the parental anti-EGFR antibody cetuximab. In vivo, the Probody was largely inert in the systemic circulation of mice, but was activated within tumor tissue and showed antitumor efficacy that was similar to that of cetuximab. The Probody demonstrated markedly improved safety and increased half-life in nonhuman primates, enabling it to be dosed safely at much higher levels than cetuximab. In addition, we found that both Probody-responsive xenograft tumors and primary tumor samples from patients were capable of activating the Probody ex vivo. Probodies may therefore improve the safety profile of therapeutic antibodies without compromising efficacy of the parental antibody and may enable the wider use of empowered antibody formats such as antibody-drug conjugates and bispecifics.


Subject(s)
Antibodies, Neoplasm/therapeutic use , ErbB Receptors/metabolism , Neoplasms/drug therapy , Neoplasms/pathology , Prodrugs/therapeutic use , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Neoplasm/pharmacology , Cell Proliferation/drug effects , Cetuximab , Humans , Immunohistochemistry , Macaca fascicularis , Mice , Mice, Nude , Prodrugs/toxicity , Skin/drug effects , Skin/pathology , Xenograft Model Antitumor Assays
9.
Food Microbiol ; 34(2): 319-27, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23541198

ABSTRACT

Antimicrobial resistance (AMR), co-selection phenomenon, and the relationship between reduced susceptibility (RSC) to ciprofloxacin (CIP) and resistance to other antimicrobials in Listeria spp. (n = 103) recovered from food processing environments (FPE) and food were investigated. Resistance of Listeria monocytogenes and other listeriae, respectively, to cefoxitin (FOX; 98% vs. 88%), CIP (7% vs. 4%), clindamycin (CLI; 33% vs. 59%) and tetracycline (6% vs. 8%) was observed, as was RSC to CIP (67% vs. 57%) and CLI (65% vs. 41%). L. monocytogenes also possessed RSC to linezolid (LZD; 6%), rifampicin (2%) and streptomycin (6%), with other listeriae displaying RSC to chloramphenicol (4%). L. monocytogenes serotype 1/2a (90%) isolates were more frequently resistant or possessed RSC to CIP compared to serotype 4b (55%) (p = 0.015). When eight strains were experimentally adapted to high concentrations of CIP, co-selection occurred as MICs to benzalkonium chloride (BAC) increased (n = 5), gentamicin MICs remained the same (n = 6) or increased 2-fold (n = 2), and led to RSC to LZD (n = 1) and resistance to CLI (n = 8). Overall, levels of resistance/RSC to CIP in food chain isolates, particularly 1/2a, are concerning. Further, reduced sensitivity to disparate antimicrobials following CIP exposure highlights the need for increased knowledge of co-selection phenomenon linked with antimicrobial agents.


Subject(s)
Anti-Bacterial Agents/pharmacology , Dairy Products/microbiology , Drug Resistance, Multiple, Bacterial , Listeria/drug effects , Seafood/microbiology , Animals , Cattle , Fishes , Food Contamination/analysis , Food Handling , Listeria/classification , Listeria/genetics , Listeria/isolation & purification
10.
J Biol Chem ; 284(35): 23344-52, 2009 Aug 28.
Article in English | MEDLINE | ID: mdl-19584055

ABSTRACT

The 3,4-dihydroxyphenyl-l-alanine (Dopa)-containing proteins of mussel byssus play a critical role in wet adhesion and have inspired versatile new synthetic strategies for adhesives and coatings. Apparently, however, not all mussel adhesive proteins are beholden to Dopa chemistry. The cDNA-deduced sequence of Pvfp-1, a highly aromatic and redox active byssal coating protein in the green mussel Perna viridis, suggests that Dopa may be replaced by a post-translational modification of tryptophan. The N-terminal tryptophan-rich domain of Pvfp-1 contains 42 decapeptide repeats with the consensus sequences ATPKPW(1)TAW(2)K and APPPAW(1)TAW(2)K. A small collagen domain (18 Gly-X-Y repeats) is also present. Tandem mass spectrometry of isolated tryptic decapeptides has detected both C(2)-hexosylated tryptophan (W(1)) and C(2)-hexosylated hydroxytryptophan (W(2)), the latter of which is redox active. The UV absorbance spectrum of W(2) is consistent with 7-hydroxytryptophan, which represents an intriguing new theme for bioinspired opportunistic wet adhesion.


Subject(s)
5-Hydroxytryptophan/metabolism , Perna/metabolism , Proteins/chemistry , Proteins/metabolism , Amino Acid Sequence , Animals , Consensus Sequence , Glycosylation , Molecular Sequence Data , Perna/chemistry , Perna/genetics , Protein Structure, Tertiary , Proteins/genetics , Sequence Alignment
11.
J Exp Biol ; 212(Pt 14): 2224-36, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19561212

ABSTRACT

The marine mussel Mytilus galloprovincialis is tethered to rocks in the intertidal zone by a holdfast known as the byssus. Functioning as a shock absorber, the byssus is composed of threads, the primary molecular components of which are collagen-containing proteins (preCOLs) that largely dictate the higher order self-assembly and mechanical properties of byssal threads. The threads contain additional matrix components that separate and perhaps lubricate the collagenous microfibrils during deformation in tension. In this study, the thread matrix proteins (TMPs), a glycine-, tyrosine- and asparagine-rich protein family, were shown to possess unique repeated sequence motifs, significant transcriptional heterogeneity and were distributed throughout the byssal thread. Deamidation was shown to occur at a significant rate in a recombinant TMP and in the byssal thread as a function of time. Furthermore, charge heterogeneity presumably due to deamidation was observed in TMPs extracted from threads. The TMPs were localized to the preCOL-containing secretory granules in the collagen gland of the foot and are assumed to provide a viscoelastic matrix around the collagenous fibers in byssal threads.


Subject(s)
Extracellular Matrix Proteins/chemistry , Mytilus/metabolism , Amino Acid Sequence , Animals , Base Sequence , Collagen/metabolism , Collagen/physiology , DNA, Complementary/chemistry , Extracellular Matrix Proteins/metabolism , Extracellular Matrix Proteins/physiology , Molecular Sequence Data , Multigene Family , Recombinant Fusion Proteins/metabolism , Repetitive Sequences, Amino Acid
12.
Biomacromolecules ; 5(4): 1351-5, 2004.
Article in English | MEDLINE | ID: mdl-15244450

ABSTRACT

In marine mussels (Mytilus), byssal threads are made in minutes from prefabricated smectic polymer liquid crystals by a process resembling reaction injection molding. The mesogens in these arrays are known to be natural block copolymers with rodlike collagen cores. Using atomic force microscopy, it was shown that these collagenous mesogens are bent-core or banana-shaped in a manner that is consistent with and predictable from their amino acid sequence. The overall bend angle in preCOL-NG in Mytilus galloprovincialis is about 130 degrees. The mesogens have a center-to-center separation of approximately 22 nm and a length of 200 nm. It is evident that the smectic structure of the prefabricated mesophases remains largely intact over 1-3 microm distances in the molded fibers and is presumably locked in place during molding by cross-linking. Like the smectic liquid crystals of many synthetic banana mesogens, the collagenous mesogens of the byssal threads exhibit SmC(2) symmetry with a characteristic tilt of 24.6 degrees. At about 100% extension, this tilt is considerably reduced and the globular end domains are no longer visible presumably because they have been unraveled.


Subject(s)
Biopolymers/chemistry , Bivalvia/chemistry , Collagen/chemistry , Animals , Bivalvia/ultrastructure , Collagen/analysis , Collagen/ultrastructure , Microscopy, Atomic Force/methods , Species Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...