Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 856
Filtrar
1.
Front Immunol ; 15: 1390498, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38694508

RESUMO

Recent advancements in genetic engineering have made it possible to modify Natural Killer (NK) cells to enhance their ability to fight against various cancers, including solid tumors. This comprehensive overview discusses the current status of genetically engineered chimeric antigen receptor NK-cell therapies and their potential for treating solid tumors. We explore the inherent characteristics of NK cells and their role in immune regulation and tumor surveillance. Moreover, we examine the strategies used to genetically engineer NK cells in terms of efficacy, safety profile, and potential clinical applications. Our investigation suggests CAR-NK cells can effectively target and regress non-hematological malignancies, demonstrating enhanced antitumor efficacy. This implies excellent promise for treating tumors using genetically modified NK cells. Notably, NK cells exhibit low graft versus host disease (GvHD) potential and rarely induce significant toxicities, making them an ideal platform for CAR engineering. The adoptive transfer of allogeneic NK cells into patients further emphasizes the versatility of NK cells for various applications. We also address challenges and limitations associated with the clinical translation of genetically engineered NK-cell therapies, such as off-target effects, immune escape mechanisms, and manufacturing scalability. We provide strategies to overcome these obstacles through combination therapies and delivery optimization. Overall, we believe this review contributes to advancing NK-cell-based immunotherapy as a promising approach for cancer treatment by elucidating the underlying mechanisms, evaluating preclinical and clinical evidence, and addressing remaining challenges.


Assuntos
Engenharia Genética , Imunoterapia Adotiva , Células Matadoras Naturais , Neoplasias , Receptores de Antígenos Quiméricos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Humanos , Neoplasias/terapia , Neoplasias/imunologia , Imunoterapia Adotiva/métodos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/imunologia , Animais
2.
Front Immunol ; 15: 1385571, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38680498

RESUMO

Surgery, chemotherapy, and endocrine therapy have improved the overall survival and postoperative recurrence rates of Luminal A, Luminal B, and HER2-positive breast cancers but treatment modalities for triple-negative breast cancer (TNBC) with poor prognosis remain limited. The effective application of the rapidly developing chimeric antigen receptor (CAR)-T cell therapy in hematological tumors provides new ideas for the treatment of breast cancer. Choosing suitable and specific targets is crucial for applying CAR-T therapy for breast cancer treatment. In this paper, we summarize CAR-T therapy's effective targets and potential targets in different subtypes based on the existing research progress, especially for TNBC. CAR-based immunotherapy has resulted in advancements in the treatment of breast cancer. CAR-macrophages, CAR-NK cells, and CAR-mesenchymal stem cells (MSCs) may be more effective and safer for treating solid tumors, such as breast cancer. However, the tumor microenvironment (TME) of breast tumors and the side effects of CAR-T therapy pose challenges to CAR-based immunotherapy. CAR-T cells and CAR-NK cells-derived exosomes are advantageous in tumor therapy. Exosomes carrying CAR for breast cancer immunotherapy are of immense research value and may provide a treatment modality with good treatment effects. In this review, we provide an overview of the development and challenges of CAR-based immunotherapy in treating different subtypes of breast cancer and discuss the progress of CAR-expressing exosomes for breast cancer treatment. We elaborate on the development of CAR-T cells in TNBC therapy and the prospects of using CAR-macrophages, CAR-NK cells, and CAR-MSCs for treating breast cancer.


Assuntos
Neoplasias da Mama , Imunoterapia Adotiva , Receptores de Antígenos Quiméricos , Microambiente Tumoral , Humanos , Imunoterapia Adotiva/métodos , Imunoterapia Adotiva/efeitos adversos , Receptores de Antígenos Quiméricos/imunologia , Receptores de Antígenos Quiméricos/genética , Feminino , Microambiente Tumoral/imunologia , Neoplasias da Mama/terapia , Neoplasias da Mama/imunologia , Animais , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Neoplasias de Mama Triplo Negativas/terapia , Neoplasias de Mama Triplo Negativas/imunologia , Linfócitos T/imunologia , Exossomos/imunologia
3.
Med Oncol ; 41(6): 127, 2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38656354

RESUMO

Chimeric Antigen Receptor (CAR) based therapies are becoming increasingly important in treating patients. CAR-T cells have been shown to be highly effective in the treatment of hematological malignancies. However, harmful therapeutic barriers have been identified, such as the potential for graft-versus-host disease (GVHD), neurotoxicity, and cytokine release syndrome (CRS). As a result, CAR NK-cell therapy is expected to be a new therapeutic option. NK cells act as cytotoxic lymphocytes, supporting the innate immune response against autoimmune diseases and cancer cells by precisely detecting and eliminating malignant cells. Genetic modification of these cells provides a dual approach to the treatment of AD and cancer. It can be used through both CAR-independent and CAR-dependent mechanisms. The use of CAR-based cell therapies has been successful in treating cancer patients, leading to further investigation of this innovative treatment for alternative diseases, including AD. The complementary roles of CAR T and CAR NK cells have stimulated exploration in this area. Our study examines the latest research on the therapeutic effectiveness of these cells in treating both cancer and ADs.


Assuntos
Doenças Autoimunes , Imunoterapia Adotiva , Células Matadoras Naturais , Neoplasias , Receptores de Antígenos Quiméricos , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Receptores de Antígenos Quiméricos/imunologia , Neoplasias/terapia , Neoplasias/imunologia , Doenças Autoimunes/terapia , Doenças Autoimunes/imunologia , Imunoterapia Adotiva/métodos , Animais
4.
Nature ; 626(8000): 727-736, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38383621

RESUMO

Natural killer (NK) cells are lymphocytes of the innate immune system. A key feature of NK cells is their ability to recognize a wide range of cells in distress, particularly tumour cells and cells infected with viruses. They combine both direct effector functions against their cellular targets and participate in the generation, shaping and maintenance of a multicellular immune response. As our understanding has deepened, several therapeutic strategies focused on NK cells have been conceived and are currently in various stages of development, from preclinical investigations to clinical trials. Here we explore in detail the complexity of NK cell biology in humans and highlight the role of these cells in cancer immunity. We also analyse the harnessing of NK cell immunity through immune checkpoint inhibitors, NK cell engagers, and infusions of preactivated or genetically modified, autologous or allogeneic NK cell products.


Assuntos
Imunoterapia Adotiva , Células Matadoras Naturais , Neoplasias , Humanos , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Neoplasias/imunologia , Neoplasias/terapia , Imunidade Inata
5.
Med ; 4(7): 457-477.e8, 2023 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-37172578

RESUMO

BACKGROUND: The advent of chimeric antigen receptor (CAR) T cell therapies has transformed the treatment of hematological malignancies; however, broader therapeutic success of CAR T cells has been limited in solid tumors because of their frequently heterogeneous composition. Stress proteins in the MICA and MICB (MICA/B) family are broadly expressed by tumor cells following DNA damage but are rapidly shed to evade immune detection. METHODS: We have developed a novel CAR targeting the conserved α3 domain of MICA/B (3MICA/B CAR) and incorporated it into a multiplexed-engineered induced pluripotent stem cell (iPSC)-derived natural killer (NK) cell (3MICA/B CAR iNK) that expressed a shedding-resistant form of the CD16 Fc receptor to enable tumor recognition through two major targeting receptors. FINDINGS: We demonstrated that 3MICA/B CAR mitigates MICA/B shedding and inhibition via soluble MICA/B while simultaneously exhibiting antigen-specific anti-tumor reactivity across an expansive library of human cancer cell lines. Pre-clinical assessment of 3MICA/B CAR iNK cells demonstrated potent antigen-specific in vivo cytolytic activity against both solid and hematological xenograft models, which was further enhanced in combination with tumor-targeted therapeutic antibodies that activate the CD16 Fc receptor. CONCLUSIONS: Our work demonstrated 3MICA/B CAR iNK cells to be a promising multi-antigen-targeting cancer immunotherapy approach intended for solid tumors. FUNDING: Funded by Fate Therapeutics and NIH (R01CA238039).


Assuntos
Receptores de Antígenos Quiméricos , Humanos , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/metabolismo , Linhagem Celular Tumoral , Imunoterapia Adotiva , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/transplante , Receptores Fc/metabolismo
6.
Blood Rev ; 60: 101073, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-36959057

RESUMO

Natural Killer (NK) cells yield promise in therapy of hematologic malignancies. The clinical experience with adoptively transferred allogeneic NK cells over past two decades has revealed safety and minimal risk of CRS or ICANS. Unlike T cells which have to be genetically altered to avoid graft vs host disease (GVHD), HLA mismatched NK cells can be infused without GVHD risk. This makes them ideal for the development of off-the-shelf products. In this review we focus on NK biology relevant to the cancer therapy, the trajectory of NK therapeutics for leukemia, lymphoma, and myeloma; and advantages of the NK cell platform. We will also discuss novel methods to enhance NK cell targeting, persistence, and function in the tumor microenvironment. The future of NK cell therapy depends on novel strategies to realize these qualities.


Assuntos
Doença Enxerto-Hospedeiro , Neoplasias Hematológicas , Humanos , Neoplasias Hematológicas/terapia , Células Matadoras Naturais/transplante , Imunoterapia Adotiva/efeitos adversos , Terapia Baseada em Transplante de Células e Tecidos , Microambiente Tumoral
7.
Biochem Biophys Res Commun ; 640: 50-55, 2023 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-36502631

RESUMO

Triple-negative breast cancer (TNBC) is a subtype of breast tumor with the highest breast cancer stem cells (BCSCs) content and resistance to conventional treatment. Due to the immunosuppressive tumor microenvironment and immunogenicity of breast cancer cells, the use of immune cells, especially natural killer cells (NK) in the treatment of solid tumors, including breast cancer, has been unsatisfactory. Therefore, identifying novel therapies is requisite for breast cancer treatment. Furthermore, the combination of cancer therapies is an effective strategy to improve therapeutic effectiveness. In this study, we inhibited telomerase (hTERT) with BIBR1532, in stimulating NK cell cytotoxicity against breast cancer cells. The MDA-MB-231 cell line was cured with IC50 level of BIBR1532 for 24 h. Afterward, cells were washed with PBS and were co-cultured with peripheral blood NK cell for 5h. Finally, we assessed the impact of telomerase inhibition on the cytotoxicity of NK cells and apoptosis of breast cancer. Also, the expression of hTERT and apoptotic-related genes were evaluated. The data revealed that inhibition of telomerase increases NK cell cytotoxicity against breast cancer. Furthermore, telomerase inhibition and NK cell synergistically enhanced cell death in breast cancer cells by suppressing hTERT, upregulation of bax, and bad expression. In conclusion, telomerase suppression makes breast cancer cells more sensitive to NK cell therapy. Consequently, the combination of telomerase inhibition and NK cells can be useful in the treatment of breast cancer cells.


Assuntos
Apoptose , Células Matadoras Naturais , Telomerase , Neoplasias de Mama Triplo Negativas , Feminino , Humanos , Linhagem Celular Tumoral , Proliferação de Células , Telomerase/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/patologia , Neoplasias de Mama Triplo Negativas/terapia , Microambiente Tumoral , Células Matadoras Naturais/transplante , Terapia Baseada em Transplante de Células e Tecidos
8.
Stem Cell Rev Rep ; 19(1): 26-45, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-35994137

RESUMO

Natural killer (NK) cells are one of the innate immune cells that play an important role in preventing and controlling tumors and viral diseases, but their role in hematopoietic stem cell transplantation (HCT) is not yet fully understood. However, according to some research, these cells can prevent infections and tumor relapse without causing graft versus host disease (GVHD). In addition to NK cells, several studies are about the anti-leukemia effects of NK cell-derived exosomes that can highlight their roles in graft-versus-leukemia (GVL). In this paper, we intend to investigate the results of various articles on the role of NK cells in allogeneic hematopoietic cell transplantation and also their exosomes in GVL. Also, we have discussed the antiviral effects of these cells in post-HCT cytomegalovirus infection.


Assuntos
Exossomos , Doença Enxerto-Hospedeiro , Transplante de Células-Tronco Hematopoéticas , Leucemia , Humanos , Células Matadoras Naturais/transplante , Leucemia/terapia , Transplante de Células-Tronco Hematopoéticas/métodos
9.
Oncoimmunology ; 11(1): 2127508, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36249274

RESUMO

Glioblastoma (GB) is the most common primary brain tumor, which is characterized by low immunogenicity of tumor cells and prevalent immunosuppression in the tumor microenvironment (TME). Targeted local combination immunotherapy is a promising strategy to overcome these obstacles. Here, we evaluated tumor-cell specific delivery of an anti-PD-1 immunoadhesin (aPD-1) via a targeted adeno-associated viral vector (AAV) as well as HER2-specific NK-92/5.28.z (anti-HER2.CAR/NK-92) cells as components for a combination immunotherapy. In co-culture experiments, target-activated anti-HER2.CAR/NK-92 cells modified surrounding tumor cells and bystander immune cells by triggering the release of inflammatory cytokines and upregulation of PD-L1. Tumor cell-specific delivery of aPD-1 was achieved by displaying a HER2-specific designed ankyrin repeat protein (DARPin) on the AAV surface. HER2-AAV mediated gene transfer into GB cells correlated with HER2 expression levels, without inducing anti-viral responses in transduced cells. Furthermore, AAV-transduction did not interfere with anti-HER2.CAR/NK-92 cell-mediated tumor cell lysis. After selective transduction of HER2+ cells, aPD-1 expression was detected at the mRNA and protein level. The aPD-1 immunoadhesin was secreted in a time-dependent manner, bound its target on PD-1-expressing cells and was able to re-activate T cells by efficiently disrupting the PD-1/PD-L1 axis. Moreover, high intratumoral and low systemic aPD-1 concentrations were achieved following local injection of HER2-AAV into orthotopic tumor grafts in vivo. aPD-1 was selectively produced in tumor tissue and could be detected up to 10 days after a single HER2-AAV injection. In subcutaneous GL261-HER2 and Tu2449-HER2 immunocompetent mouse models, administration of the combination therapy significantly prolonged survival, including complete tumor control in several animals in the GL261-HER2 model. In summary, local therapy with aPD-1 encoding HER2-AAVs in combination with anti-HER2.CAR/NK-92 cells may be a promising novel strategy for GB immunotherapy with the potential to enhance efficacy and reduce systemic side effects of immune-checkpoint inhibitors.


Assuntos
Glioblastoma , Adenoviridae/genética , Animais , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Citocinas , Glioblastoma/genética , Glioblastoma/terapia , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/transplante , Camundongos , RNA Mensageiro , Receptor ErbB-2/metabolismo , Terapias em Estudo , Microambiente Tumoral
10.
Proc Natl Acad Sci U S A ; 119(25): e2122379119, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35696582

RESUMO

Acute myeloid leukemia (AML) remains a therapeutic challenge, and a paucity of tumor-specific targets has significantly hampered the development of effective immune-based therapies. Recent paradigm-changing studies have shown that natural killer (NK) cells exhibit innate memory upon brief activation with IL-12 and IL-18, leading to cytokine-induced memory-like (CIML) NK cell differentiation. CIML NK cells have enhanced antitumor activity and have shown promising results in early phase clinical trials in patients with relapsed/refractory AML. Here, we show that arming CIML NK cells with a neoepitope-specific chimeric antigen receptor (CAR) significantly enhances their antitumor responses to nucleophosphmin-1 (NPM1)-mutated AML while avoiding off-target toxicity. CIML NK cells differentiated from peripheral blood NK cells were efficiently transduced to express a TCR-like CAR that specifically recognizes a neoepitope derived from the cytosolic oncogenic NPM1-mutated protein presented by HLA-A2. These CAR CIML NK cells displayed enhanced activity against NPM1-mutated AML cell lines and patient-derived leukemic blast cells. CAR CIML NK cells persisted in vivo and significantly improved AML outcomes in xenograft models. Single-cell RNA sequencing and mass cytometry analyses identified up-regulation of cell proliferation, protein folding, immune responses, and major metabolic pathways in CAR-transduced CIML NK cells, resulting in tumor-specific, CAR-dependent activation and function in response to AML target cells. Thus, efficient arming of CIML NK cells with an NPM1-mutation-specific TCR-like CAR substantially improves their innate antitumor responses against an otherwise intracellular mutant protein. These preclinical findings justify evaluating this approach in clinical trials in HLA-A2+ AML patients with NPM1c mutations.


Assuntos
Memória Imunológica , Células de Memória Imunológica , Imunoterapia Adotiva , Células Matadoras Naturais , Leucemia Mieloide Aguda , Nucleofosmina , Receptores de Antígenos Quiméricos , Antígeno HLA-A2/imunologia , Humanos , Células de Memória Imunológica/imunologia , Células de Memória Imunológica/transplante , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Mutação , Nucleofosmina/genética , Nucleofosmina/imunologia , Receptores de Antígenos Quiméricos/genética , Receptores de Antígenos Quiméricos/imunologia
11.
Thorac Cancer ; 13(14): 2050-2056, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35670036

RESUMO

BACKGROUND: A previous trial showed that autologous ex-vivo expanded NK cell (SNK01) treatment combined with pembrolizumab showed better efficacy than pembrolizumab monotherapy in advanced non-small cell lung cancer (NSCLC). This study was a 2-year follow-up of that previous study to determine the long-term efficacy of the combination treatment. METHODS: This trial included 20 patients with advanced NSCLC with a PD-L1 tumor proportion score of 1% or greater who failed prior to front-line platinum-based therapy. The patients received pembrolizumab with low-dose SNK01 (2 × 109 cells/dose) or high-dose SNK01 (4 × 109 cells/dose), or pembrolizumab monotherapy. The primary study endpoint was overall survival (OS), and the secondary endpoint was progression-free survival (PFS). RESULTS: Two patients were excluded following serious adverse events. Among the 11 patients who died, five were from the NK groups (41.6%, n = 5/12), and six received pembrolizumab monotherapy (100%, n = 6/6). The estimated 2-year survival rate was 58.3% versus 16.7% (pembrolizumab plus SNK01 vs. pembrolizumab monotherapy). The hazard ratio of pembrolizumab plus SNK01 compared with pembrolizumab monotherapy was 0.32 (95% CI: 0.1, 1.08, p-value: 0.066). Although the median PFS was significantly higher in the pembrolizumab plus SNK01 group than in the pembrolizumab alone group, OS and PFS did not differ statistically between patients who received low doses of NK cells and those who received high doses of NK cells. CONCLUSIONS: Autologous NK cells can enhance the long-term OS and PFS for NSCLC. A larger study is needed to confirm this result. Clinical Research Information Service number: KCT0003463.


Assuntos
Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Anticorpos Monoclonais Humanizados/farmacologia , Anticorpos Monoclonais Humanizados/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Antígeno B7-H1/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/patologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Humanos , Células Matadoras Naturais/transplante , Neoplasias Pulmonares/patologia , Intervalo Livre de Progressão
12.
Front Immunol ; 13: 879452, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35720374

RESUMO

Background: To explore the feasibility and safety of natural killer (NK) cell therapy in HCC, we performed a prospective, open-label, phase I trial to evaluate the synergistic effect of locoregional high-dose autologous NK cell therapy in combination with hepatic arterial infusion chemotherapy (HAIC). Methods: Patients with locally advanced HCC who were refractory to the standard treatment were eligible for this study. Patients received expanded and activated NK cells for 5 consecutive days in a dose-escalating manner (dose 2.5×108, 5×108, 10×108 NK cells/injection) through hepatic arterial infusion following 4 cycles of HAIC with 5-fluorouracil (750 mg/m2) and cisplatin (25 mg/m2). The primary endpoint was the safety of NK cell-based immunotherapy, and the secondary endpoints were objective response rate (ORR), progression-free survival (PFS), overall survival (OS), and immunologic responses. Results: Of the 11 patients enrolled, the confirmed ORR was 63.6% (complete response [CR]: 36.4%, confirmed partial response [PR]: 27.3%). Stable disease (SD) and progressive disease (PD) were observed in two patients (18.2%) each, resulting in a disease control rate (DCR) of 81.8%. The median PFS and OS were 10.3 and 41.6 months, respectively. There were no incidences of decompensation or severe adverse events during HAIC, and no adverse events related to NK cell infusion were noted. Conclusion: The combination of HAIC and locoregional high-dose NK cell therapy is a safe and effective treatment for locally advanced HCC patients who were refractory to the standard treatment. This result warrants further development of this novel treatment to establish its efficacy in HCC. Clinical Trial Registration: cris.nih.go.kr, identifier KCT0003973.


Assuntos
Carcinoma Hepatocelular , Terapia Baseada em Transplante de Células e Tecidos , Neoplasias Hepáticas , Protocolos de Quimioterapia Combinada Antineoplásica , Carcinoma Hepatocelular/patologia , Carcinoma Hepatocelular/terapia , Terapia Baseada em Transplante de Células e Tecidos/efeitos adversos , Humanos , Células Matadoras Naturais/transplante , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/terapia , Estudos Prospectivos
13.
Cytotherapy ; 24(8): 802-817, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35589475

RESUMO

T cell-based therapies like genetically modified immune cells expressing chimeric antigen receptors have shown robust anti-cancer activity in vivo, especially in patients with blood cancers. However, extending this approach to an "off-the-shelf" setting can be challenging, as allogeneic T cells carry a significant risk of graft-versus-host disease (GVHD). By contrast, allogeneic natural killer (NK) cells recognize malignant cells without the need for prior antigen exposure and have been used safely in multiple cancer settings without the risk of GVHD. However, similar to T cells, NK cell function is negatively impacted by tumor-induced transforming growth factor beta (TGF-ß) secretion, which is a ubiquitous and potent immunosuppressive mechanism employed by most malignancies. Allogeneic NK cells for adoptive immunotherapy can be sourced from peripheral blood (PB) or cord blood (CB), and the authors' group and others have previously shown that ex vivo expansion and gene engineering can overcome CB-derived NK cells' functional immaturity and poor cytolytic activity, including in the presence of exogenous TGF-ß.  However, a direct comparison of the effects of TGF-ß-mediated immune suppression on ex vivo-expanded CB- versus PB-derived NK cell therapy products has not previously been performed. Here the authors show that PB- and CB-derived NK cells have distinctive gene signatures that can be overcome by ex vivo expansion. Additionally, exposure to exogenous TGF-ß results in an upregulation of inhibitory receptors on NK cells, a novel immunosuppressive mechanism not previously described. Finally, the authors provide functional and genetic evidence that both PB- and CB-derived NK cells are equivalently susceptible to TGF-ß-mediated immune suppression. The authors believe these results provide important mechanistic insights to consider when using ex vivo-expanded, TGF-ß-resistant PB- or CB-derived NK cells as novel immunotherapy agents for cancer.


Assuntos
Doença Enxerto-Hospedeiro , Imunoterapia Adotiva , Fator de Crescimento Transformador beta , Linhagem Celular Tumoral , Sangue Fetal , Doença Enxerto-Hospedeiro/terapia , Humanos , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/transplante , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/uso terapêutico
14.
Pediatr Blood Cancer ; 69(8): e29676, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35441789

RESUMO

BACKGROUND: Natural killer (NK) cell therapy has been shown to be effective in the treatment of some cancers. However, the effects of this adoptive immunotherapy have not been investigated for Wilms tumor (WT). In this study, the effects of adoptive NK-cell transfer on a patient-derived xenograft (PDX) model of anaplastic WT were evaluated, and the impacts of cell source and ex vivo activation strategy on the therapeutic efficacy of NK-cell product were appraised. METHODS: NK cells were isolated from human peripheral blood mononuclear cells (NKPB ) and human cord blood (NKCB ), and were expanded and activated using a cytokine cocktail. Another group of NK cells (NKET ) was produced through activation with the exosomes extracted from previously challenged NKPB cells with WT. PDX-bearing mice were treated with clinically relevant doses of NKPB , NKCB , NKET , standard chemotherapy, and placebo (phosphate-buffered saline). RESULTS: PDX models treated with NKCB showed a better survival rate, though the difference among the study groups was not significant. Compared with the placebo control group, NKCB significantly improved the histopathologic response, NKPB significantly inhibited the proliferation of neoplastic cells, and NKET led to a significant decrease in the metastasis score (all p-values <.05). Standard chemotherapy provided the greatest tumor growth inhibition and the lowest mitotic count, though it did not show any significant advantage over NK-cell therapies in any of the outcome parameters in two-by-two comparisons. CONCLUSIONS: This study spotlights the efficacy of adoptive NK-cell transfer as a potential treatment candidate for high-risk WT.


Assuntos
Neoplasias Renais , Tumor de Wilms , Animais , Linhagem Celular Tumoral , Citotoxicidade Imunológica , Humanos , Imunoterapia Adotiva , Neoplasias Renais/terapia , Células Matadoras Naturais/transplante , Leucócitos Mononucleares , Camundongos , Tumor de Wilms/terapia
15.
Comput Math Methods Med ; 2022: 8361665, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35047059

RESUMO

OBJECTIVE: To explore the effect of autoimmune cell therapy on immune cells in patients with chronic obstructive pulmonary disease (COPD) and to provide a reference for clinical treatment of COPD. METHODS: Sixty patients with stable COPD were randomly divided into control group and treatment group (n = 30). The control group was given conventional treatment, and the treatment group was given one autoimmune cell therapy on the basis of conventional treatment. The serum levels of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the peripheral blood were detected by flow cytometry. Possible adverse reactions were detected at any time during treatment. RESULTS: There were no significant differences in the contents of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the serum of the control group (P > 0.05). Compared with before treatment, the contents of CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the serum of the treatment group were significantly increased (P < 0.05). The ratio of CD4 + /CD8+ T cells in both control and treatment groups did not change significantly during treatment (P > 0.05). There were no significant differences in serum CD3+ T cells, CD4+ T cells, CD8+ cells, B cells, and NK cells in the treatment group at 30 days and 90 days after treatment (P > 0.05), but they were significantly higher than those in the control group (P < 0.05). CONCLUSION: Autoimmune cell therapy can significantly increase the level of immune cells in the body and can be maintained for a long period of time, which has certain clinical benefits for recurrent respiratory tract infections and acute exacerbation in patients with COPD.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos/métodos , Doença Pulmonar Obstrutiva Crônica/imunologia , Doença Pulmonar Obstrutiva Crônica/terapia , Idoso , Idoso de 80 Anos ou mais , Autoimunidade , Linfócitos B/imunologia , Linfócitos B/transplante , Transfusão de Sangue Autóloga/métodos , Transfusão de Sangue Autóloga/estatística & dados numéricos , Terapia Baseada em Transplante de Células e Tecidos/estatística & dados numéricos , Biologia Computacional , Feminino , Humanos , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Transfusão de Leucócitos/métodos , Transfusão de Leucócitos/estatística & dados numéricos , Masculino , Pessoa de Meia-Idade , Doença Pulmonar Obstrutiva Crônica/sangue , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/transplante
16.
Anticancer Res ; 42(2): 1181-1187, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35093924

RESUMO

BACKGROUND/AIM: Adoptive immunotherapy (AIT) using autologous zoledronate-activated killer (ZAK) cells has been performed for developing a novel modality of cancer treatment. In this study, data series from incurable pancreatic cancer were analyzed. PATIENTS AND METHODS: Patients were treated with AIT using intravenous administration of ZAK cells every 3 to 4 weeks in combination with standard chemotherapy and possible clinical benefits were examined. RESULTS: Seventy-five patients were treated. A median overall survival (OS) time of 6.7 months was achieved for all patients and 13.1 months for those treated 5 times or more, that increased to 14.6 and 18.3 months, respectively, when the previous treatment period of chemotherapy alone was included in the analysis. The disease control rate was 58.5 %. Multivariate regression analysis showed a significant positive correlation between the survival and baseline value of lymphocyte percentage in white blood cell counts (p=0.031). CONCLUSION: The data suggest that AIT using ZAK cells in combination with chemotherapy is safe and feasible and may be effective in prolonging survival for patients with incurable pancreatic cancer. The lymphocyte percentage at baseline may be a good biomarker for predicting the survival benefit of ZAK cell AIT.


Assuntos
Imunoterapia Adotiva/métodos , Células Matadoras Naturais/transplante , Neoplasias Pancreáticas/terapia , Adulto , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Terapia Combinada , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Japão/epidemiologia , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Ativação Linfocitária/efeitos dos fármacos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/patologia , Estudos Prospectivos , Terapia de Salvação , Análise de Sobrevida , Resultado do Tratamento , Ácido Zoledrônico/farmacologia , Ácido Zoledrônico/uso terapêutico , Neoplasias Pancreáticas
17.
Nat Commun ; 13(1): 121, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013215

RESUMO

HIV is difficult to eradicate due to the persistence of a long-lived reservoir of latently infected cells. Previous studies have shown that natural killer cells are important to inhibiting HIV infection, but it is unclear whether the administration of natural killer cells can reduce rebound viremia when anti-retroviral therapy is discontinued. Here we show the administration of allogeneic human peripheral blood natural killer cells delays viral rebound following interruption of anti-retroviral therapy in humanized mice infected with HIV-1. Utilizing genetically barcoded virus technology, we show these natural killer cells efficiently reduced viral clones rebounding from latency. Moreover, a kick and kill strategy comprised of the protein kinase C modulator and latency reversing agent SUW133 and allogeneic human peripheral blood natural killer cells during anti-retroviral therapy eliminated the viral reservoir in a subset of mice. Therefore, combinations utilizing latency reversal agents with targeted cellular killing agents may be an effective approach to eradicating the viral reservoir.


Assuntos
Fármacos Anti-HIV/farmacologia , Linfócitos T CD4-Positivos/imunologia , Infecções por HIV/terapia , HIV-1/efeitos dos fármacos , Células Matadoras Naturais/imunologia , Inibidores de Proteínas Quinases/farmacologia , Viremia/terapia , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/imunologia , Medula Óssea/virologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/virologia , Técnicas de Cocultura , Feminino , Infecções por HIV/genética , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/genética , HIV-1/imunologia , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Células Matadoras Naturais/transplante , Masculino , Camundongos , Camundongos Transgênicos , Proteína Quinase C/genética , Proteína Quinase C/imunologia , Baço/efeitos dos fármacos , Baço/imunologia , Baço/virologia , Carga Viral/efeitos dos fármacos , Viremia/genética , Viremia/imunologia , Viremia/virologia , Latência Viral/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
18.
Hum Immunol ; 83(1): 86-98, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34583856

RESUMO

The global outbreak of coronavirus-2019 (COVID-19) still claims more lives daily around the world due to the lack of a definitive treatment and the rapid tendency of virus to mutate, which even jeopardizes vaccination efficacy. At the forefront battle against SARS-CoV-2, an effective innate response to the infection has a pivotal role in the initial control and treatment of disease. However, SARS-CoV-2 subtly interrupts the equations of immune responses, disrupting the cytolytic antiviral effects of NK cells, while seriously activating infected macrophages and other immune cells to induce an unleashed "cytokine storm", a dangerous and uncontrollable inflammatory response causing life-threatening symptoms in patients. Notably, the NK cell exhaustion with ineffective cytolytic function against the sources of exaggerated cytokine release, acts as an Achilles' heel which exacerbates the severity of COVID-19. Given this, approaches that improve NK cell cytotoxicity may benefit treatment protocols. As a suggestion, adoptive transfer of NK or CAR-NK cells with proper cytotolytic potentials and the lowest capacity of cytokine-release (for example CD56dim NK cells brightly express activating receptors), to severe COVID-19 patients may provide an effective cure especially in cases suffering from cytokine storms. More intriguingly, the ongoing evidence for persistent clonal expansion of NK memory cells characterized by an activating phenotype in response to viral infections, can benefit the future studies on vaccine development and adoptive NK cell therapy in COVID-19. Whether vaccinated volunteers or recovered patients can also be considered as suitable candidates for cell donation could be the subject of future research.


Assuntos
Transferência Adotiva , COVID-19/terapia , Síndrome da Liberação de Citocina/terapia , Citocinas/imunologia , Células Matadoras Naturais/transplante , SARS-CoV-2/imunologia , Transferência Adotiva/efeitos adversos , Animais , COVID-19/imunologia , COVID-19/metabolismo , COVID-19/virologia , Síndrome da Liberação de Citocina/imunologia , Síndrome da Liberação de Citocina/metabolismo , Síndrome da Liberação de Citocina/virologia , Citocinas/metabolismo , Citotoxicidade Imunológica , Interações Hospedeiro-Patógeno , Humanos , Memória Imunológica , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Células Matadoras Naturais/virologia , SARS-CoV-2/patogenicidade , Resultado do Tratamento
19.
Leukemia ; 36(1): 155-164, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34312462

RESUMO

In this phase I/II clinical trial, we investigated the safety and efficacy of high doses of mb-IL21 ex vivo expanded donor-derived NK cells to decrease relapse in 25 patients with myeloid malignancies receiving haploidentical stem-cell transplantation (HSCT). Three doses of donor NK cells (1 × 105-1 × 108 cells/kg/dose) were administered on days -2, +7, and +28. Results were compared with an independent contemporaneously treated case-matched cohort of 160 patients from the CIBMTR database.After a median follow-up of 24 months, the 2-year relapse rate was 4% vs. 38% (p = 0.014), and disease-free survival (DFS) was 66% vs. 44% (p = 0.1) in the cases and controls, respectively. Only one relapse occurred in the study group, in a patient with the high level of donor-specific anti-HLA antibodies (DSA) presented before transplantation. The 2-year relapse and DFS in patients without DSA was 0% vs. 40% and 72% vs. 44%, respectively with HR for DFS in controls of 2.64 (p = 0.029). NK cells in recipient blood were increased at day +30 in a dose-dependent manner compared with historical controls, and had a proliferating, mature, highly cytotoxic, NKG2C+/KIR+ phenotype.Administration of donor-derived expanded NK cells after haploidentical transplantation was safe, associated with NK cell-dominant immune reconstitution early post-transplant, preserved T-cell reconstitution, and improved relapse and DFS. TRIAL REGISTRATION: NCT01904136 ( https://clinicaltrials.gov/ct2/show/NCT01904136 ).


Assuntos
Neoplasias Hematológicas/terapia , Transplante de Células-Tronco Hematopoéticas/mortalidade , Células Matadoras Naturais/transplante , Recidiva Local de Neoplasia/terapia , Doadores não Relacionados/estatística & dados numéricos , Adolescente , Adulto , Idoso , Estudos de Casos e Controles , Feminino , Seguimentos , Neoplasias Hematológicas/imunologia , Neoplasias Hematológicas/patologia , Humanos , Células Matadoras Naturais/imunologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/imunologia , Recidiva Local de Neoplasia/patologia , Prognóstico , Taxa de Sobrevida , Condicionamento Pré-Transplante , Transplante Haploidêntico , Adulto Jovem
20.
Cancer Immunol Immunother ; 71(3): 589-599, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34282496

RESUMO

Liver transplantation (LT) is a viable treatment option for cirrhosis patients with hepatocellular carcinoma (HCC). However, recurrence is the rate-limiting factor of long-term survival. To prevent this, we conducted the phase I study of the adoptive transfer of deceased donor liver-derived natural killer (NK) cells. Liver NK cells were extracted from donor liver graft perfusate and were stimulated in vitro with IL-2. The patient received an intravenous infusion of NK cells 3-5 days after LT. Eighteen LT recipients were treated. There were no severe cell infusion-related adverse events or acute rejection episodes. One patient withdrew from the study because the pathological observation revealed sarcoma instead of HCC. All patients who received this immunotherapy completed the follow-up for at least 2 years without evidence of HCC recurrence (median follow-up, 96 months [range, 17-121 months]). Considering that 9 (52.9%) of the 17 patients pathologically exceeded the Milan criteria, liver NK cell infusion is likely to be useful for preventing HCC recurrence after LT. This is the first-in-human immunotherapy study using deceased donor liver-derived NK cells to prevent HCC recurrence after LT. This treatment was well tolerated and resulted in no HCC recurrence after LT.Clinical trial registration www.clinicaltrials.gov ; NCT01147380; registration date: June 17, 2010.


Assuntos
Carcinoma Hepatocelular/terapia , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/transplante , Neoplasias Hepáticas/terapia , Transplante de Fígado , Fígado/imunologia , Transferência Adotiva , Adulto , Idoso , Biomarcadores , Terapia Combinada , Estudos de Viabilidade , Feminino , Sobrevivência de Enxerto , Humanos , Imuno-Histoquímica , Células Matadoras Naturais/metabolismo , Transplante de Fígado/efeitos adversos , Transplante de Fígado/métodos , Masculino , Pessoa de Meia-Idade , Projetos Piloto , Resultado do Tratamento , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA