Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49
Filter
1.
J Immunother Cancer ; 12(2)2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38417916

ABSTRACT

BACKGROUND: The antitumor activity of natural killer (NK) cells can be enhanced by specific targeting with therapeutic antibodies that trigger antibody-dependent cell-mediated cytotoxicity (ADCC) or by genetic engineering to express chimeric antigen receptors (CARs). Despite antibody or CAR targeting, some tumors remain resistant towards NK cell attack. While the importance of ICAM-1/LFA-1 interaction for natural cytotoxicity of NK cells is known, its impact on ADCC induced by the ErbB2 (HER2)-specific antibody trastuzumab and ErbB2-CAR-mediated NK cell cytotoxicity against breast cancer cells has not been investigated. METHODS: Here we used NK-92 cells expressing high-affinity Fc receptor FcγRIIIa in combination with trastuzumab or ErbB2-CAR engineered NK-92 cells (NK-92/5.28.z) as well as primary human NK cells combined with trastuzumab or modified with the ErbB2-CAR and tested cytotoxicity against cancer cells varying in ICAM-1 expression or alternatively blocked LFA-1 on NK cells. Furthermore, we specifically stimulated Fc receptor, CAR and/or LFA-1 to study their crosstalk at the immunological synapse and their contribution to degranulation and intracellular signaling in antibody-targeted or CAR-targeted NK cells. RESULTS: Blockade of LFA-1 or absence of ICAM-1 significantly reduced cell killing and cytokine release during trastuzumab-mediated ADCC against ErbB2-positive breast cancer cells, but not so in CAR-targeted NK cells. Pretreatment with 5-aza-2'-deoxycytidine induced ICAM-1 upregulation and reversed NK cell resistance in ADCC. Trastuzumab alone did not sufficiently activate NK cells and required additional LFA-1 co-stimulation, while activation of the ErbB2-CAR in CAR-NK cells induced efficient degranulation independent of LFA-1. Total internal reflection fluorescence single molecule imaging revealed that CAR-NK cells formed an irregular immunological synapse with tumor cells that excluded ICAM-1, while trastuzumab formed typical peripheral supramolecular activation cluster (pSMAC) structures. Mechanistically, the absence of ICAM-1 did not affect cell-cell adhesion during ADCC, but rather resulted in decreased signaling via Pyk2 and ERK1/2, which was intrinsically provided by CAR-mediated targeting. Furthermore, while stimulation of the inhibitory NK cell checkpoint molecule NKG2A markedly reduced FcγRIIIa/LFA-1-mediated degranulation, retargeting by CAR was only marginally affected. CONCLUSIONS: Downregulation of ICAM-1 on breast cancer cells is a critical escape mechanism from trastuzumab-triggered ADCC. In contrast, CAR-NK cells are able to overcome cancer cell resistance caused by ICAM-1 reduction, highlighting the potential of CAR-NK cells in cancer immunotherapy.


Subject(s)
Breast Neoplasms , Receptors, Chimeric Antigen , Humans , Female , Intercellular Adhesion Molecule-1 , Receptors, Chimeric Antigen/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Down-Regulation , Tumor Escape , Cell Line, Tumor , Killer Cells, Natural , Trastuzumab/pharmacology , Antibodies , Receptors, Fc/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism
2.
Cytotherapy ; 25(5): 451-457, 2023 05.
Article in English | MEDLINE | ID: mdl-36610812

ABSTRACT

The NK-92 cell line, established in 1992, mirrors all the characteristics of highly active blood natural killer (NK) cells but with much broader and greater cytotoxicity. The cell line was established from the blood cells of a patient with lymphoma and has been made widely available for research since it was deposited into the American Type Culture Collection in 1998. The worldwide distribution of NK-92 cells has led to a plethora of scientific discoveries that have greatly increased the understanding of NK-cell biology. NK-92 cells also have been developed for clinical use, overcoming the challenges of obtaining and expanding NK cells from donor or patient blood. More than 100 patients with cancer have now been treated all over the world with unmodified or genetically engineered NK-92 cells. Modified cells include high-affinity Fc-receptor expressing NK-92 cells (haNKR) and various chimeric antigen receptor targeted haNK cells (t-haNKTM). Infusions of either unmodified or modified NK-92 cells have been reported to be safe and efficacious, leading in some cases to disease remission even in patients who had failed multiple previous lines of therapy. It is the purpose of this review to distill the plethora of scientific data on NK-92 cells and its genetic variants, highlighting relevant experimental findings that have contributed to a better understanding of NK cell biology and summarize the therapeutic potential of these cells for treatment of cancer and infections.


Subject(s)
Lymphoma , Receptors, Chimeric Antigen , Humans , Cell Line, Tumor , Killer Cells, Natural , Immunotherapy , Receptors, Chimeric Antigen/metabolism , Immunotherapy, Adoptive
3.
Front Vet Sci ; 10: 1285354, 2023.
Article in English | MEDLINE | ID: mdl-38234984

ABSTRACT

Compared with the options available to human patients with cancer, treatment choices for dogs are often more limited. Chemotherapy is frequently the first-line treatment for many cancers. However, its efficacy can be limited, and its side effects can affect the quality of the remaining life. This paper briefly summarizes the experience with Viscum album L. (mistletoe) extract in human patients as a stipulation to consider treatment with mistletoe extract for canines with cancer. The mistletoe extract contains -among others - lectins and viscotoxins that have documented anti-proliferative effect on cancer cells as well as immune-stimulatory function. Importantly, it also improves the well-being of patients with cancer due to its lectin ML-1 content, which can trigger the release of endorphins. Being cross-reactive with canine cells and having a relatively low side effect profile, it raises the question of whether mistletoe preparations might be considered as part of the treatment approach for dogs with cancer.

4.
Cytotherapy ; 24(8): 827-834, 2022 08.
Article in English | MEDLINE | ID: mdl-35400595

ABSTRACT

BACKGROUND AIMS: Chimeric antigen receptor (CAR) T-cell therapy can be associated with significant toxicities. CAR-engineered natural killer (NK) cells provide a safer alternative while maintaining anti-tumor effects. Activated NK (aNK) cells are a clinical-grade cellular product obtained from the NK-92 cell line that have demonstrated both safety and potent cytotoxicity toward a wide range of cancers in phase 1 trials. Genetically engineered variants of aNK cells expressing a high-affinity Fc receptor (haNK) or co-expressing a CAR (t-haNK) are currently in phase 1/2 clinical trials. A key factor in the efficacy of cellular immunotherapies is biodistribution and tumor infiltration, which affect the local effector:target ratio. The chemokines CCL19 and CCL21 can drive recruitment of CCR7 receptor-expressing immune cells to secondary lymphoid organs. METHODS: Since NK-92 cells do not spontaneously express CCR7, clinical-grade aNK cells were transfected with a non-viral vector containing the CCR7 receptor, an anti-CD19 CAR and a high-affinity CD16 Fc receptor. RESULTS: CCR7-engineered CD19 t-haNK showed significant migration in vitro toward K562 cells engineered to secrete CCL19. This observation was confirmed in a NOD.Cg-PrkdcscidIl2rgtm1Wjl/SzJ (NSG) mouse model in which subcutaneous tumors of CCL19-expressing K562 cells displayed a higher number of infiltrating CCR7_CD19 t-haNK cells than CCR7-negative CD19 t-haNK cells. In NSG mice inoculated either intravenously or subcutaneously with CCL19-secreting Raji cells, treatment with CCR7_CD19 t-haNK improved survival and tumor control compared with CD19 t-haNK or vehicle. CONCLUSIONS: Expression of CCR7 receptor by off-the-shelf t-haNK cells improves their homing toward lymph node chemokines both in vitro and in vivo, resulting in superior tumor control.


Subject(s)
Immunotherapy, Adoptive , Lymphoma , Receptors, CCR7 , Animals , Antigens, CD19 , Cell Line, Tumor , Chemokine CCL19/genetics , Chemokine CCL19/metabolism , Humans , Immunotherapy, Adoptive/methods , Killer Cells, Natural , Lymphoma/therapy , Mice , Mice, Inbred NOD , Receptors, CCR7/genetics , Receptors, CCR7/metabolism , Receptors, Chimeric Antigen/metabolism , Receptors, Fc/metabolism , Tissue Distribution
5.
Front Immunol ; 12: 665784, 2021.
Article in English | MEDLINE | ID: mdl-34421888

ABSTRACT

Despite all good intentions, dogs are still running behind humans in effective cancer immunotherapies. The more effective treatments in humans, like infusions of CAR-T and NK-cells are not broadly pursued for canines due to significant costs, the rather complicated logistics and the lack of targetable surface antigens. Monoclonal antibodies are challenging to develop considering the limited knowledge about canine target antigens and about their mode of action. Although immunogenic vaccines could be less costly, this approach is hampered by the fact that cancer by itself is immuno-suppressive and any preceding chemotherapy may suppress any clinically meaningful immune response. This review - rather than providing a comprehensive listing of all available immunotherapies for dogs, aims at pointing out the issues that are holding back this field but which hopefully can be addressed so that dogs can "catch up" with what is available to humans.


Subject(s)
Dog Diseases/therapy , Immunotherapy , Neoplasms/therapy , Animals , Dogs , Humans
6.
J Immunother Cancer ; 9(1)2021 01.
Article in English | MEDLINE | ID: mdl-33468562

ABSTRACT

BACKGROUND: On encountering a susceptible target, natural killer (NK) cells mediate cytotoxicity through highly regulated steps of directed degranulation. Cytotoxic granules converge at the microtubule organizing center and are polarized toward the immunological synapse (IS), followed by granule exocytosis. NK cell retargeting by chimeric antigen receptors (CARs) or mAbs represents a promising strategy for overcoming tumor cell resistance. However, little is known about the lytic granule dynamics of such retargeted NK cells toward NK-cell-resistant tumors. METHODS: Here, we used spinning disk confocal microscopy for live-cell imaging to analyze granule-mediated NK cell cytotoxicity in ErbB2-targeted CAR-expressing NK-92 cells (NK-92/5.28.z) and high-affinity FcR transgenic NK-92 cells plus Herceptin toward ErbB2-positive breast cancer cells (MDA-MB-453), which are resistant to parental NK-92. RESULTS: Unmodified NK-92 cells cocultured with resistant cancer cells showed stable conjugate formation and granule clustering, but failed to polarize granules to the IS. In contrast, retargeting by CAR or FcR+Herceptin toward the MDA-MB-453 cells enabled granule polarization to the IS, resulting in highly effective cytotoxicity. We found that in NK-92 the phosphoinositide 3-kinase pathway was activated after contact with resistant MDA-MB-453, while phospholipase C-γ (PLCγ) and mitogen-activated protein kinase (MEK)/extracellular signal-regulated kinase (ERK) were not activated. In contrast, retargeting by CAR or antibody-dependent cell-mediated cytotoxicity (ADCC) provided the missing PLCγ and MEK/ERK signals. CONCLUSIONS: These observations suggest that NK cells can create conjugates with resistant cancer cells and respond by granule clustering, but the activation signals are insufficient to induce granule polarization and consequent release of lytic enzymes. Retargeting by CAR and/or the FcR/mAb (ADCC) axis provide the necessary signals, leading to granule polarization and thereby overcoming tumor cell resistance.


Subject(s)
Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Killer Cells, Natural/cytology , Receptor, ErbB-2/immunology , Trastuzumab/pharmacology , Antibody-Dependent Cell Cytotoxicity , Breast Neoplasms/therapy , Cell Line, Tumor , Coculture Techniques , Cytotoxicity, Immunologic , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunotherapy, Adoptive/methods , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/transplantation , Mitogen-Activated Protein Kinases/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Receptors, Chimeric Antigen/metabolism , Signal Transduction/drug effects
7.
Vet Sci ; 5(4)2018 Dec 06.
Article in English | MEDLINE | ID: mdl-30563208

ABSTRACT

Infusions with immune cells, such as lymphocytes or natural killer (NK) cells, represent one of several modalities of immunotherapy. In human patients with advanced B-cell leukemia or lymphoma, infusions with chimeric antigen receptor (CAR) T-lymphocytes have shown promising responses. However, the scientific and clinical development of cell-based therapies for dogs, who get cancer of similar types as humans, is lagging behind. One reason is that immune cells and their functionality in dogs are less well characterized, largely due a lack of canine-specific reagents to detect surface markers, and specific cytokines to isolate and expand their immune cells. This review summarizes the current status of canine cancer immunotherapies, with focus on autologous and allogeneic T-lymphocytes, as well as NK cells, and discusses potential initiatives that would allow therapies with canine immune cells to "catch up" with the advances in humans.

8.
Front Immunol ; 9: 133, 2018.
Article in English | MEDLINE | ID: mdl-29459862

ABSTRACT

A number of excellent reviews on the potential of canine cancer immunotherapy are available, but many extrapolate from observations in humans when in fact only very few immunotherapies have been developed for canines that have shown efficacy in well-designed studies. Pharmaceutical and biotech companies are aware that the market for more expensive immunotherapies in canines is limited resulting in limited funding for clinical trials. However, dogs and other pets deserve access to this new form of cancer therapy. The purpose of this brief review is to summarize the current status of available immunotherapies for dogs and their near-term prospects, provided we can effectively translate discoveries and progress in humans to canines.


Subject(s)
Dog Diseases/therapy , Immunotherapy , Neoplasms/therapy , Animals , Dogs , Humans
9.
Cytotherapy ; 19(2): 235-249, 2017 02.
Article in English | MEDLINE | ID: mdl-27887866

ABSTRACT

BACKGROUND AIMS: Natural killer (NK) cells can rapidly respond to transformed and stressed cells and represent an important effector cell type for adoptive immunotherapy. In addition to donor-derived primary NK cells, continuously expanding cytotoxic cell lines such as NK-92 are being developed for clinical applications. METHODS: To enhance their therapeutic utility for the treatment of B-cell malignancies, we engineered NK-92 cells by lentiviral gene transfer to express chimeric antigen receptors (CARs) that target CD19 and contain human CD3ζ (CAR 63.z), composite CD28-CD3ζ or CD137-CD3ζ signaling domains (CARs 63.28.z and 63.137.z). RESULTS: Exposure of CD19-positive targets to CAR NK-92 cells resulted in formation of conjugates between NK and cancer cells, NK-cell degranulation and selective cytotoxicity toward established B-cell leukemia and lymphoma cells. Likewise, the CAR NK cells displayed targeted cell killing of primary pre-B-ALL blasts that were resistant to parental NK-92. Although all three CAR NK-92 cell variants were functionally active, NK-92/63.137.z cells were less effective than NK-92/63.z and NK-92/63.28.z in cell killing and cytokine production, pointing to differential effects of the costimulatory CD28 and CD137 domains. In a Raji B-cell lymphoma model in NOD-SCID IL2R γnull mice, treatment with NK-92/63.z cells, but not parental NK-92 cells, inhibited disease progression, indicating that selective cytotoxicity was retained in vivo. CONCLUSIONS: Our data demonstrate that it is feasible to generate CAR-engineered NK-92 cells with potent and selective antitumor activity. These cells may become clinically useful as a continuously expandable off-the-shelf cell therapeutic agent.


Subject(s)
Cytotoxicity, Immunologic , Killer Cells, Natural , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Lymphoma/therapy , Recombinant Fusion Proteins/metabolism , Animals , Antigens, CD19/immunology , B-Lymphocytes/immunology , CD3 Complex/genetics , CD3 Complex/metabolism , Cell Line, Tumor , Cell Proliferation , Cytotoxicity, Immunologic/genetics , Epitopes/genetics , HEK293 Cells , Humans , Immunotherapy, Adoptive/methods , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Leukemia, Lymphocytic, Chronic, B-Cell/immunology , Lymphocyte Activation/genetics , Lymphoma/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Male , Mice , Mice, Inbred NOD , Mice, SCID , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 9/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
10.
Oncotarget ; 7(52): 86359-86373, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27861156

ABSTRACT

Natural killer (NK) cells are known to play a role in mediating innate immunity, in enhancing adaptive immune responses, and have been implicated in mediating anti-tumor responses via antibody-dependent cell-mediated cytotoxicity (ADCC) by reactivity of CD16 with the Fc region of human IgG1 antibodies. The NK-92 cell line, derived from a lymphoma patient, has previously been well characterized and adoptive transfer of irradiated NK-92 cells has demonstrated safety and shown preliminary evidence of clinical benefit in cancer patients. The NK-92 cell line, devoid of CD16, has now been engineered to express the high affinity (ha) CD16 V158 FcγRIIIa receptor, as well as engineered to express IL-2; IL-2 has been shown to replenish the granular stock of NK cells, leading to enhanced perforin- and granzyme-mediated lysis of tumor cells. The studies reported here show high levels of granzyme in haNK cells, and demonstrate the effects of irradiation of haNK cells on multiple phenotypic markers, viability, IL-2 production, and lysis of a spectrum of human tumor cells. Studies also compare endogenous irradiated haNK lysis of tumor cells with that of irradiated haNK-mediated ADCC using cetuximab, trastuzumab and pertuzumab monoclonal antibodies. These studies thus provide the rationale for the potential use of irradiated haNK cells in adoptive transfer studies for a range of human tumor types. Moreover, since only approximately 10% of humans are homozygous for the high affinity V CD16 allele, these studies also provide the rationale for the use of irradiated haNK cells in combination with IgG1 anti-tumor monoclonal antibodies.


Subject(s)
Adoptive Transfer , Granzymes/immunology , Killer Cells, Natural/immunology , Receptors, IgG/genetics , Animals , Antibody-Dependent Cell Cytotoxicity , B7-H1 Antigen/analysis , Cell Line, Tumor , GPI-Linked Proteins/genetics , GPI-Linked Proteins/immunology , Genetic Engineering , Humans , Immunoglobulin G/therapeutic use , Interleukin-2/genetics , Killer Cells, Natural/enzymology , Killer Cells, Natural/radiation effects , Male , Mice , Middle Aged , Receptors, IgG/immunology
11.
Front Immunol ; 7: 91, 2016.
Article in English | MEDLINE | ID: mdl-27014270

ABSTRACT

Natural killer (NK) cells are potent cytotoxic effector cells for cancer therapy and potentially for severe viral infections. However, there are technical challenges to obtain sufficient numbers of functionally active NK cells from a patient's blood since they represent only 10% of the lymphocytes and are often dysfunctional. The alternative is to obtain cells from a healthy donor, which requires depletion of the allogeneic T cells to prevent graft-versus-host reactions. Cytotoxic cell lines have been established from patients with clonal NK-cell lymphoma. Those cells can be expanded in culture in the presence of IL-2. Except for the NK-92 cell line, though, none of the other six known NK cell lines has consistently and reproducibly shown high antitumor cytotoxicity. Only NK-92 cells can easily be genetically manipulated to recognize specific tumor antigens or to augment monoclonal antibody activity through antibody-dependent cellular cytotoxicity. NK-92 is also the only cell line product that has been infused into patients with advanced cancer with clinical benefit and minimal side effects.

12.
J Cell Mol Med ; 20(7): 1287-94, 2016 07.
Article in English | MEDLINE | ID: mdl-27008316

ABSTRACT

Many B-cell acute and chronic leukaemias tend to be resistant to killing by natural killer (NK) cells. The introduction of chimeric antigen receptors (CAR) into T cells or NK cells could potentially overcome this resistance. Here, we extend our previous observations on the resistance of malignant lymphoblasts to NK-92 cells, a continuously growing NK cell line, showing that anti-CD19-CAR (αCD19-CAR) engineered NK-92 cells can regain significant cytotoxicity against CD19 positive leukaemic cell lines and primary leukaemia cells that are resistant to cytolytic activity of parental NK-92 cells. The 'first generation' CAR was generated from a scFv (CD19) antibody fragment, coupled to a flexible hinge region, the CD3ζ chain and a Myc-tag and cloned into a retrovirus backbone. No difference in cytotoxic activity of NK-92 and transduced αCD19-CAR NK-92 cells towards CD19 negative targets was found. However, αCD19-CAR NK-92 cells specifically and efficiently lysed CD19 expressing B-precursor leukaemia cell lines as well as lymphoblasts from leukaemia patients. Since NK-92 cells can be easily expanded to clinical grade numbers under current Good Manufactoring Practice (cGMP) conditions and its safety has been documented in several phase I clinical studies, treatment with CAR modified NK-92 should be considered a treatment option for patients with lymphoid malignancies.


Subject(s)
B-Lymphocytes/immunology , Genetic Engineering , Killer Cells, Natural/immunology , Lymphoma, B-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , Cell Line, Tumor , Cell Lineage , Cytotoxicity, Immunologic , Humans , Lymphoma, B-Cell/pathology , Retroviridae/metabolism , Single-Chain Antibodies/metabolism , Tissue Donors , Transfection
13.
Cancer Immunol Immunother ; 65(4): 485-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26559813

ABSTRACT

Natural killer (NK) cells are increasingly considered as immunotherapeutic agents in particular in the fight against cancers. NK cell therapies are potentially broadly applicable and, different from their T cell counterparts, do not cause graft-versus-host disease. Efficacy and clinical in vitro or in vivo expansion of primary NK cells will however always remain variable due to individual differences of donors or patients. Long-term storage of clinical NK cell lots to allow repeated clinical applications remains an additional challenge. In contrast, the established and well-characterized cell line NK-92 can be easily and reproducibly expanded from a good manufacturing practice (GMP)-compliant cryopreserved master cell bank. Moreover, no cost-intensive cell purification methods are required. To date, NK-92 has been intensively studied. The cells displayed superior cytotoxicity against a number of tumor types tested, which was confirmed in preclinical mouse studies. Subsequent clinical testing demonstrated safety of NK-92 infusions even at high doses. Despite the phase I nature of the trials conducted so far, some efficacy was noted, particularly against lung tumors. Furthermore, to overcome tumor resistance and for specific targeting, NK-92 has been engineered to express a number of different chimeric antigen receptors (CARs), including targeting, for example, CD19 or CD20 (anti-B cell malignancies), CD38 (anti-myeloma) or human epidermal growth factor receptor 2 (HER2; ErbB2; anti-epithelial cancers). The concept of an NK cell line as an allogeneic cell therapeutic produced 'off-the-shelf' on demand holds great promise for the development of effective treatments.


Subject(s)
Cytotoxicity, Immunologic , Immunotherapy, Adoptive/methods , Killer Cells, Natural/transplantation , Neoplasms/therapy , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Clinical Trials as Topic , Humans , Killer Cells, Natural/immunology , Neoplasms/immunology , Treatment Outcome
14.
Cytotherapy ; 17(3): 245-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25533934

ABSTRACT

BACKGROUND AIMS: Natural killer (NK) cells from peripheral or cord blood-especially if they are obtained from a human leukocyte antigen-mismatched (allogeneic) donor-are increasingly being considered for treatment of malignant diseases and to prevent or treat relapse after stem cell transplant. However, in addition to proving their efficacy, there are some more logistical and technical issues that must be addressed before NK cell infusions will be fully accepted by the medical community. METHODS: Issues include (i) the expansion of sufficient numbers of cells under conditions suitable, (ii) cryopreservation and (iii) optimization/standardization of shipping conditions if the cells are used at distant sites. Because the patient's own autologous cells usually are not fully functional because of inhibition by "self" major histocompatibility complex expression, better methods must be developed to target NK cells to tumor cells and overcome self-inhibition. RESULTS: Tumor-directed NK-cell therapy can be best accomplished through genetic engineering of NK cells expressing receptors for tumor antigens or combination with monoclonal antibodies that preferentially kill tumors through antibody-dependent cellular cytotoxicity. If allogeneic NK cells are used, T-lymphocytes in the cell collections that can cause acute graft-versus-host disease in the recipient must be removed. CONCLUSIONS: In addition to showing efficacy in clinical trials, the production of NK cells for treatment must be cost-effective to be eligible for reimbursement by third-party players.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Immunotherapy, Adoptive/methods , Killer Cells, Natural/transplantation , Neoplasms/therapy , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity/immunology , Cell Culture Techniques , Cell Line , Cell- and Tissue-Based Therapy/methods , Cryopreservation , Fetal Blood/cytology , Genetic Engineering , Graft vs Host Disease/immunology , HLA Antigens/immunology , Histocompatibility Antigens Class I/immunology , Humans , Killer Cells, Natural/immunology , Neoplasms/immunology , T-Lymphocytes/immunology
15.
Mol Ther ; 23(2): 330-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25373520

ABSTRACT

Natural killer (NK) cells are an important effector cell type for adoptive cancer immunotherapy. Similar to T cells, NK cells can be modified to express chimeric antigen receptors (CARs) to enhance antitumor activity, but experience with CAR-engineered NK cells and their clinical development is still limited. Here, we redirected continuously expanding and clinically usable established human NK-92 cells to the tumor-associated ErbB2 (HER2) antigen. Following GMP-compliant procedures, we generated a stable clonal cell line expressing a humanized CAR based on ErbB2-specific antibody FRP5 harboring CD28 and CD3ζ signaling domains (CAR 5.28.z). These NK-92/5.28.z cells efficiently lysed ErbB2-expressing tumor cells in vitro and exhibited serial target cell killing. Specific recognition of tumor cells and antitumor activity were retained in vivo, resulting in selective enrichment of NK-92/5.28.z cells in orthotopic breast carcinoma xenografts, and reduction of pulmonary metastasis in a renal cell carcinoma model, respectively. γ-irradiation as a potential safety measure for clinical application prevented NK cell replication, while antitumor activity was preserved. Our data demonstrate that it is feasible to engineer CAR-expressing NK cells as a clonal, molecularly and functionally well-defined and continuously expandable cell therapeutic agent, and suggest NK-92/5.28.z cells as a promising candidate for use in adoptive cancer immunotherapy.


Subject(s)
Gene Expression , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Receptor, ErbB-2/immunology , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Recombinant Fusion Proteins/genetics , Animals , Breast Neoplasms/genetics , Breast Neoplasms/immunology , Breast Neoplasms/therapy , Cell Line, Transformed , Cell Line, Tumor , Clonal Evolution , Cytotoxicity, Immunologic , Disease Models, Animal , Epitopes, T-Lymphocyte/immunology , Female , Genetic Vectors/genetics , Humans , Immunophenotyping , Immunotherapy , Lentivirus/genetics , Lymphocyte Culture Test, Mixed , Neoplasms/genetics , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Phenotype , Transduction, Genetic , Xenograft Model Antitumor Assays
16.
Oncoimmunology ; 3: e28147, 2014.
Article in English | MEDLINE | ID: mdl-25340009

ABSTRACT

T lymphocytes engineered to express a chimeric antigen receptor (CAR) are being celebrated as a major breakthrough of anticancer immunotherapy. Natural killer cells have not received similar attention as CAR effectors, although the use of these relatively short-lived cytotoxic cells is associated with several advantages.

17.
Cytotherapy ; 15(12): 1563-70, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24094496

ABSTRACT

BACKGROUND AIMS: Natural killer (NK) cells, either naive or genetically engineered, are increasingly considered for cellular therapy of patients with malignancies. When using NK cells from peripheral blood, the number of expanded NK cells can be highly variable and the need for NK cell enrichment can make the process expensive. The NK-92 cell line (CD56+/CD3-) that was isolated from a patient with lymphoma has predictable high cytotoxic activity and can be expanded under good manufacturing practice conditions in recombinant interleukin-2. METHODS: Fifteen patients (age, 9-71 years) with advanced, treatment-resistant malignancies, either solid tumors/sarcomas (n = 13) or leukemia/lymphoma (n = 2), received two infusions of NK-92 cells, given 48 h apart. Three cohorts of patients were treated with escalating doses of NK-92 cells (n = 7 at 1 × 10(9), n = 6 at 3 × 10(9) and n = 2 at 1 × 10(10) cells/m(2)). RESULTS: No infusion-related or long-term side effects were observed. The dose of 10(10) cells/m(2) was considered the maximum expandable cell dose with the use of an established culture bag system. Three fourths of patients with lung cancer had some anti-tumor response. Only one patient of seven had development of human leukocyte antigen antibodies. The persistence of NK-92 cells (male origin) in the circulation was confirmed by Y chromosome-specific polymerase chain reaction in two female patients. CONCLUSIONS: Infusions of NK-92 cells up to 10(10) cells/m(2) were well tolerated. Despite the allogeneic nature of NK-92, development of human leukocyte antigen antibodies in these patients with cancer appears to be rare. The cells can persist in the recipient's circulation for at least 48 h. Some encouraging responses were seen in patients with advanced lung cancer.


Subject(s)
Cell- and Tissue-Based Therapy , Immunotherapy , Killer Cells, Natural/transplantation , Leukemia, Lymphoid/therapy , Sarcoma/therapy , Adolescent , Adult , Aged , Child , Drug Resistance, Neoplasm/genetics , Female , Humans , Killer Cells, Natural/cytology , Leukemia, Lymphoid/pathology , Male , Middle Aged , Neoplasm Staging , Sarcoma/pathology , Transplantation, Homologous/adverse effects , Transplantation, Homologous/methods
18.
Cytotherapy ; 15(10): 1185-94, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23768925

ABSTRACT

Although T-lymphocytes have received most of the attention in immunotherapy trials, new discoveries around natural killer (NK) cells suggest that they also should be suitable effector cells for cellular therapy of cancer. In addition to direct cytotoxicity, NK cells produce an array of immune-active cytokines, among them interferons and granulocyte-macrophage colony-stimulating factor, which places them at the crossroads of innate and adaptive immunity. They also augment monoclonal antibody activity through antibody-mediated cellular cytotoxicity and can be transfected with chimeric antigen receptors. One of the stumbling blocks for NK cell-based therapies has been the inability to predictably obtain and expand larger numbers from donors, but also to achieve sufficiently high transfection efficiency of target genes. The first clinical trials with NK cells suggest some benefit, but more definite evidence is needed to justify this relatively expensive treatment.


Subject(s)
Immunotherapy, Adoptive/methods , Killer Cells, Natural/transplantation , Neoplasms/immunology , Neoplasms/therapy , Animals , Cell Proliferation , Clinical Trials as Topic , Genetic Therapy , Humans , Immunotherapy, Adoptive/trends , Killer Cells, Natural/immunology , Molecular Targeted Therapy
19.
Transfusion ; 53(2): 412-8; quiz 411, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22738379

ABSTRACT

BACKGROUND: In the setting of allogeneic stem cell transplantation (SCT), infusing natural killer (NK) cells from a major histocompatibility complex (MHC)-mismatched donor can mediate an antileukemic effect. The graft-versus-tumor effect after autologous stem cell transplantation (ASCT) may result in less disease relapse. STUDY DESIGN AND METHODS: We performed a Phase I clinical trial to assess the safety and feasibility of infusing distantly processed donor NK-enriched mononuclear cell (NK-MNC) infusions from a MHC haplotype-mismatched (haploidentical) donor to patients who recently underwent ASCT for a hematologic malignancy. On Day 1, peripheral blood MNCs were obtained by steady-state leukapheresis and sent from Boston to the Production Assistance for Cellular Therapies (PACT) facility at the University of Minnesota, where immunomagnetic depletion of CD3 cells was performed on Day 2. NK-MNC products were then returned to Boston on Day 2 for infusion on Day 3. Toxicity, cellular product characteristics, and logistic events were monitored. RESULTS: At a median of 90 days (range, 49-191 days) after ASCT, 13 patients were treated with escalating doses of NK-MNCs per kilogram from 10(5) to 2 × 10(7) . Adverse effects included Grade 2 rigors and muscle aches, but no Grade 3 or 4 events and no graft-versus-host disease or marrow suppression. One air courier delay occurred. NK-MNC products were viable with cytotoxic activity after transport. CONCLUSION: CD3-depleted, MHC-mismatched allogeneic NK-MNC infusions can be safely and feasibly administered to patients after ASCT after distant processing and transport, justifying further development of this approach.


Subject(s)
Blood Donors , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Killer Cells, Natural/transplantation , Leukapheresis/methods , Adult , Aged , Family , Feasibility Studies , Female , Haplotypes , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immune Tolerance/physiology , Infusions, Intravenous , Killer Cells, Natural/cytology , Male , Middle Aged , Transplantation, Autologous/adverse effects , Treatment Outcome , Young Adult
20.
Oncoimmunology ; 2(10): e26527, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-24404423

ABSTRACT

Multiple natural killer (NK) cell-based anticancer therapies are currently under development. Here, we compare the efficiency of genetically modified NK-92 cells expressing chimeric antigen receptors (CARs) at killing NK cell-resistant B-lymphoid leukemia cells to the antibody-dependent cell-mediated cytotoxicity (ADCC) of NK-92 cells expressing a high affinity variant of the IgG Fc receptor (FcγRIII). First, we compared in vitro the abilities of NK-92 cells expressing CD20-targeting CARs to kill primary chronic lymphocytic leukemia (CLL) cells derived from 9 patients with active, untreated disease to the cytotoxicity of NK-92 cells expressing FcγRIII combined with either of the anti-CD20 monoclonal antibodies (mAbs) rituximab or ofatumumab. We found that CAR-expressing NK-92 cells effectively kill NK cell-resistant primary CLL cells and that such a cytotoxic response is significantly stronger than that resulting from ADCC. For studying CAR-expressing NK cell-based immunotherapy in vivo, we established xenograft mouse models of residual leukemia using the human BCR-ABL1+ cell lines SUP-B15 (CD19+CD20-) and TMD-5 (CD19+CD20+), two acute lymphoblastic leukemia (ALL) lines that are resistant to parental NK-92 cells. Intravenous injection of NK-92 cells expressing CD19-targeting CARs eliminated SUP-B15 cells, whereas they had no such effect on TMD-5 cells. However, the intrafemoral injection of NK-92 cells expressing CD19-targeting CAR resulted in the depletion of TMD-5 cells from the bone marrow environment. Comparative studies in which NK-92 cells expressing either CD19- or CD20-targeting CARs were directly injected into subcutaneous CD19+CD20+ Daudi lymphoma xenografts revealed that CD20-targeting CAR is superior to its CD19-specific counterpart in controlling local tumor growth. In summary, we show here that CAR-expressing NK-92 cells can be functionally superior to ADCC (as mediated by anti-CD20 mAbs) in the elimination of primary CLL cells. Moreover, we provide data demonstrating that the systemic administration of CAR-expressing NK-92 cells can control lymphoblastic leukemia in immunocompromised mice. Our results also suggest that the direct injection of CAR-expressing NK-92 cells to neoplastic lesions could be an effective treatment modality against lymphoma.

SELECTION OF CITATIONS
SEARCH DETAIL