Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters











Publication year range
1.
Cytotherapy ; 24(11): 1087-1094, 2022 11.
Article in English | MEDLINE | ID: mdl-36050244

ABSTRACT

BACKGROUND: Natural killer (NK) cell genome editing promises to enhance the innate and alloreactive anti-tumor potential of NK cell adoptive transfer. DNA transposons are versatile non-viral gene vectors now being adapted to primary NK cells, representing important tools for research and clinical product development. AIMS AND METHODS: We set out to generate donor-derived, primary chimeric antigen receptor (CAR)-NK cells by combining the TcBuster transposon system with Epstein-Barr virus-transformed lymphoblastoid feeder cell-mediated activation and expansion. RESULTS: This approach allowed for clinically relevant NK-cell expansion capability and CAR expression, which was further enhanced by immunomagnetic selection based on binding to the CAR target protein.The resulting CAR-NK cells targeting the myeloid associated antigen CLL-1 efficiently targeted CLL-1-positive AML cell lines and primary AML populations, including a population enriched for leukemia stem cells. Subsequently, concurrent delivery of CRISPR/Cas9 cargo was applied to knockout the NK cell cytokine checkpoint cytokine-inducible SH2-containing protein (CIS, product of the CISH gene), resulting in enhanced cytotoxicity and an altered NK cell phenotype. CONCLUSIONS: This report contributes a promising application of transposon engineering to donor-derived NK cells and emphasizes the importance of feeder mediated NK cell activation and expansion to current protocols.


Subject(s)
Epstein-Barr Virus Infections , Leukemia, Lymphocytic, Chronic, B-Cell , Leukemia, Myeloid, Acute , Receptors, Chimeric Antigen , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , DNA Transposable Elements/genetics , Gene Editing , Herpesvirus 4, Human/genetics , Humans , Immunotherapy, Adoptive/methods , Killer Cells, Natural , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Receptors, Chimeric Antigen/genetics , Receptors, Chimeric Antigen/metabolism
2.
Cell Stem Cell ; 23(2): 181-192.e5, 2018 Aug 02.
Article in English | MEDLINE | ID: mdl-30082067

ABSTRACT

Chimeric antigen receptors (CARs) significantly enhance the anti-tumor activity of immune effector cells. Although most studies have evaluated CAR expression in T cells, here we evaluate different CAR constructs that improve natural killer (NK) cell-mediated killing. We identified a CAR containing the transmembrane domain of NKG2D, the 2B4 co-stimulatory domain, and the CD3ζ signaling domain to mediate strong antigen-specific NK cell signaling. NK cells derived from human iPSCs that express this CAR (NK-CAR-iPSC-NK cells) have a typical NK cell phenotype and demonstrate improved anti-tumor activity compared with T-CAR-expressing iPSC-derived NK cells (T-CAR-iPSC-NK cells) and non-CAR-expressing cells. In an ovarian cancer xenograft model, NK-CAR-iPSC-NK cells significantly inhibited tumor growth and prolonged survival compared with PB-NK cells, iPSC-NK cells, or T-CAR-iPSC-NK cells. Additionally, NK-CAR-iPSC-NK cells demonstrate in vivo activity similar to that of T-CAR-expressing T cells, although with less toxicity. These NK-CAR-iPSC-NK cells now provide standardized, targeted "off-the-shelf" lymphocytes for anti-cancer immunotherapy.


Subject(s)
Cell Engineering , Induced Pluripotent Stem Cells/cytology , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Receptors, Chimeric Antigen/metabolism , Animals , Cells, Cultured , Female , Humans , K562 Cells , Killer Cells, Natural/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Ovarian Neoplasms/genetics , Receptors, Chimeric Antigen/genetics
3.
Methods Mol Biol ; 1441: 277-84, 2016.
Article in English | MEDLINE | ID: mdl-27177674

ABSTRACT

Natural killer (NK) cells are an attractive cell population for immunotherapy. Adoptive transfer of NK cells has been tested in multiple clinical trials including acute myeloid leukemia (AML) and ovarian cancer, although limitations do exist especially for treatment of solid tumors. In order to overcome these limitations, mouse xenograft models are needed for evaluation of various NK cell populations, as well as routes of NK cell administration. Here, we describe the methods used for the establishment of an intraperitoneal (ip) ovarian cancer mouse xenograft model with ip delivery of NK cells. This model has been successfully employed with multiple ovarian cell lines and could be applied to other tumor models where the tumor's primary location is in the peritoneal cavity. It is also compatible with multiple routes of NK cell administration. Bioluminescent imaging for monitoring tumor formation and response provides for easy visualization of NK cell tumor inhibition. This xenograft model is superior to other models because the tumor is implanted into the same physiological space where ovarian cancer is found, which allows for improved mimicking of actual disease.


Subject(s)
Interleukin-2/metabolism , Killer Cells, Natural/transplantation , Ovarian Neoplasms/therapy , Animals , Cell Line, Tumor , Cytotoxicity, Immunologic , Female , Humans , Immunotherapy , Injections, Intraperitoneal , Killer Cells, Natural/immunology , Luminescent Measurements , Mice , Ovarian Neoplasms/immunology , Xenograft Model Antitumor Assays
4.
Stem Cells ; 34(1): 93-101, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26503833

ABSTRACT

Natural killer (NK) cells can provide effective immunotherapy for ovarian cancer. Here, we evaluated the ability of NK cells isolated from peripheral blood (PB) and NK cells derived from induced pluripotent stem cell (iPSC) to mediate killing of ovarian cancer cells in a mouse xenograft model. A mouse xenograft model was used to evaluate the intraperitoneal delivery of three different NK cell populations: iPSC-derived NK cells, PB-NK cells that had been activated and expanded in long-term culture, and overnight activated PB-NK cells that were isolated through CD3/CD19 depletion of PB B and T cells. Bioluminescent imaging was used to monitor tumor burden of luciferase expressing tumor lines. Tumors were allowed to establish prior to administering NK cells via intraperitoneal injection. These studies demonstrate a single dose of any of the three NK cell populations significantly reduced tumor burden. When mice were given three doses of either iPSC-NK cells or expanded PB-NK cells, the median survival improved from 73 days in mice untreated to 98 and 97 days for treated mice, respectively. From these studies, we conclude iPSC-derived NK cells mediate antiovarian cancer killing at least as well as PB-NK cells, making these cells a viable resource for immunotherapy for ovarian cancer. Due to their ability to be easily differentiated into NK cells and their long-term expansion potential, iPSCs can be used to produce large numbers of well-defined NK cells that can be banked and used to treat a large number of patients including treatment with multiple doses if necessary.


Subject(s)
Induced Pluripotent Stem Cells/cytology , Killer Cells, Natural/cytology , Ovarian Neoplasms/therapy , Animals , Cell Line, Tumor , Cell Proliferation , Female , Flow Cytometry , Humans , Immunotherapy , Mice , Ovarian Neoplasms/blood
5.
Front Immunol ; 6: 195, 2015.
Article in English | MEDLINE | ID: mdl-25972867

ABSTRACT

Natural killer (NK) cells represent an attractive lymphocyte population for cancer immunotherapy due to their ability to lyse tumor targets without prior sensitization and without need for human leukocyte antigens-matching. Chimeric antigen receptors (CARs) are able to enhance lymphocyte targeting and activation toward diverse malignancies. CARs consist of an external recognition domain (typically a small chain variable fragment) directed at a specific tumor antigen that is linked with one or more intracellular signaling domains that mediate lymphocyte activation. Most CAR studies have focused on their expression in T cells. However, use of CARs in NK cells is starting to gain traction because they provide a method to redirect these cells more specifically to target refractory cancers. CAR-mediated anti-tumor activity has been demonstrated using NK cell lines, as well as NK cells isolated from peripheral blood, and NK cells produced from human pluripotent stem cells. This review will outline the CAR constructs that have been reported in NK cells with a focus on comparing the use of different signaling domains in combination with other co-activating domains.

6.
Cytotherapy ; 15(10): 1297-306, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23993303

ABSTRACT

BACKGROUND AIMS: There is an urgent need for novel therapeutic strategies for relapsed ovarian cancer. Dramatic clinical anti-tumor effects have been observed with interleukin (IL)-2 activated natural killer (NK) cells; however, intravenous delivery of NK cells in patients with ovarian cancer has not been successful in ameliorating disease. We investigated in vivo engraftment of intraperitoneally (IP) delivered NK cells in an ovarian cancer xenograft model to determine if delivery mode can affect tumor cell killing and circumvent lack of NK cell expansion. METHODS: An ovarian cancer xenograft mouse model was established to evaluate efficacy of IP-delivered NK cells. Tumor burden was monitored by bioluminescent imaging of luciferase-expressing ovarian cancer cells. NK cell persistence, tumor burden and NK cell trafficking were evaluated. Transplanted NK cells were evaluated by flow cytometry and cytotoxicity assays. RESULTS: IP delivery of human NK cells plus cytokines led to high levels of circulating NK and was effective in clearing intraperitoneal ovarian cancer burden in xenografted mice. NK cells remained within the peritoneal cavity 54 days after injection and had markers of maturation. Additionally, surviving NK cells were able to kill ovarian cancer cells at a rate similar to pre-infusion levels, supporting that in vivo functionality of human NK cells can be maintained after IP infusion. CONCLUSIONS: IP delivery of NK cells leads to stable engraftment and antitumor response in an ovarian cancer xenograft model. These data support further pre-clinical and clinical evaluation of IP delivery of allogeneic NK cells in ovarian cancer.


Subject(s)
Adenocarcinoma/therapy , Cancer Vaccines , Immunotherapy, Adoptive/methods , Killer Cells, Natural/transplantation , Ovarian Neoplasms/therapy , Peritoneal Cavity/pathology , Adenocarcinoma/immunology , Animals , Antigens, Differentiation/metabolism , Cell Proliferation , Cell Survival , Cytotoxicity, Immunologic , Female , Humans , Injections, Intraperitoneal , Interleukin-2/immunology , K562 Cells , Killer Cells, Natural/immunology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Ovarian Neoplasms/immunology , Recurrence , Tumor Burden , Xenograft Model Antitumor Assays
7.
Mol Pharmacol ; 84(2): 236-43, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23696245

ABSTRACT

Drug resistance is a serious challenge in cancer treatment and can be acquired through multiple mechanisms. These molecular changes may introduce varied extents of resistance to different therapies and need to be characterized for optimal therapy choice. A recently discovered small molecule, ethyl-2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate) (CXL017), reveals selective cytotoxicity toward drug-resistant leukemia. A drug-resistant acute myeloid leukemia cell line, HL60/MX2, also failed to acquire resistance to CXL017 upon chronic exposure and regained sensitivity toward standard therapies. In this study, we investigated the mechanisms responsible for HL60/MX2 cells' drug resistance and the molecular basis for its resensitization. Results show that the HL60/MX2 cell line has an elevated level of Mcl-1 protein relative to the parental cell line, HL60, and its resensitized cell line, HL60/MX2/CXL017, whereas it has a reduced level of topoisomerase IIß. Mcl-1 overexpression in HL60/MX2 cells is mainly regulated through phospho-extracellular signal-regulated protein kinases 1 and 2-mediated Mcl-1 stabilization, whereas the reduction of topoisomerase IIß in HL60/MX2 cells is controlled through genetic downregulation. Upregulating Mcl-1 introduces multidrug resistance to standard therapies, whereas its downregulation results in significant cell death. Downregulating topoisomerase IIß confers resistance specifically to mitoxantrone, not to other topoisomerase II inhibitors. Overall, these data suggest that Mcl-1 overexpression is a critical determinant for cross-resistance to standard therapies, whereas topoisomerase IIß downregulation is specific to mitoxantrone resistance.


Subject(s)
DNA Topoisomerases, Type II/biosynthesis , DNA-Binding Proteins/biosynthesis , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/metabolism , Mitoxantrone/pharmacology , Myeloid Cell Leukemia Sequence 1 Protein/biosynthesis , Benzopyrans/pharmacology , Cell Death/drug effects , Cell Death/genetics , Cell Line, Tumor , DNA Topoisomerases, Type II/genetics , DNA Topoisomerases, Type II/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Down-Regulation , Drug Resistance, Multiple , Drug Resistance, Neoplasm , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/genetics , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Up-Regulation/drug effects
8.
Stem Cells Transl Med ; 2(4): 274-83, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23515118

ABSTRACT

Adoptive transfer of antitumor lymphocytes has gained intense interest in the field of cancer therapeutics over the past two decades. Human natural killer (NK) cells are a promising source of lymphocytes for anticancer immunotherapy. NK cells are part of the innate immune system and exhibit potent antitumor activity without need for human leukocyte antigen matching and without prior antigen exposure. Moreover, the derivation of NK cells from pluripotent stem cells could provide an unlimited source of lymphocytes for off-the-shelf therapy. To date, most studies on hematopoietic cell development from human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) have used incompletely defined conditions and been on a limited scale. Here, we have used a two-stage culture system to efficiently produce NK cells from hESCs and iPSCs in the absence of cell sorting and without need for xenogeneic stromal cells. This novel combination of embryoid body formation using defined conditions and membrane-bound interleukin 21-expressing artificial antigen-presenting cells allows production of mature and functional NK cells from several different hESC and iPSC lines. Although different hESC and iPSC lines had varying efficiencies in hematopoietic development, all cell lines tested could produce functional NK cells. These methods can be used to generate enough cytotoxic NK cells to treat a single patient from fewer than 250,000 input hESCs/iPSCs. Additionally, this strategy provides a genetically amenable platform to study normal NK cell development and education in vitro.


Subject(s)
Cell Culture Techniques/methods , Induced Pluripotent Stem Cells/cytology , Killer Cells, Natural/cytology , Neoplasms/therapy , Animals , Antigen-Presenting Cells/cytology , Cell Line , Cell Proliferation , Embryoid Bodies/cytology , Feeder Cells/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Immunotherapy , Mice , Neoplasms/immunology , Stromal Cells/cytology
9.
ACS Chem Biol ; 8(2): 327-35, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23102022

ABSTRACT

Multidrug resistance (MDR) is a major hurdle in the treatment of cancer, and there is a pressing need for new therapies. We have recently developed ethyl 2-amino-6-(3,5-dimethoxyphenyl)-4-(2-ethoxy-2-oxoethyl)-4H-chromene-3-carboxylate (CXL017), derived from a dual inhibitor of Bcl-2 and SERCA proteins, sHA 14-1, with selective cytotoxicity toward MDR cancer cell lines in vitro. In this study, we present new evidence for its therapeutic potential in treatment of MDR cancers and offer mechanistic insights toward its preferential targeting of drug-resistant cancer. CXL017 selectively suppressed the growth of tumors derived from the MDR cancer cell line, HL60/MX2, in vivo. In addition, even after chronic exposure to CXL017, HL60/MX2 failed to develop stable resistance to CXL017, whereas it acquired >2000-fold resistance to cytarabine (Ara-C), the major first-line chemotherapy for the treatment of acute myeloid leukemia (AML). Remarkably, instead of acquiring further cross-resistance, HL60/MX2 cells exposed to CXL017 were resensitized to standard therapies (10- to 100-fold). Western blotting analyses revealed that CXL017 exposure significantly down-regulated Mcl-1 and Bax and up-regulated Noxa, Bim, Bcl-X(L), SERCA2, and SERCA3 proteins, along with a reduction in endoplasmic reticulum (ER) calcium content. Given the well-established functions of Bcl-2 family proteins and ER calcium in drug resistance, our results suggest that the down-regulation of Mcl-1 and the up-regulation of Noxa and Bim along with the decrease in ER calcium content are likely responsible for CXL017-induced resensitization of MDR cancer cells. These data also demonstrate the unique potential of CXL017 to overcome MDR in cancer treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Benzopyrans/pharmacology , Cytarabine/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Leukemia, Myeloid, Acute/drug therapy , Antineoplastic Combined Chemotherapy Protocols/chemistry , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzopyrans/chemistry , Cell Survival/drug effects , Cytarabine/chemistry , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , HL-60 Cells , Humans , Leukemia, Myeloid, Acute/pathology , Structure-Activity Relationship , Tumor Cells, Cultured
10.
J Med Chem ; 55(11): 5566-81, 2012 Jun 14.
Article in English | MEDLINE | ID: mdl-22582991

ABSTRACT

Multidrug resistance (MDR) against standard therapies poses a serious challenge in cancer treatment, and there is a clinical need for new anticancer agents that would selectively target MDR malignancies. Our previous studies have identified a 4H-chromene system, CXL017 (4) as an example, that can preferentially kill MDR cancer cells. To further improve its potency, we have performed detailed structure-activity relationship (SAR) studies at the 3, 4, and 6 positions of the 4H-chromene system. The results reveal that the 3 and 4 positions prefer rigid and hydrophobic functional groups while the 6 position prefers a meta or para-substituted aryl functional group and the substituent should be small and hydrophilic. We have also identified and characterized nine MDR cancer cells that acquire MDR through different mechanisms and demonstrated the scope of our new lead, 9g, to selectively target different MDR cancers, which holds promise to help manage MDR in cancer treatment.


Subject(s)
Antineoplastic Agents/chemical synthesis , Benzopyrans/chemical synthesis , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Benzopyrans/chemistry , Benzopyrans/pharmacology , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , Sarcoplasmic Reticulum Calcium-Transporting ATPases/metabolism , Structure-Activity Relationship
SELECTION OF CITATIONS
SEARCH DETAIL