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










Publication year range
1.
J Cancer Res Clin Oncol ; 149(7): 2757-2769, 2023 Jul.
Article in English | MEDLINE | ID: mdl-35776199

ABSTRACT

PURPOSE: A barrier to widespread adoption of chimeric antigen receptor (CAR) T-cell therapy is toxicity. To address this, we recently developed a novel antibody-T-cell receptor (AbTCR) platform (trademarked as ARTEMIS®) which was designed to leverage natural immune receptor signaling and regulation. The AbTCR platform includes a gamma/delta (γδ) TCR-based AbTCR construct and a separate co-stimulatory molecule, both engineered to be tumor-specific. Here, we aim to assess the safety and preliminary efficacy of a CD19-directed AbTCR T-cell therapy. METHODS: We generated ET019003 T cells, which are autologous CD19-directed AbTCR T cells. We then conducted an early phase I study to evaluate the safety and preliminary efficacy of ET019003 T cells for the treatment of CD19-positive relapsed/refractory (r/r) B-cell lymphoma. RESULTS: Sixteen patients enrolled in this study and 12 patients were treated. Of the 12 patients treated, 6 patients (50%) achieved a complete response (CR), and 4 (33%) achieved a partial response (PR) (best objective response rate [ORR] of 83%). CRs were durable, including 2 patients with ongoing CRs for 22.7 months and 23.2 months. ET019003 was well-tolerated with an attractive safety profile. No patients experienced severe (grade ≥ 3) cytokine release syndrome (CRS) and only 1 patient experienced immune effector cell-associated neurotoxicity syndrome (ICANS) of any grade. Significant elevations of cytokine levels were not seen, even in patients with marked expansion of ET019003 T cells. CONCLUSION: This study provides initial clinical validation of the AbTCR platform as a novel cancer treatment with the potential to provide durable clinical benefit with low toxicity. TRIAL REGISTRATION: NCT03642496; Date of registration: August 22, 2018.


Subject(s)
Lymphoma, B-Cell , Neurotoxicity Syndromes , Humans , Receptors, Antigen, T-Cell , Lymphoma, B-Cell/therapy , Lymphoma, B-Cell/etiology , Neurotoxicity Syndromes/etiology , Immunotherapy, Adoptive/adverse effects , Antibodies , Antigens, CD19 , Cell- and Tissue-Based Therapy
2.
Nat Commun ; 11(1): 5998, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33243988

ABSTRACT

Intratumoral heterogeneity is a common feature of many myeloid leukemias and a significant reason for treatment failure and relapse. Thus, identifying the cells responsible for residual disease and leukemia re-growth is critical to better understanding how they are regulated. Here, we show that a knock-in reporter mouse for the stem cell gene Musashi 2 (Msi2) allows identification of leukemia stem cells in aggressive myeloid malignancies, and provides a strategy for defining their core dependencies. Specifically, we carry out a high throughput screen using Msi2-reporter blast crisis chronic myeloid leukemia (bcCML) and identify several adhesion molecules that are preferentially expressed in therapy resistant bcCML cells and play a key role in bcCML. In particular, we focus on syndecan-1, whose deletion triggers defects in bcCML growth and propagation and markedly improves survival of transplanted mice. Further, live imaging reveals that the spatiotemporal dynamics of leukemia cells are critically dependent on syndecan signaling, as loss of this signal impairs their localization, migration and dissemination to distant sites. Finally, at a molecular level, syndecan loss directly impairs integrin ß7 function, suggesting that syndecan exerts its influence, at least in part, by coordinating integrin activity in bcCML. These data present a platform for delineating the biological underpinnings of leukemia stem cell function, and highlight the Sdc1-Itgß7 signaling axis as a key regulatory control point for bcCML growth and dissemination.


Subject(s)
Blast Crisis/therapy , Leukemia, Myeloid, Acute/therapy , Neoplastic Stem Cells/pathology , RNA-Binding Proteins/genetics , Syndecan-1/antagonists & inhibitors , Animals , Antineoplastic Agents/therapeutic use , Blast Crisis/genetics , Blast Crisis/pathology , Chemoradiotherapy/methods , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Gene Knock-In Techniques , Gene Knockout Techniques , Genes, Reporter/genetics , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , High-Throughput Screening Assays , Humans , Imatinib Mesylate/pharmacology , Imatinib Mesylate/therapeutic use , Integrin beta Chains/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice, Transgenic , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/radiation effects , RNA-Seq , Signal Transduction/drug effects , Syndecan-1/genetics , Syndecan-1/metabolism
3.
Cell Discov ; 4: 62, 2018.
Article in English | MEDLINE | ID: mdl-30479831

ABSTRACT

The clinical use of genetically modified T-cell therapies has led to unprecedented response rates in leukemia and lymphoma patients treated with anti-CD19 chimeric antigen receptor (CAR)-T. Despite this clinical success, FDA-approved T-cell therapies are currently limited to B-cell malignancies, and challenges remain with managing cytokine-related toxicities. We have designed a novel antibody-T-cell receptor (AbTCR) platform where we combined the Fab domain of an antibody with the γ and δ chains of the TCR as the effector domain. We demonstrate the ability of anti-CD19-AbTCR-T cells to trigger antigen-specific cytokine production, degranulation, and killing of CD19-positive cancer cells in vitro and in xenograft mouse models. By using the same anti-CD19 binding moiety on an AbTCR compared to a CAR platform, we demonstrate that AbTCR activates cytotoxic T-cell responses with a similar dose-response as CD28/CD3ζ CAR, yet does so with less cytokine release and results in T cells with a less exhausted phenotype. Moreover, in comparative studies with the clinically validated CD137 (4-1BB)-based CAR, CTL019, our anti-CD19-AbTCR shows less cytokine release and comparable tumor inhibition in a patient-derived xenograft leukemia model.

4.
Sci Transl Med ; 9(372)2017 01 11.
Article in English | MEDLINE | ID: mdl-28077677

ABSTRACT

Diabetes mellitus (DM) is a metabolic disease frequently associated with impaired bone healing. Despite its increasing prevalence worldwide, the molecular etiology of DM-linked skeletal complications remains poorly defined. Using advanced stem cell characterization techniques, we analyzed intrinsic and extrinsic determinants of mouse skeletal stem cell (mSSC) function to identify specific mSSC niche-related abnormalities that could impair skeletal repair in diabetic (Db) mice. We discovered that high serum concentrations of tumor necrosis factor-α directly repressed the expression of Indian hedgehog (Ihh) in mSSCs and in their downstream skeletogenic progenitors in Db mice. When hedgehog signaling was inhibited during fracture repair, injury-induced mSSC expansion was suppressed, resulting in impaired healing. We reversed this deficiency by precise delivery of purified Ihh to the fracture site via a specially formulated, slow-release hydrogel. In the presence of exogenous Ihh, the injury-induced expansion and osteogenic potential of mSSCs were restored, culminating in the rescue of Db bone healing. Our results present a feasible strategy for precise treatment of molecular aberrations in stem and progenitor cell populations to correct skeletal manifestations of systemic disease.


Subject(s)
Femoral Fractures/drug therapy , Fracture Healing/drug effects , Hedgehog Proteins/pharmacology , Mesenchymal Stem Cells/cytology , Stem Cell Niche , Animals , Bone and Bones/pathology , Cell Proliferation , Cell Separation , Diabetes Mellitus, Experimental/pathology , Female , Flow Cytometry , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Osteogenesis , Signal Transduction
5.
Clin Cancer Res ; 23(2): 478-488, 2017 Jan 15.
Article in English | MEDLINE | ID: mdl-27535982

ABSTRACT

PURPOSE: The majority of tumor-specific antigens are intracellular and/or secreted and therefore inaccessible by conventional chimeric antigen receptor (CAR) T-cell therapy. Given that all intracellular/secreted proteins are processed into peptides and presented by class I MHC on the surface of tumor cells, we used alpha-fetoprotein (AFP), a specific liver cancer marker, as an example to determine whether peptide-MHC complexes can be targets for CAR T-cell therapy against solid tumors. EXPERIMENTAL DESIGN: We generated a fully human chimeric antigen receptor, ET1402L1-CAR (AFP-CAR), with exquisite selectivity and specificity for the AFP158-166 peptide complexed with human leukocyte antigen (HLA)-A*02:01. RESULTS: We report that T cells expressing AFP-CAR selectively degranulated, released cytokines, and lysed liver cancer cells that were HLA-A*02:01+/AFP+ while sparing cells from multiple tissue types that were negative for either expressed proteins. In vivo, intratumoral injection of AFP-CAR T cells significantly regressed both Hep G2 and AFP158-expressing SK-HEP-1 tumors in SCID-Beige mice (n = 8 for each). Moreover, intravenous administration of AFP-CAR T cells in Hep G2 tumor-bearing NSG mice lead to rapid and profound tumor growth inhibition (n = 6). Finally, in an established intraperitoneal liver cancer xenograft model, AFP-CAR T cells showed robust antitumor activity (n = 6). CONCLUSIONS: This study demonstrates that CAR T-cell immunotherapy targeting intracellular/secreted solid tumor antigens can elicit a potent antitumor response. Our approach expands the spectrum of antigens available for redirected T-cell therapy against solid malignancies and offers a promising new avenue for liver cancer immunotherapy. Clin Cancer Res; 23(2); 478-88. ©2016 AACR.


Subject(s)
Immunotherapy , Liver Neoplasms/therapy , Receptors, Antigen, T-Cell/immunology , alpha-Fetoproteins/immunology , Animals , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Hep G2 Cells , Humans , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Mice , Molecular Targeted Therapy , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes, Cytotoxic/drug effects , Xenograft Model Antitumor Assays , alpha-Fetoproteins/antagonists & inhibitors , alpha-Fetoproteins/genetics
6.
Proc Natl Acad Sci U S A ; 113(47): E7545-E7553, 2016 11 22.
Article in English | MEDLINE | ID: mdl-27815529

ABSTRACT

Inflammation disrupts tissue architecture and function, thereby contributing to the pathogenesis of diverse diseases; the signals that promote or restrict tissue inflammation thus represent potential targets for therapeutic intervention. Here, we report that genetic or pharmacologic Hedgehog pathway inhibition intensifies colon inflammation (colitis) in mice. Conversely, genetic augmentation of Hedgehog response and systemic small-molecule Hedgehog pathway activation potently ameliorate colitis and restrain initiation and progression of colitis-induced adenocarcinoma. Within the colon, the Hedgehog protein signal does not act directly on the epithelium itself, but on underlying stromal cells to induce expression of IL-10, an immune-modulatory cytokine long known to suppress inflammatory intestinal damage. IL-10 function is required for the full protective effect of small-molecule Hedgehog pathway activation in colitis; this pharmacologic augmentation of Hedgehog pathway activity and stromal IL-10 expression are associated with increased presence of CD4+Foxp3+ regulatory T cells. We thus identify stromal cells as cellular coordinators of colon inflammation and suggest their pharmacologic manipulation as a potential means to treat colitis.


Subject(s)
Colitis/metabolism , Dextran Sulfate/adverse effects , Hedgehog Proteins/metabolism , Interleukin-10/metabolism , Signal Transduction , Animals , CD4 Antigens/metabolism , Colitis/chemically induced , Colitis/drug therapy , Disease Models, Animal , Disease Progression , Forkhead Transcription Factors/metabolism , Hedgehog Proteins/drug effects , Humans , Mice , Mutation , Signal Transduction/drug effects , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/pharmacology , T-Lymphocytes, Regulatory/metabolism , Zinc Finger Protein GLI1/genetics
7.
Nat Commun ; 7: 12169, 2016 07 18.
Article in English | MEDLINE | ID: mdl-27425143

ABSTRACT

Although we know a great deal about the phenotype and function of haematopoietic stem/progenitor cells, a major challenge has been mapping their dynamic behaviour within living systems. Here we describe a strategy to image cells in vivo with high spatial and temporal resolution, and quantify their interactions using a high-throughput computational approach. Using these tools, and a new Msi2 reporter model, we show that haematopoietic stem/progenitor cells display preferential spatial affinity for contacting the vascular niche, and a temporal affinity for making stable associations with these cells. These preferences are markedly diminished as cells mature, suggesting that programs that control differentiation state are key determinants of spatiotemporal behaviour, and thus dictate the signals a cell receives from specific microenvironmental domains. These collectively demonstrate that high-resolution imaging coupled with computational analysis can provide new biological insight, and may in the long term enable creation of a dynamic atlas of cells within their native microenvironment.


Subject(s)
Computer Simulation , Hematopoietic Stem Cells/cytology , Imaging, Three-Dimensional , Animals , Cell Tracking , Computer Systems , Female , Genes, Reporter , Green Fluorescent Proteins/metabolism , Male , Mice , RNA-Binding Proteins/metabolism , Time Factors
8.
Nature ; 534(7607): 407-411, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27281208

ABSTRACT

Pancreatic intraepithelial neoplasia is a pre-malignant lesion that can progress to pancreatic ductal adenocarcinoma, a highly lethal malignancy marked by its late stage at clinical presentation and profound drug resistance. The genomic alterations that commonly occur in pancreatic cancer include activation of KRAS2 and inactivation of p53 and SMAD4 (refs 2-4). So far, however, it has been challenging to target these pathways therapeutically; thus the search for other key mediators of pancreatic cancer growth remains an important endeavour. Here we show that the stem cell determinant Musashi (Msi) is a critical element of pancreatic cancer progression both in genetic models and in patient-derived xenografts. Specifically, we developed Msi reporter mice that allowed image-based tracking of stem cell signals within cancers, revealing that Msi expression rises as pancreatic intraepithelial neoplasia progresses to adenocarcinoma, and that Msi-expressing cells are key drivers of pancreatic cancer: they preferentially harbour the capacity to propagate adenocarcinoma, are enriched in circulating tumour cells, and are markedly drug resistant. This population could be effectively targeted by deletion of either Msi1 or Msi2, which led to a striking defect in the progression of pancreatic intraepithelial neoplasia to adenocarcinoma and an improvement in overall survival. Msi inhibition also blocked the growth of primary patient-derived tumours, suggesting that this signal is required for human disease. To define the translational potential of this work we developed antisense oligonucleotides against Msi; these showed reliable tumour penetration, uptake and target inhibition, and effectively blocked pancreatic cancer growth. Collectively, these studies highlight Msi reporters as a unique tool to identify therapy resistance, and define Msi signalling as a central regulator of pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Drug Resistance, Neoplasm/drug effects , Molecular Imaging , Nerve Tissue Proteins/genetics , Pancreatic Neoplasms/drug therapy , RNA-Binding Proteins/genetics , Animals , Carcinoma in Situ/genetics , Carcinoma in Situ/pathology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/genetics , Disease Models, Animal , Disease Progression , Drug Resistance, Neoplasm/genetics , Female , Gene Deletion , Genes, Reporter/genetics , Humans , Male , Mice , Models, Genetic , Neoplastic Cells, Circulating/metabolism , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/metabolism , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/pharmacokinetics , Oligonucleotides, Antisense/therapeutic use , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , RNA-Binding Proteins/metabolism , Signal Transduction/drug effects , Survival Rate , Xenograft Model Antitumor Assays
9.
Cancer Res ; 75(5): 792-7, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25681272

ABSTRACT

Asymmetric division is an evolutionarily conserved process that generates daughter cells with different fates through the unequal partitioning of fate determinants. While asymmetric division is critically important in generating diversity during development, its dysregulation can also promote oncogenesis. In particular, signals that shift the normal balance of symmetric and asymmetric division can lead to a differentiation arrest and trigger cancer progression. Here, we discuss the studies that have provided increasing support for this idea. Beginning with original work carried out in Drosophila, we trace more recent work in mammalian systems that suggest that the subversion of asymmetric division can contribute significantly to the development and progression of both hematologic malignancies and solid cancers.


Subject(s)
Neoplasms/pathology , Neoplastic Stem Cells/pathology , Animals , Cell Differentiation/physiology , Cell Division/physiology , Disease Progression , Humans , Neoplasms/genetics , Neoplasms/metabolism
10.
Nat Genet ; 46(3): 245-52, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24487275

ABSTRACT

Cell fate can be controlled through asymmetric division and segregation of protein determinants, but the regulation of this process in the hematopoietic system is poorly understood. Here we show that the dynein-binding protein Lis1 is critically required for hematopoietic stem cell function and leukemogenesis. Conditional deletion of Lis1 (also known as Pafah1b1) in the hematopoietic system led to a severe bloodless phenotype, depletion of the stem cell pool and embryonic lethality. Further, real-time imaging revealed that loss of Lis1 caused defects in spindle positioning and inheritance of cell fate determinants, triggering accelerated differentiation. Finally, deletion of Lis1 blocked the propagation of myeloid leukemia and led to a marked improvement in survival, suggesting that Lis1 is also required for oncogenic growth. These data identify a key role for Lis1 in hematopoietic stem cells and mark its directed control of asymmetric division as a critical regulator of normal and malignant hematopoietic development.


Subject(s)
1-Alkyl-2-acetylglycerophosphocholine Esterase/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Leukemia, Myeloid/pathology , Leukemia, Myeloid/physiopathology , Microtubule-Associated Proteins/physiology , 1-Alkyl-2-acetylglycerophosphocholine Esterase/antagonists & inhibitors , 1-Alkyl-2-acetylglycerophosphocholine Esterase/deficiency , 1-Alkyl-2-acetylglycerophosphocholine Esterase/genetics , Animals , Carcinogenesis , Cell Division , Cell Line, Tumor , Female , Hematopoiesis , Humans , K562 Cells , Leukemia, Myeloid/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/deficiency , Microtubule-Associated Proteins/genetics , Phenotype , Pregnancy , Spindle Apparatus/pathology
11.
Cancer Cell ; 21(2): 140-2, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22340585

ABSTRACT

Cancer stem cells lie at the root of chronic myelogenous leukemia (CML) and mediate its continued growth. Their resistance to current therapies results in an inability to eradicate the disease. In this issue of Cancer Cell, Li et al. identify SIRT1 as a new target for eliminating CML cancer stem cells.

12.
Nature ; 466(7307): 765-8, 2010 Aug 05.
Article in English | MEDLINE | ID: mdl-20639863

ABSTRACT

Chronic myelogenous leukaemia (CML) can progress from a slow growing chronic phase to an aggressive blast crisis phase, but the molecular basis of this transition remains poorly understood. Here we have used mouse models of CML to show that disease progression is regulated by the Musashi-Numb signalling axis. Specifically, we find that the chronic phase is marked by high levels of Numb expression whereas the blast crisis phase has low levels of Numb expression, and that ectopic expression of Numb promotes differentiation and impairs advanced-phase disease in vivo. As a possible explanation for the decreased levels of Numb in the blast crisis phase, we show that NUP98-HOXA9, an oncogene associated with blast crisis CML, can trigger expression of the RNA-binding protein Musashi2 (Msi2), which in turn represses Numb. Notably, loss of Msi2 restores Numb expression and significantly impairs the development and propagation of blast crisis CML in vitro and in vivo. Finally we show that Msi2 expression is not only highly upregulated during human CML progression but is also an early indicator of poorer prognosis. These data show that the Musashi-Numb pathway can control the differentiation of CML cells, and raise the possibility that targeting this pathway may provide a new strategy for the therapy of aggressive leukaemias.


Subject(s)
Cell Differentiation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , RNA-Binding Proteins/metabolism , Animals , Blast Crisis/genetics , Blast Crisis/metabolism , Blast Crisis/pathology , Cell Differentiation/genetics , Disease Progression , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prognosis , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Receptor, Notch1/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...