Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
Mol Ther Methods Clin Dev ; 27: 415-430, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36381305

ABSTRACT

Cord blood (CB)-derived natural killer (NK) cells that are genetically engineered to express a chimeric antigen receptor (CAR) are an attractive off-the-shelf therapy for the treatment of cancer, demonstrating a robust safety profile in vivo. For poor prognosis brain tumors such as glioblastoma multiforme (GBM), novel therapies are urgently needed. Although CAR-T cells demonstrate efficacy in preclinical GBM models, an off-the-shelf product may exhibit unwanted side effects like graft-versus-host disease. Hence, we developed an off-the-shelf CAR-NK cell approach using a B7H3 CAR and showed that CAR-transduced NK cells have robust cytolytic activity against GBM cells in vitro. However, transforming growth factor (TGF)-ß within the tumor microenvironment has devastating effects on the cytolytic activity of both unmodified and CAR-transduced NK cells. To overcome this potent immune suppression, we demonstrated that co-transducing NK cells with a B7H3 CAR and a TGF-ß dominant negative receptor (DNR) preserves cytolytic function in the presence of exogenous TGF-ß. This study demonstrates that a novel DNR and CAR co-expression strategy may be a promising therapeutic for recalcitrant CNS tumors like GBM.

2.
Nano Res ; 15(3): 2300-2314, 2022 Mar.
Article in English | MEDLINE | ID: mdl-36089987

ABSTRACT

Despite the promise of immunotherapy such as the immune checkpoint inhibitors (ICIs) anti-PD-1 and anti-CTLA-4 for advanced melanoma, only 26%-52% of patients respond, and many experience grade III/IV immune-related adverse events. Motivated by the need for an effective therapy for patients non-responsive to clinically approved ICIs, we have developed a novel nanoimmunotherapy that combines locally administered Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) with systemically administered agonistic anti-CD137 monoclonal antibody therapy (aCD137). PBNP-PTT was administered at various thermal doses to melanoma cells in vitro, and was combined with aCD137 in vivo to test treatment effects on melanoma tumor progression, animal survival, immunological protection against tumor rechallenge, and hepatotoxicity. When administered at a melanoma-specific thermal dose, PBNP-PTT elicits immunogenic cell death (ICD) in melanoma cells and upregulates markers associated with antigen presentation and immune cell co-stimulation in vitro. Consequently, PBNP-PTT eliminates primary melanoma tumors in vivo, yielding long-term tumor-free survival. However, the antitumor immune effects generated by PBNP-PTT cannot eliminate secondary tumors, despite significantly slowing their growth. The addition of aCD137 enables significant abscopal efficacy and improvement of survival, functioning through activated dendritic cells and tumor-infiltrating CD8+ T cells, and generates CD4+ and CD8+ T cell memory that manifests in the rejection of tumor rechallenge, with no long-term hepatotoxicity. This study describes for the first time a novel and effective nanoimmunotherapy combination of PBNP-PTT with aCD137 mAb therapy for melanoma.

3.
Nat Commun ; 13(1): 2155, 2022 04 20.
Article in English | MEDLINE | ID: mdl-35444206

ABSTRACT

T cell factor 1 (TCF1) is required for memory and stem-like CD8+ T cell functions. How TCF1 partners with other transcription factors to regulate transcription remains unclear. Here we show that negative elongation factor (NELF), an RNA polymerase II (Pol II) pausing factor, cooperates with TCF1 in T cell responses to cancer. Deletion of mouse Nelfb, which encodes the NELFB subunit, in mature T lymphocytes impairs immune responses to both primary tumor challenge and tumor antigen-mediated vaccination. Nelfb deletion causes more exhausted and reduced memory T cell populations, whereas its ectopic expression boosts antitumor immunity and efficacy of chimeric antigen receptor T-cell immunotherapy. Mechanistically, NELF is associated with TCF1 and recruited preferentially to the enhancers and promoters of TCF1 target genes. Nelfb ablation reduces Pol II pausing and chromatin accessibility at these TCF1-associated loci. Our findings thus suggest an important and rate-limiting function of NELF in anti-tumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes , RNA Polymerase II , Animals , Mice , Promoter Regions, Genetic , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
4.
Mol Ther Methods Clin Dev ; 23: 296-306, 2021 Dec 10.
Article in English | MEDLINE | ID: mdl-34729377

ABSTRACT

The synovial sarcoma X breakpoint 2 (SSX2) belongs to a multigene family of cancer-testis antigens and can be found overexpressed in multiple malignancies. Its restricted expression in immune-privileged normal tissues suggest that SSX2 may be a relevant target antigen for chimeric antigen receptor (CAR) therapy. We have developed a T cell receptor (TCR)-like antibody (Fab/3) that binds SSX2 peptide 41-49 (KASEKIFYV) in the context of HLA-A∗-0201. The sequence of Fab/3 was utilized to engineer a CAR with the CD3 zeta intra-cellular domain along with either a CD28 or 4-1BB costimulatory endodomain. Human T cells from HLA-A2+ donors were transduced to mediate anti-tumor activity against acute myeloid leukemia (AML) tumor cells. Upon challenge with HLA-A2+/SSX2+ AML tumor cells, CAR-expressing T cells released interferon-γ and eliminated the tumor cells in a long-term co-culture assay. Using the HLA-A2+ T2 cell line, we demonstrated a strong specificity of the single-chain variable fragment (scFv) for SSX2 p41-49 and the closely related SSX3 p41-49, with no response against the others SSX-homologous peptides or unrelated homologous peptides. Since SSX3 has not been observed in tumor cells and expression cannot be induced by pharmacological intervention, SSX241-49 represents an attractive target for CAR-based cellular therapy to treat multiple types of cancer.

5.
J Transl Med ; 17(1): 321, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31547819

ABSTRACT

BACKGROUND: Medulloblastoma (MB), the most common pediatric brain cancer, presents with a poor prognosis in a subset of patients with high risk disease, or at recurrence, where current therapies are ineffective. Cord blood (CB) natural killer (NK) cells may be promising off-the-shelf effector cells for immunotherapy due to their recognition of malignant cells without the need for a known target, ready availability from multiple banks, and their potential to expand exponentially. However, they are currently limited by immune suppressive cytokines secreted in the MB tumor microenvironment including Transforming Growth Factor ß (TGF-ß). Here, we address this challenge in in vitro models of MB. METHODS: CB-derived NK cells were modified to express a dominant negative TGF-ß receptor II (DNRII) using retroviral transduction. The ability of transduced CB cells to maintain function in the presence of medulloblastoma-conditioned media was then assessed. RESULTS: We observed that the cytotoxic ability of nontransduced CB-NK cells was reduced in the presence of TGF-ß-rich, medulloblastoma-conditioned media (21.21 ± 1.19% killing at E:T 5:1 in the absence vs. 14.98 ± 2.11% in the presence of medulloblastoma-conditioned media, n = 8, p = 0.02), but was unaffected in CB-derived DNRII-transduced NK cells (21.11 ± 1.84% killing at E:T 5:1 in the absence vs. 21.81 ± 3.37 in the presence of medulloblastoma-conditioned media, n = 8, p = 0.85. We also observed decreased expression of CCR2 in untransduced NK cells (mean CCR2 MFI 826 ± 117 in untransduced NK + MB supernatant from mean CCR2 MFI 1639.29 ± 215 in no MB supernatant, n = 7, p = 0.0156), but not in the transduced cells. Finally, we observed that CB-derived DNRII-transduced NK cells may protect surrounding immune cells by providing a cytokine sink for TGF-ß (decreased TGF-ß levels of 610 ± 265 pg/mL in CB-derived DNRII-transduced NK cells vs. 1817 ± 342 pg/mL in untransduced cells; p = 0.008). CONCLUSIONS: CB NK cells expressing a TGF-ß DNRII may have a functional advantage over unmodified NK cells in the presence of TGF-ß-rich MB, warranting further investigation on its potential applications for patients with medulloblastoma.


Subject(s)
Cerebellar Neoplasms/immunology , Killer Cells, Natural/immunology , Medulloblastoma/immunology , Transforming Growth Factor beta/metabolism , Cell Line, Tumor , Down-Regulation , Fetal Blood/cytology , Humans , Killer Cells, Natural/transplantation , Neutralization Tests , Receptors, CCR2/metabolism , Transplantation, Homologous
6.
Clin Cancer Res ; 25(14): 4400-4412, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31010834

ABSTRACT

PURPOSE: The ability of natural killer (NK) cells to lyse allogeneic targets, without the need for explicit matching or priming, makes them an attractive platform for cell-based immunotherapy. Umbilical cord blood is a practical source for generating banks of such third-party NK cells for "off-the-shelf" cell therapy applications. NK cells are highly cytolytic, and their potent antitumor effects can be rapidly triggered by a lack of HLA expression on interacting target cells, as is the case for a majority of solid tumors, including neuroblastoma. Neuroblastoma is a leading cause of pediatric cancer-related deaths and an ideal candidate for NK-cell therapy. However, the antitumor efficacy of NK cells is limited by immunosuppressive cytokines in the tumor microenvironment, such as TGFß, which impair NK cell function and survival. EXPERIMENTAL DESIGN: To overcome this, we genetically modified NK cells to express variant TGFß receptors, which couple a mutant TGFß dominant-negative receptor to NK-specific activating domains. We hypothesized that with these engineered receptors, inhibitory TGFß signals are effectively converted to activating signals. RESULTS: Modified NK cells exhibited higher cytotoxic activity against neuroblastoma in a TGFß-rich environment in vitro and superior progression-free survival in vivo, as compared with their unmodified controls. CONCLUSIONS: Our results support the development of "off-the-shelf" gene-modified NK cells, that overcome TGFß-mediated immune evasion, in patients with neuroblastoma and other TGFß-secreting malignancies.


Subject(s)
Genetic Engineering , Immunotherapy/methods , Killer Cells, Natural/immunology , Neuroblastoma/drug therapy , Neuroblastoma/immunology , Receptor, Transforming Growth Factor-beta Type II/immunology , Tumor Microenvironment/immunology , Animals , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cell Line, Tumor , Female , Humans , Killer Cells, Natural/metabolism , Male , Mice , Mice, Inbred NOD , Neuroblastoma/pathology , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism , Xenograft Model Antitumor Assays
7.
Oncotarget ; 9(86): 35611-35622, 2018 Nov 02.
Article in English | MEDLINE | ID: mdl-30479692

ABSTRACT

Incubation of umbilical cord blood (UCB) derived regulatory T-cells (Tregs) with fucosyltransferase enzyme improves their ability to home to the target tissue to prevent graft vs. host disease (GVHD). We report results of 5 patients (Double UCB Transplant, n=2; Peripheral Blood Matched Unrelated Donor Transplant, n=3) who received UCB-Tregs (Dose level = 1×106/kg), infused one day prior to the donor graft. All patients received their designated UCB-Treg dose without any infusion reaction. The ratio of conventional T-cells in donor graft was at least 10 times higher than infused UCB-Tregs (ratio range, 12-356). All patients engrafted at median of 13 days (range, 8-17 days). One patient died due to brain hemorrhage on day 45. A bi-modal increase of plasma IL-10 level occurred on day 7 and day 21 and notably, plasma IL-2 level dropped significantly in all patients at Day 7. All evaluable patients developed ≥grade II acute GVHD and at 1 year follow up, all were alive and without evidence of disease relapse. No increase in the chronic GVHD biomarkers (REG3a and Elafin) was observed at day 7. At the time of last follow up, all evaluable patients were off immune-suppression. Stage 2 of this clinical trial examining UCB-Treg at dose level= 1×107/kg is currently underway.

8.
Oncotarget ; 9(72): 33694-33701, 2018 Sep 14.
Article in English | MEDLINE | ID: mdl-30263095

ABSTRACT

Regulatory T cells (Tregs) are an important component of the immune system involved in regulation of immune cell proliferation and inflammatory responses and preventing autoimmune diseases. The use of Tregs in cellular therapy has recently been explored in clinical trials specifically evaluating the role of ex vivo expanded Tregs in the prevention of graft-versus-host disease during stem cell transplantation. The possibility of Treg use in the clinic requires clinical grade expansion of Tregs for development of cell therapy protocols and proper homing of Tregs to the intended target. Here we demonstrate a novel medium composition to expand CB Tregs, specifically upregulation the homing and activation markers CD62L and cutaneous lymphocyte antigen (CLA). CLA expression was uniquely acquired during activation of Tregs with subsequent loss or lack of expression with media change. This finding highlights the importance of proper growth conditions unique to Tregs that can alter expression of proteins and establishes a baseline for expanding marker specific Tregs that home and target unique tissues.

9.
Br J Haematol ; 177(3): 457-466, 2017 05.
Article in English | MEDLINE | ID: mdl-28295190

ABSTRACT

Multiple myeloma (MM) is a disease with known immune dysregulation. Natural killer (NK) cells have shown preclinical activity in MM. We conducted a first-in-human study of umbilical cord blood-derived (CB) NK cells for MM patients undergoing high dose chemotherapy and autologous haematopoietic stem cell transplantation (auto-HCT). Patients received lenalidomide (10 mg) on days -8 to -2, melphalan 200 mg/m2 on day -7, CB-NK cells on day -5 and auto-HCT on day 0. Twelve patients were enrolled, three on each of four CB-NK cell dose levels: 5 × 106 , 1 × 107 , 5 × 107 and 1 × 108 CB-NK cells/kg. Ten patients had either high-risk chromosomal changes or a history of relapsed/progressed disease. There were no infusional toxicities and no graft-versus-host disease. One patient failed to engraft due to poor autologous graft quality and was rescued with a back-up autologous graft. Overall, 10 patients achieved at least a very good partial response as their best response, including eight with near complete response or better. With a median follow-up of 21 months, four patients have progressed or relapsed, two of whom have died. CB-NK cells were detected in vivo in six patients, with an activated phenotype (NKG2D+ /NKp30+ ). These data warrant further development of this novel cellular therapy.


Subject(s)
Fetal Blood/immunology , Hematopoietic Stem Cell Transplantation/methods , Killer Cells, Natural/transplantation , Multiple Myeloma/therapy , Adoptive Transfer/methods , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Separation/methods , Combined Modality Therapy , Female , Flow Cytometry/methods , Graft Survival , Humans , Lenalidomide , Male , Melphalan/administration & dosage , Middle Aged , Thalidomide/administration & dosage , Thalidomide/analogs & derivatives , Treatment Outcome
10.
Cytotherapy ; 19(3): 408-418, 2017 03.
Article in English | MEDLINE | ID: mdl-28109751

ABSTRACT

Cord blood (CB) natural killer (NK) cells are promising effector cells for tumor immunotherapy but are currently limited by immune-suppressive cytokines in the tumor microenvironment, such as transforming growth factor (TGF-ß). We observed that TGF-ß inhibits expression of activating receptors such as NKG2D and DNAM1 and decreases killing activity against glioblastoma tumor cells through inhibition of perforin secretion. To overcome the detrimental effects of TGF-ß, we engrafted a dominant negative TGF-ß receptor II (DNRII) on CB-derived NK cells by retroviral transduction and evaluated their ability to kill glioblastoma cells in the presence of TGF-ß. After manufacture using Good Manufacturing Practice-compliant methodologies and transduction with DNRII, CB-derived DNRII-transduced NK cells expanded to clinically relevant numbers and retained both their killing ability and their secretion of interferon-γ upon activation. More important, these cells maintained both perforin expression and NKG2D/DNMA1 expression in the presence of TGF-ß allowing for recognition and killing of glioblastoma tumor cells. Hence, NK cells expressing a DNRII should have a functional advantage over unmodified NK cells in the presence of TGF-ß-secreting tumors and may be an important therapeutic approach for patients with cancer.


Subject(s)
Brain Neoplasms/therapy , Fetal Blood/cytology , Genetic Therapy/methods , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , Killer Cells, Natural/transplantation , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Brain Neoplasms/immunology , Cell Line, Tumor , Cytokines/metabolism , Fetal Blood/immunology , Fetal Blood/transplantation , Genes, Dominant , Glioblastoma/immunology , Humans , Interferon-gamma/metabolism , Jurkat Cells , K562 Cells , Killer Cells, Natural/cytology , Killer Cells, Natural/metabolism , Perforin/metabolism , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
11.
Cytotherapy ; 18(10): 1312-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27497700

ABSTRACT

Regulatory T cells (Tregs) play a fundamental role in the maintenance of self-tolerance and immune homeostasis. Defects in Treg function and/or frequencies have been reported in multiple disease models. Amyotrophic lateral sclerosis (ALS) is a progressive neurodegenerative disorder affecting upper and lower motor neurons. Compelling evidence supports a neuroprotective role for Tregs in this disease. Indeed, rapid progression in ALS patients is associated with decreased FoxP3 expression and Treg frequencies. Thus, we propose that strategies to restore Treg number and function may slow disease progression in ALS. In this study, we developed a robust, Good Manufacturing Practice (GMP)-compliant procedure to enrich and expand Tregs from ALS patients. Tregs isolated from these patients were phenotypically similar to those from healthy individuals but were impaired in their ability to suppress T-cell effector function. In vitro expansion of Tregs for 4 weeks in the presence of GMP-grade anti-CD3/CD28 beads, interleukin (IL)-2 and rapamcyin resulted in a 25- to 200-fold increase in their number and restored their immunoregulatory activity. Collectively, our data facilitate and support the implementation of clinical trials of adoptive therapy with ex vivo expanded and highly suppressive Tregs in patients with ALS.


Subject(s)
Adoptive Transfer/standards , Amyotrophic Lateral Sclerosis/pathology , Cell Separation , Cell- and Tissue-Based Therapy/standards , Primary Cell Culture , T-Lymphocytes, Regulatory/pathology , Adoptive Transfer/methods , Amyotrophic Lateral Sclerosis/immunology , Case-Control Studies , Cell Separation/methods , Cell Separation/standards , Cell- and Tissue-Based Therapy/methods , Cells, Cultured , Guideline Adherence/standards , Humans , Immune Tolerance , Interleukin-2/metabolism , Primary Cell Culture/methods , Primary Cell Culture/standards , T-Lymphocytes, Regulatory/immunology
12.
Cytotherapy ; 18(11): 1410-1421, 2016 11.
Article in English | MEDLINE | ID: mdl-27421740

ABSTRACT

Natural killer (NK) cells are members of the innate immune system that recognize target cells via activating and inhibitory signals received through cell receptors. Derived from the lymphoid lineage, NK cells are able to produce cytokines and exert a cytotoxic effect on viral infected and malignant cells. It is their unique ability to lyse target cells rapidly and without prior education that renders NK cells a promising effector cell for adoptive cell therapy. However, both viruses and tumors employ evasion strategies to avoid attack by NK cells, which represent biological challenges that need to be harnessed to fully exploit the cytolytic potential of NK cells. Using genetic modification, the function of NK cells can be enhanced to improve their homing, cytolytic activity, in vivo persistence and safety. Examples include gene modification to express chemokine, high-affinity Fc receptor and chimeric antigen receptors, suicide genes and the forced expression of cytokines such as interleukin (IL)-2 and IL-15. Preclinical studies have clearly demonstrated that such approaches are effective in improving NK-cell function, homing and safety. In this review, we summarize the recent advances in the genetic manipulations of NK cells and their application for cellular immunotherapeutic strategies.


Subject(s)
Genetic Engineering , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Neoplasms/immunology , Neoplasms/therapy , Animals , Humans , Immune Evasion , Treatment Outcome
13.
Methods Mol Biol ; 1441: 203-13, 2016.
Article in English | MEDLINE | ID: mdl-27177668

ABSTRACT

As part of the innate immune system, natural killer (NK) cells are regarded as promising effector cells for adoptive cell therapy approaches to treat patients with cancer. In some cases, genetic modification of the NK cells may be considered but such manipulation has to be integrated into the expansion method to allow the generation of clinically relevant numbers of gene-modified NK cells. Therefore, an efficient gene transfer procedure is needed.Our group developed a retrovirus-based transduction protocol capable of robust expansion of gene-modified NK cells with a high rate of transgene expression. Actively dividing cells is a prerequisite for efficient gene transfer when using a retroviral vector. In the procedure presented here, strong activation of the NK cells was provided by a combination of IL-15 and the K-562 feeder cells. Beside the interest in developing a simple procedure compliant with good manufacturing practice (GMP) for the production of therapeutic products, this approach also provides a valuable means of generating genetically modified primary NK cells for future preclinical studies.


Subject(s)
Interleukin-15/genetics , Killer Cells, Natural/cytology , Transduction, Genetic , Cells, Cultured , Feeder Cells/cytology , Feeder Cells/immunology , Genetic Vectors , Humans , Interleukin-15/metabolism , K562 Cells , Killer Cells, Natural/immunology , Retroviridae/genetics
14.
Mol Ther ; 24(6): 1135-1149, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27019998

ABSTRACT

Chimeric antigen receptor (CAR) T cells have shown great promise in the treatment of hematologic malignancies but more variable results in the treatment of solid tumors and the persistence and expansion of CAR T cells within patients has been identified as a key correlate of antitumor efficacy. Lack of immunological "space", functional exhaustion, and deletion have all been proposed as mechanisms that hamper CAR T-cell persistence. Here we describe the events following activation of third-generation CAR T cells specific for GD2. CAR T cells had highly potent immediate effector functions without evidence of functional exhaustion in vitro, although reduced cytokine production reversible by PD-1 blockade was observed after longer-term culture. Significant activation-induced cell death (AICD) of CAR T cells was observed after repeated antigen stimulation, and PD-1 blockade enhanced both CAR T-cell survival and promoted killing of PD-L1(+) tumor cell lines. Finally, we assessed CAR T-cell persistence in patients enrolled in the CARPETS phase 1 clinical trial of GD2-specific CAR T cells in the treatment of metastatic melanoma. Together, these data suggest that deletion also occurs in vivo and that PD-1-targeted combination therapy approaches may be useful to augment CAR T-cell efficacy and persistence in patients.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Melanoma/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/transplantation , Animals , Cell Line, Tumor , Cell Survival/drug effects , Gangliosides/immunology , Humans , Lymphocyte Activation , Melanoma/immunology , Mice , Neoplasm Metastasis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
15.
Mol Ther Methods Clin Dev ; 3: 16012, 2016.
Article in English | MEDLINE | ID: mdl-27006970

ABSTRACT

Current cell processing technologies for gene and cell therapies are often slow, expensive, labor intensive and are compromised by high cell losses and poor selectivity thus limiting the efficacy and availability of clinical cell therapies. We employ cell-specific on-demand mechanical intracellular impact from laser pulse-activated plasmonic nanobubbles (PNB) to process heterogeneous human cell grafts ex vivo with dual simultaneous functionality, the high cell type specificity, efficacy and processing rate for transfection of target CD3+ cells and elimination of subsets of unwanted CD25+ cells. The developed bulk flow PNB system selectively processed human cells at a rate of up to 100 million cell/minute, providing simultaneous transfection of CD3+ cells with the therapeutic gene (FKBP12(V36)-p30Caspase9) with the efficacy of 77% and viability 95% (versus 12 and 60%, respectively, for standard electroporation) and elimination of CD25+ cells with 99% efficacy. PNB flow technology can unite and replace several methodologies in an all-in-one universal ex vivo simultaneous procedure to precisely and rapidly prepare a cell graft for therapy. PNB's can process various cell systems including cord blood, stem cells, and bone marrow.

16.
Blood ; 125(19): 2885-92, 2015 May 07.
Article in English | MEDLINE | ID: mdl-25778529

ABSTRACT

Delayed engraftment is a major limitation of cord blood transplantation (CBT), due in part to a defect in the cord blood (CB) cells' ability to home to the bone marrow. Because this defect appears related to low levels of fucosylation of cell surface molecules that are responsible for binding to P- and E-selectins constitutively expressed by the marrow microvasculature, and thus for marrow homing, we conducted a first-in-humans clinical trial to correct this deficiency. Patients with high-risk hematologic malignancies received myeloablative therapy followed by transplantation with 2 CB units, one of which was treated ex vivo for 30 minutes with the enzyme fucosyltransferase-VI and guanosine diphosphate fucose to enhance the interaction of CD34(+) stem and early progenitor cells with microvessels. The results of enforced fucosylation for 22 patients enrolled in the trial were then compared with those for 31 historical controls who had undergone double unmanipulated CBT. The median time to neutrophil engraftment was 17 days (range, 12-34 days) compared with 26 days (range, 11-48 days) for controls (P = .0023). Platelet engraftment was also improved: median was 35 days (range, 18-100 days) compared with 45 days (range, 27-120 days) for controls (P = .0520). These findings support ex vivo fucosylation of multipotent CD34(+) CB cells as a clinically feasible means to improve engraftment efficiency in the double CBT setting. The trial is registered to www.clinicaltrials.gov as #NCT01471067.


Subject(s)
Blood Platelets/cytology , Fetal Blood/cytology , Fucose/metabolism , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Neutrophils/transplantation , Adolescent , Adult , Aged , Blood Platelets/immunology , Cohort Studies , E-Selectin/metabolism , Feasibility Studies , Female , Fetal Blood/immunology , Fucosyltransferases/metabolism , Graft vs Host Disease , Hematologic Neoplasms/immunology , Hematologic Neoplasms/mortality , Hematopoietic Stem Cells/immunology , Humans , Male , Middle Aged , Neutrophils/cytology , Neutrophils/immunology , P-Selectin/metabolism , Platelet Transfusion , Prognosis , Survival Rate , Young Adult
17.
J Immunother ; 38(1): 12-23, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25415284

ABSTRACT

Cancer immunotherapy has long been used in the treatment of metastatic melanoma, and an anti-CTLA-4 monoclonal antibody treatment has recently been approved by the US Food and Drug Administration. Targeted therapies such as small molecule kinase inhibitors targeting deregulated mitogen-activated protein kinase (MAPK) signaling have markedly improved melanoma control in up to 50% of metastatic disease patients and have likewise been recently approved. Combination therapies for melanoma have been proposed as a way to exploit the high-level but short-term responses associated with kinase inhibitor therapies and the low-level but longer-term responses associated with immunotherapy. Cancer immunotherapy now includes adoptive transfer of autologous tumor-specific chimeric antigen receptor (CAR) T cells and this mode of therapy is a candidate for combination with small molecule drugs. This paper describes CART cells that target GD2-expressing melanoma cells and investigates the effects of approved MAPK pathway-targeted therapies for melanoma [vemurafenib (Vem), dabrafenib (Dab), and trametinib (Tram)] on the viability, activation, proliferation, and cytotoxic T lymphocyte activity of these CAR T cells, as well as on normal peripheral blood mononuclear cells. We report that, although all these drugs lead to inhibition of stimulated T cells at high concentrations in vitro, only Vem inhibited T cells at concentrations equivalent to reported plasma concentrations in treated patients. Although the combination of Dab and Tram also resulted in inhibition of T-cell effector functions at some therapeutic concentrations, Dab itself had little adverse effect on CAR T-cell function. These findings may have implications for novel therapeutic combinations of adoptive CAR T-cell immunotherapy and MAPK pathway inhibitors.


Subject(s)
Antineoplastic Agents/pharmacology , Lymphocyte Activation/drug effects , Melanoma/immunology , Protein Kinase Inhibitors/pharmacology , Receptors, Antigen, T-Cell/immunology , Cell Line , Cell Survival/drug effects , Flow Cytometry , Humans , Imidazoles/pharmacology , Immunotherapy/methods , In Vitro Techniques , Indoles/pharmacology , Leukocytes, Mononuclear/immunology , Lymphocyte Activation/immunology , MAP Kinase Signaling System/drug effects , Oximes/pharmacology , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Pyridones/pharmacology , Pyrimidinones/pharmacology , Sulfonamides/pharmacology , T-Lymphocytes, Cytotoxic , Transfection , Vemurafenib
18.
Blood ; 125(9): 1502-6, 2015 Feb 26.
Article in English | MEDLINE | ID: mdl-25428215

ABSTRACT

Adoptive therapy with regulatory T cells (Tregs) to prevent graft-versus-host disease (GVHD) would benefit from a strategy to improve homing to the sites of inflammation. We hypothesized that adding fucose to human Tregs, forming the Sialyl Lewis X moiety on P-selectin glycoprotein ligand-1, would improve their trafficking pattern. The selectin pathway recruiter, α-1,3-fucosyltransferase-VI enzyme, significantly increased Treg surface fucosylation (66% vs 8%). In a xenogenic GVHD mouse model, fucosylated Tregs showed prolonged periods of in vivo persistence. When given at a lower dose compared with the untreated Tregs, the murine recipients of fucosylated Tregs maintained weight, had ameliorated clinical GVHD, and improved survival (70% vs 30%; P < .0001). These preclinical data indicate that fucosylated human Tregs is an effective strategy for prevention of GVHD and, as such, warrants consideration for future clinical trials.


Subject(s)
Disease Models, Animal , Fucose/metabolism , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , T-Lymphocytes, Regulatory/immunology , Animals , Blotting, Western , Cells, Cultured , E-Selectin/metabolism , Female , Fetal Blood/cytology , Fetal Blood/immunology , Fetal Blood/metabolism , Flow Cytometry , Fucosyltransferases/metabolism , Graft vs Host Disease/mortality , Humans , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Oligosaccharides/metabolism , Sialyl Lewis X Antigen
20.
Blood ; 124(13): 2034-45, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25051962

ABSTRACT

A subset of regulatory B cells (Bregs) in mice negatively regulate T-cell immune responses through the secretion of regulatory cytokines such as IL-10 and direct cell-cell contact and have been linked to experimental models of autoimmunity, inflammation, and cancer. However, the regulatory function of Bregs in human disease is much less clear. Here we demonstrate that B cells with immunoregulatory properties are enriched within both the CD19(+)IgM(+)CD27(+) memory and CD19(+)CD24(hi)CD38(hi) transitional B-cell subsets in healthy human donors. Both subsets suppressed the proliferation and interferon-γ production of CD3/CD28-stimulated autologous CD4(+) T cells in a dose-dependent manner, and both relied on IL-10 secretion as well as cell-cell contact, likely mediated through CD80 and CD86, to support their full suppressive function. Moreover, after allogeneic stem cell transplantation, Bregs from patients with chronic graft-versus-host disease (cGVHD) were less frequent and less likely to produce IL-10 than were Bregs from healthy donors and patients without cGVHD. These findings suggest that Bregs may be involved in the pathogenesis of cGVHD and support future investigation of regulatory B cell-based therapy in the treatment of this disease.


Subject(s)
B-Lymphocyte Subsets/immunology , B-Lymphocytes, Regulatory/immunology , Graft vs Host Disease/immunology , Immunoglobulin M/immunology , Immunologic Memory , ADP-ribosyl Cyclase 1/metabolism , Antigens, CD19/metabolism , B-Lymphocyte Subsets/metabolism , B-Lymphocytes, Regulatory/metabolism , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD24 Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Cell Communication , Cells, Cultured , Chronic Disease , Cytokines/biosynthesis , Graft vs Host Disease/metabolism , Humans , Immunomodulation , Immunophenotyping , Inflammation Mediators/metabolism , Interleukin-10/biosynthesis , Lymphocyte Activation/immunology , Lymphocyte Count , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7
SELECTION OF CITATIONS
SEARCH DETAIL
...