Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Biotechnol Bioeng ; 117(12): 3664-3676, 2020 12.
Article in English | MEDLINE | ID: mdl-32716053

ABSTRACT

Matrix metalloproteinase-12 (MMP-12), also known as macrophage elastase, is a potent inflammatory mediator and therefore an important pharmacological target. Clinical trial failures of broad-spectrum compound MMP inhibitors suggested that specificity is the key for a successful therapy. To provide the required selectivity, monoclonal antibody (mAb)-based inhibitors are on the rise. However, poor production of active recombinant human MMP-12 catalytic domain (cdMMP-12) presented a technical hurdle for its inhibitory mAb development. We hypothesized that this problem could be solved by designing an expression-optimized cdMMP-12 mutant without structural disruptions at its reaction cleft and surrounding area, and thus isolated active-site inhibitory mAbs could maintain their binding and inhibition functions toward wild-type MMP-12. We combined three advances in the field-PROSS algorithm for cdMMP-12 mutant design, convex paratope antibody library construction, and functional selection for inhibitory mAbs. As a result, isolated Fab inhibitors showed nanomolar affinity and potency toward cdMMP-12 with high selectivity and high proteolytic stability. Particularly, Fab LH11 targeted the reaction cleft of wild-type cdMMP-12 with 75 nM binding KD and 23 nM inhibition IC50 . We expect that our methods can promote the development of mAbs inhibiting important proteases, many of which are recalcitrant to functional production.


Subject(s)
Antibodies, Monoclonal/chemistry , Matrix Metalloproteinase 12/chemistry , Matrix Metalloproteinase Inhibitors/chemistry , Antibodies, Monoclonal/genetics , Humans , Matrix Metalloproteinase 12/genetics , Protein Domains
2.
Hum Gene Ther ; 34(21-22): 1145-1161, 2023 Nov.
Article in English | MEDLINE | ID: mdl-36851890

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy has transformed the treatment of hematological malignancies but has yet to achieve similar success in solid tumors due to a lack of persistence and function in the tumor microenvironment. We previously reported the augmentation of CAR T cell therapy in an engineered solid tumor model through the secretion of anti-PD-1 single-chain fragment variable region (scFv), as shown by enhanced CAR T cell antitumor efficacy, expansion, and vitality. We have since improved the platform to create a superior cellular product-CAR T cells secreting single-chain trimeric 4-1BB ligand fused to anti-PD-1 scFv (αPD1-41BBL). 4-1BB signaling promotes cytotoxic T lymphocyte proliferation and survival but targeting 4-1BB with agonist antibodies in the clinic has been hindered by low antitumor activity and high toxicity. CAR T cells using 4-1BB endodomain for costimulatory signals have demonstrated milder antitumor response and longer persistence compared to CAR T cells costimulated by CD28 endodomain. We have, for the first time, engineered CD28-costimulated CAR T cells to secrete a fusion protein containing the soluble trimeric 4-1BB ligand. In vitro and in vivo, CAR19.αPD1-41BBL T cells exhibited reduced inhibitory receptor upregulation, enhanced persistence and proliferation, and a less differentiated memory status compared to CAR T cells without additional 4-1BB:4-1BBL costimulation. Accordingly, CAR19.αPD1-41BBL T cell-treated mice displayed significantly improved tumor growth control and overall survival. Spurred on by our preclinical success targeting CD19 as a model antigen, we produced mesothelin-targeting CAR T cells and confirmed the enhanced solid tumor efficacy of αPD1-41BBL-secreting CAR T cells.


Subject(s)
Neoplasms , Receptors, Chimeric Antigen , Animals , Mice , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell , CD28 Antigens , 4-1BB Ligand/metabolism , Neoplasms/therapy , Immunotherapy, Adoptive , Tumor Microenvironment
3.
Hum Gene Ther ; 33(5-6): 223-236, 2022 03.
Article in English | MEDLINE | ID: mdl-34225478

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy mediates unprecedented benefit in certain leukemias and lymphomas, but has yet to achieve similar success in combating solid tumors. A substantial body of work indicates that the accumulation of adenosine in the solid tumor microenvironment (TME) plays a crucial role in abrogating immunotherapies. Adenosine deaminase 1 (ADA) catabolizes adenosine into inosine and is indispensable for a functional immune system. We have, for the first time, engineered CAR T cells to overexpress ADA. To potentially improve the pharmacokinetic profile of ADA, we have modified the overexpressed ADA in two ways, through the incorporation of a (1) albumin-binding domain or (2) collagen-binding domain. ADA and modified ADA were successfully expressed by CAR T cells and augmented CAR T cell exhaustion resistance. In a preclinical engineered ovarian carcinoma xenograft model, ADA and collagen-binding ADA overexpression significantly enhanced CAR T cell expansion, tumor tissue infiltration, tumor growth control, and overall survival, whereas albumin-binding ADA overexpression did not. Furthermore, in a syngeneic colon cancer solid tumor model, the overexpression of mouse ADA by cancer cells significantly reduced tumor burden and remodeled the TME to favor antitumor immunity. The overexpression of ADA for enhanced cell therapy is a safe, straightforward, reproducible genetic modification that can be utilized in current CAR T cell constructs to result in an armored CAR T product with superior therapeutic potential.


Subject(s)
Receptors, Chimeric Antigen , Adenosine/metabolism , Adenosine Deaminase/genetics , Albumins/metabolism , Animals , Cell Line, Tumor , Collagen/metabolism , Humans , Immunotherapy, Adoptive , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes , Xenograft Model Antitumor Assays
4.
Mol Ther Oncolytics ; 21: 144-157, 2021 Jun 25.
Article in English | MEDLINE | ID: mdl-33981830

ABSTRACT

Despite the remarkable success of chimeric antigen receptor-modified T (CAR-T) cell therapy for blood malignancies, the clinical efficacy of this novel therapy in solid tumor treatment is largely limited by the immunosuppressive tumor microenvironment (TME). For instance, immune checkpoints (e.g., programmed cell death protein 1 [PD-1]/programmed death ligand 1 [PD-L1]) in TME play an important role in inhibiting T cell proliferation and functions. Transforming growth factor ß (TGF)-ß secreted by cancer cells in TME induces regulatory T cells (Tregs) and inhibits cytotoxic T cells. To overcome the inhibitory effect of immune checkpoints, we have previously engineered CAR-T cells to secrete anti-PD-1 to block the PD-1/PD-L1 pathway activity, a step demonstrating superior antitumor efficacy compared with conventional CAR-T cells. In this study, we engineered CAR-T cells that secrete bispecific trap protein co-targeting PD-1 and TGF-ß, with the aim of further improving antitumor immunity. Compared with conventional CAR-T cells and anti-PD-1-secreting CAR-T cells, data from in vitro and in vivo experiments showed that CAR-T cells with trap protein secretion further attenuated inhibitory T cell signaling, enhanced T cell persistence and expansion, and improved effector function and resistance to exhaustion. In the xenograft mouse model, CAR-T cells with trap protein secretion exhibited significantly enhanced antitumor immunity and efficacy. With these observations, we demonstrate the potential of trap protein self-secreting CAR-T cells as a potent therapy for solid tumors.

5.
Biomaterials ; 217: 119265, 2019 10.
Article in English | MEDLINE | ID: mdl-31271861

ABSTRACT

Cancer immunotherapy has recently burst onto the center stage of cancer treatment and research. T lymphocyte adoptive cellular transfer (ACT), a form of cancer immunotherapy, has spawned unprecedented complete remissions for terminal patients with certain leukemias and lymphomas. Unfortunately, the successes have been overshadowed by the disappointing clinical results of ACT administered to treat solid tumors, in addition to the toxicities associated with the treatment, a lack of efficacy in a significant proportion of the patient population, and cancer relapse following the treatment. Biomaterials hold the promise of addressing these shortcomings. ACT consists of two main stages - T lymphocyte ex vivo expansion followed by reinfusion into the patient - and biomaterials can improve the efficacy of ACT at both stages. In this review, we highlight recent advances in the use of biomaterials for T lymphocyte adoptive cellular cancer immunotherapy and discuss the challenges at each stage.


Subject(s)
Biocompatible Materials/chemistry , Immunotherapy , T-Lymphocytes/immunology , Cell Proliferation , Humans , Nanoparticles/chemistry , T-Lymphocytes/cytology , Tissue Scaffolds/chemistry
6.
Theranostics ; 9(25): 7872-7888, 2019.
Article in English | MEDLINE | ID: mdl-31695805

ABSTRACT

Quantitative mass spectrometry (MS) continues to deepen our understanding of the immune system, quickly becoming the gold standard for obtaining high-throughput, quantitative data on biomolecules. The development of targeted and multiplexed assays for biomarker quantification makes MS an attractive tool both for diagnosing diseases and for quantifying the effects of immunotherapeutics. Because of its accuracy, the use of MS for identifying biomarkers of disease reduces the potential for misdiagnosis and overtreatment. Advances in workflows for sample processing have drastically reduced processing time and complexities due to sample preparation, making MS a more accessible technology. In this review, we present how recent developments in proteomics and metabolomics make MS an essential component of enhancing and monitoring the efficacy of immunotherapeutic treatments.


Subject(s)
Biomarkers/metabolism , Metabolomics/methods , Proteomics/methods , Animals , Humans , Immunologic Factors/immunology , Immunologic Factors/metabolism , Immunotherapy/methods , Tandem Mass Spectrometry/methods , Therapeutic Uses
7.
Sci Rep ; 9(1): 810, 2019 01 28.
Article in English | MEDLINE | ID: mdl-30692582

ABSTRACT

Magnesium (Mg) and its alloys have shown attractive biocompatibility and mechanical strength for medical applications, but low corrosion resistance of Mg in physiological environment limits its broad clinical translation. Hydroxyapatite (HA) nanoparticles (nHA) are promising coating materials for decreasing degradation rates and prolonging mechanical strength of Mg-based implants while enhancing bone healing due to their osteoconductivity and osteoinductivity. Conformal HA coatings with nano-to-submicron structures, namely nHA and mHA coatings, were deposited successfully on Mg plates and rods using a transonic particle acceleration (TPA) process under two different conditions, characterized, and investigated for their effects on Mg degradation in vitro. The nHA and mHA coatings enhanced corrosion resistance of Mg and retained 86-90% of ultimate compressive strength after in vitro immersion in rSBF for 6 weeks, much greater than non-coated Mg that only retained 66% of strength. Mg-based rods with or without coatings showed slower degradation than the respective Mg-based plates in rSBF after 6 weeks, likely because of the greater surface-to-volume ratio of Mg plates than Mg rods. This indicates that Mg-based plate and screw devices may undergo different degradation even when they have the same coatings and are implanted at the same or similar anatomical locations. Therefore, in addition to locations of implantation, the geometry, dimension, surface area, volume, and mass of Mg-based implants and devices should be carefully considered in their design and processing to ensure that they not only provide adequate structural and mechanical stability for bone fixation, but also support the functions of bone cells, as clinically required for craniomaxillofacial (CMF) and orthopedic implants. When the nHA and mHA coated Mg and non-coated Mg plates were cultured with bone marrow derived mesenchymal stem cells (BMSCs) using the in vitro direct culture method, greater cell adhesion densities were observed under indirect contact conditions than that under direct contact conditions for the nHA and mHA coated Mg. In comparison with non-coated Mg, the nHA and mHA coated Mg reduced BMSC adhesion densities directly on the surface, but increased the average BMSC adhesion densities under indirect contact. Further long-term studies in vitro and in vivo are necessary to elucidate the effects of nHA and mHA coatings on cell functions and tissue healing.


Subject(s)
Coated Materials, Biocompatible/chemistry , Durapatite/chemistry , Magnesium/chemistry , Mesenchymal Stem Cells/cytology , Absorbable Implants , Biochemical Phenomena , Cell Adhesion , Cells, Cultured , Corrosion , Humans , Materials Testing , Nanoparticles , Particle Size , Surface Properties
SELECTION OF CITATIONS
SEARCH DETAIL