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










Publication year range
1.
Front Immunol ; 15: 1371353, 2024.
Article in English | MEDLINE | ID: mdl-38605958

ABSTRACT

Background: BVAC-C, a B cell- and monocyte-based immunotherapeutic vaccine transfected with recombinant HPV E6/E7, was well tolerated in HPV-positive recurrent cervical carcinoma patients in a phase I study. This phase IIa study investigates the antitumor activity of BVAC-C in patients with HPV 16- or 18-positive cervical cancer who had experienced recurrence after a platinum-based combination chemotherapy. Patients and methods: Patients were allocated to 3 arms; Arm 1, BVAC-C injection at 0, 4, 8 weeks; Arm 2, BVAC-C injection at 0, 4, 8, 12 weeks; Arm 3, BVAC-C injection at 0, 4, 8, 12 weeks with topotecan at 2, 6, 10, 14 weeks. Primary endpoints were safety and objective response rate (ORR) as assessed by an independent radiologist according to Response Evaluation Criteria in Solid Tumors version 1.1. Secondary endpoints included the disease control rate (DCR), duration of response (DOR), progression-free survival (PFS), and overall survival (OS). Results: Of the 30 patients available for analysis, the ORR was 19.2% (Arm 1: 20.0% (3/15), Arm 2: 33.3% (2/6), Arm3: 0%) and the DCR was 53.8% (Arm 1: 57.1%, Arm 2: 28.6%, Arm3: 14.3%). The median DOR was 7.5 months (95% CI 7.1-not reported), the median PFS was 5.8 months (95% CI 4.2-10.3), and the median OS was 17.7 months (95% CI 12.0-not reported). All evaluated patients showed not only inflammatory cytokine responses (IFN-γ or TNF-α) but also potent E6/E7-specific T cell responses upon vaccinations. Immune responses of patients after vaccination were correlated with their clinical responses. Conclusion: BVAC-C represents a promising treatment option and a manageable safety profile in the second-line setting for this patient population. Further studies are needed to identify potential biomarkers of response. Clinical trial registration: ClinicalTrials.gov, identifier NCT02866006.


Subject(s)
Cancer Vaccines , Papillomavirus Infections , Uterine Cervical Neoplasms , Female , Humans , Uterine Cervical Neoplasms/drug therapy , Human papillomavirus 16 , Neoplasm Recurrence, Local/pathology , Cancer Vaccines/adverse effects
2.
Cancer Lett ; 520: 38-47, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34224797

ABSTRACT

Understanding the rationale of combining immunotherapy and other anticancer treatment modalities is of great interest because of interpatient variability in single-agent immunotherapy. Here, we demonstrated that topoisomerase I inhibitors, a class of chemotherapeutic drugs, can alter the tumor immune landscape, corroborating their antitumor effects combined with immunotherapy. We observed that topotecan-conditioned TC-1 tumors were occupied by a vast number of monocytic cells that highly express CD11c, CD64, and costimulatory molecules responsible for the favorable changes in the tumor microenvironment. Ly6C+MHC-II+CD11chiCD64hi cells, referred to as topotecan-induced monocyte-derived dendritic cells (moDCs), proliferate and activate antigen-specific CD8+ T cells to levels equivalent to those of conventional DCs. Phenotypic changes in Ly6C+ cells towards moDCs were similarly induced by exposure to topotecan in vitro, which was more profoundly facilitated in the presence of tumor cells. Notably, anti-M-CSFR reversed the acquisition of DC-like properties of topotecan-induced moDCs, leading to the abolition of the antitumor effect of topotecan combined with a cancer vaccine. In short, topoisomerase I inhibitors generate monocyte-derived antigen-presenting cells in tumors, which could be mediated by M-CSF-M-CSFR signaling.


Subject(s)
Antigen-Presenting Cells/immunology , Immunotherapy , Neoplasms/therapy , Topoisomerase I Inhibitors/pharmacology , Animals , Antigens, Ly/immunology , CD11c Antigen/immunology , Cancer Vaccines/immunology , Cancer Vaccines/pharmacology , Cell Proliferation/genetics , Coculture Techniques , Combined Modality Therapy , Dendritic Cells/drug effects , Dendritic Cells/immunology , Humans , Immunity, Innate/drug effects , Immunity, Innate/immunology , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Neoplasms/immunology , Neoplasms/pathology , Receptors, IgG/immunology , T-Lymphocytes/immunology , Topoisomerase I Inhibitors/immunology , Topotecan/pharmacology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
3.
Vaccines (Basel) ; 8(3)2020 Jul 22.
Article in English | MEDLINE | ID: mdl-32707803

ABSTRACT

For cancer vaccines, the selection of optimal tumor-associated antigens (TAAs) that can maximize the immunogenicity of the vaccine without causing unwanted adverse effects is challenging. In this study, we developed two engineered Human epidermal growth factor receptor 2 (HER2) antigens, K965 and K1117, and compared their immunogenicity to a previously reported truncated HER2 antigen, K684, within a B cell and monocyte-based vaccine (BVAC). We found that BVAC-K965 and BVAC-K1117 induced comparable antigen-specific antibody responses and antigen-specific T cell responses to BVAC-K684. Interestingly, BVAC-K1117 induced more potent antitumor activity than the other vaccines in murine CT26-HER2 tumor models. In addition, BVAC-K1117 showed enhanced antitumor effects against truncated p95HER2-expressing CT26 tumors compared to BVAC-K965 and BVAC-K684 based on the survival analysis by inducing T cell responses against intracellular domain (ICD) epitopes. The increased ICD epitope-specific T cell responses induced by BVAC-K1117 compared to BVAC-K965 and BVAC-K684 were recapitulated in human leukocyte antigen (HLA)-untyped human PBMCs and HLA-A*0201 PBMCs. Furthermore, we also observed synergistic antitumor effects between BVAC-K1117 and anti-PD-L1 antibody treatment against CT26-HER2 tumors. Collectively, our findings demonstrate that inclusion of a sufficient number of ICD epitopes of HER2 in cellular vaccines can improve the antitumor activity of the vaccine and provide a way to optimize the efficacy of anticancer cellular vaccines targeting HER2.

4.
Cancer Immunol Res ; 8(5): 698-709, 2020 05.
Article in English | MEDLINE | ID: mdl-32122993

ABSTRACT

Although treatment with the glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) agonistic antibody (DTA-1) has shown antitumor activity in various tumor models, the underlying mechanism is not fully understood. Here, we demonstrate that interleukin (IL)-21-producing follicular helper T (Tfh) cells play a crucial role in DTA-1-induced tumor inhibition. The administration of DTA-1 increased IL21 expression by Tfh cells in an antigen-specific manner, and this activation led to enhanced antitumor cytotoxic T lymphocyte (CTL) activity. Mice treated with an antibody that neutralizes the IL21 receptor exhibited decreased antitumor activity when treated with DTA-1. Tumor growth inhibition by DTA-1 was abrogated in Bcl6 fl/fl Cd4 Cre mice, which are genetically deficient in Tfh cells. IL4 was required for optimal induction of IL21-expressing Tfh cells by GITR costimulation, and c-Maf mediated this pathway. Thus, our findings identify GITR costimulation as an inducer of IL21-expressing Tfh cells and provide a mechanism for the antitumor activity of GITR agonism.


Subject(s)
Antibodies, Monoclonal/pharmacology , Cytokines/metabolism , Glucocorticoid-Induced TNFR-Related Protein/agonists , Interleukins/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Glucocorticoid-Induced TNFR-Related Protein/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/drug therapy , Neoplasms/metabolism
5.
FASEB J ; 34(3): 4462-4481, 2020 03.
Article in English | MEDLINE | ID: mdl-31989715

ABSTRACT

Myeloid progenitor cells have generally been considered the predominant source of myeloid cells under steady-state conditions. Here we show that NK cells contributed to a myeloid cell lineage pool in naïve and tumor-bearing mice. Using fate tracing of NKp46+ cells, we found that myeloid cells could be derived from NK cells. Notably, among mature CD11b+ CD27+ NK cells, c-Kit+ CD24+ NK cells were capable of differentiating into a range of myeloid lineages in vitro and produced neutrophils and monocytes in vivo. The differentiation was completely inhibited by NK-stimulating cytokines. In addition to the potential for differentiation into myeloid cells, c-Kit+ CD24+ NK cells retained NK cell phenotypes and effector functions. Mechanistically, GATA-2 was necessary for the differentiation of c-Kit+ CD24+ NK cells. Therefore, we discovered that GATA-2-dependent differentiation of c-Kit+ CD24+ NK cells contributes to myeloid cell development and identified a novel pathway for myeloid lineage commitment under physiological conditions.


Subject(s)
Cell Proliferation/physiology , Myeloid Cells/cytology , Myeloid Cells/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , CD11b Antigen/genetics , CD11b Antigen/metabolism , CD24 Antigen/genetics , CD24 Antigen/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Lentivirus/genetics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Monocytes/metabolism , Natural Cytotoxicity Triggering Receptor 1/genetics , Natural Cytotoxicity Triggering Receptor 1/metabolism , Neutrophils/metabolism , Phagocytosis/genetics , Phagocytosis/physiology , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
6.
Front Immunol ; 10: 1887, 2019.
Article in English | MEDLINE | ID: mdl-31474983

ABSTRACT

Monocyte-derived dendritic cells (moDCs) have been shown to robustly expand during infection; however, their roles in anti-infectious immunity remain unclear. Here, we found that moDCs were dramatically increased in the secondary lymphoid organs during acute LCMV infection in an interferon-γ (IFN-γ)-dependent manner. We also found that priming by moDCs enhanced the differentiation of memory CD8+ T cells compared to differentiation primed by conventional dendritic cells (cDCs) through upregulation of Eomesodermin (Eomes) and T cell factor-1 (TCF-1) expression in CD8+ T cells. Consequently, impaired memory formation of CD8+ T cells in mice that had reduced numbers of moDCs led to defective clearance of pathogens upon rechallenge. Mechanistically, attenuated interleukin-2 (IL-2) signaling in CD8+ T cells primed by moDCs was responsible for the enhanced memory programming of CD8+ T cells. Therefore, our findings unveil a specialization of the antigen-presenting cell subsets in the fate determination of CD8+ T cells during infection and pave the way for the development of a novel therapeutic intervention on infection.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunologic Memory/immunology , Lymphocyte Activation/immunology , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Hepatocyte Nuclear Factor 1-alpha/metabolism , Interferon-gamma/immunology , Interleukin-2/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Listeriosis/pathology , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/pathology , Lymphocytic choriomeningitis virus/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Box Domain Proteins/metabolism
7.
Cancer Res ; 78(18): 5315-5326, 2018 09 15.
Article in English | MEDLINE | ID: mdl-30012672

ABSTRACT

PD-1-based cancer immunotherapy is a successful example of immune checkpoint blockade that provides long-term durable therapeutic effects in patients with cancer across a wide spectrum of cancer types. Accumulating evidence suggests that anti-PD-1 therapy enhances antitumor immunity by reversing the function of exhausted T cells in the tumor environment. However, the responsiveness rate of patients with cancer to anti-PD-1 therapy remains low, providing an urgent need for optimization and improvement. In this study, we designed an anti-PD-1-resistant mouse tumor model and showed that unresponsiveness to anti-PD-1 is associated with a gradual increase in CD8 T-cell exhaustion. We also found that invariant natural killer T cell stimulation by the synthetic ligand α-galactosylceramide (αGC) can enhance the antitumor effect in anti-PD-1-resistant tumors by restoring the effector function of tumor antigen-specific exhausted CD8 T cells. IL2 and IL12 were among the cytokines produced by αGC stimulation critical for reinvigorating exhausted CD8 T cells in tumor-bearing mice and patients with cancer. Furthermore, we observed a synergistic increase in the antitumor effect between αGC-loaded antigen-presenting cells and PD-1 blockade in a therapeutic murine tumor model. Our study suggests NKT cell stimulation as a promising therapeutic strategy for the treatment of patients with anti-PD-1-resistant cancer.Significance: These findings provide mechanistic insights into the application of NKT cell stimulation as a potent adjuvant for immunotherapy against advanced cancer. Cancer Res; 78(18); 5315-26. ©2018 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/cytology , Killer Cells, Natural/cytology , Neoplasms/immunology , Programmed Cell Death 1 Receptor/metabolism , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Cytotoxicity, Immunologic , Female , Galactosylceramides/pharmacology , Humans , Immunotherapy , Interleukin-12/metabolism , Interleukin-2/metabolism , Ligands , Lymphocyte Activation , Lymphocytes, Tumor-Infiltrating/cytology , Melanoma, Experimental , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Natural Killer T-Cells/immunology
8.
Nat Commun ; 8: 15776, 2017 06 06.
Article in English | MEDLINE | ID: mdl-28585539

ABSTRACT

During cancer immunoediting, loss of major histocompatibility complex class I (MHC-I) in neoplasm contributes to the evasion of tumours from host immune system. Recent studies have demonstrated that most natural killer (NK) cells that are found in advanced cancers are defective, releasing the malignant MHC-I-deficient tumours from NK-cell-dependent immune control. Here, we show that a natural killer T (NKT)-cell-ligand-loaded tumour-antigen expressing antigen-presenting cell (APC)-based vaccine effectively eradicates these advanced tumours. During this process, we find that the co-expression of Tim-3 and PD-1 marks functionally exhausted NK cells in advanced tumours and that MHC-I downregulation in tumours is closely associated with the induction of NK-cell exhaustion in both tumour-bearing mice and cancer patients. Furthermore, the recovery of NK-cell function by IL-21 is critical for the anti-tumour effects of the vaccine against advanced tumours. These results reveal the process involved in the induction of NK-cell dysfunction in advanced cancers and provide a guidance for the development of strategies for cancer immunotherapy.


Subject(s)
Cancer Vaccines/pharmacology , Genes, MHC Class I , Interleukins/immunology , Killer Cells, Natural/immunology , Animals , Biomarkers, Tumor/immunology , Cancer Vaccines/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Hepatitis A Virus Cellular Receptor 2/immunology , Hepatitis A Virus Cellular Receptor 2/metabolism , Humans , Interleukins/metabolism , Interleukins/pharmacology , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/immunology , Urinary Bladder Neoplasms/pathology , Xenograft Model Antitumor Assays
9.
Cancer Res ; 74(22): 6705-16, 2014 Nov 15.
Article in English | MEDLINE | ID: mdl-25273090

ABSTRACT

Extramedullary myelopoiesis occurs commonly in tumor-bearing animals and is known to lead to accumulation of peripheral myeloid-derived suppressor cells (MDSC), which play an important role in immune escape. However, the cellular and molecular mechanisms by which tumors induce extramedullary myelopoiesis are poorly understood. In this study, we found that osteopontin expressed by tumor cells enhances extramedullary myelopoiesis in a CD44-dependent manner through the Erk1/2-MAPK pathway. Osteopontin-mediated extramedullary myelopoiesis was directly associated with increased MDSCs in tumor-bearing hosts. More importantly, osteopontin silencing in tumor cells delayed both tumor growth and extramedullary myelopoiesis, while the same treatment did not affect tumor growth in vitro. Finally, treatment with an antibody against osteopontin inhibited tumor growth and synergized with cell-based immunotherapeutic vaccines in mediating antitumor immunity. Our findings unveil a novel immunosuppressive role for tumor-derived osteopontin and offer a rationale for its therapeutic targeting in cancer treatment.


Subject(s)
Myelopoiesis , Neoplasms, Experimental/immunology , Osteopontin/physiology , Animals , Cancer Vaccines/immunology , Cell Line, Tumor , Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/physiology , Hyaluronan Receptors/physiology , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/physiology , Stem Cells/physiology
10.
Eur J Immunol ; 44(6): 1672-84, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24659444

ABSTRACT

Myeloid-derived suppressor cells (MDSCs), which suppress diverse innate and adaptive immune responses and thereby provide an evasion mechanism for tumors, are emerging as a key population linking inflammation to cancer. Although many inflammatory factors that induce MDSCs in the tumor microenvironment are known, the crucial components and the underlying mechanisms remain elusive. In this study, we proposed a novel mechanism by which serum amyloid A3 (SAA3), a well-known inflammatory factor, connects MDSCs with cancer progression. We found that SAA3 expression in BALB/c mice increased in monocytic MDSCs (Mo MDSCs) with tumor growth. The induction of SAA3 by apo-SAA treatment in Mo MDSCs enhanced their survival and suppressive activity, while it inhibited GM-CSF-induced differentiation. Endogenous SAA3 itself contributed to the increase in the survival and suppressive activity of Mo MDSCs. We demonstrated that SAA3 induced TLR2 signaling, in turn increasing the autocrine secretion of TNF-α, that led to STAT3 activation. In addition, activated STAT3 enhanced the suppressive activity of Mo MDSCs. Furthermore, SAA3 induction in Mo MDSCs contributed to accelerating tumor progression in vivo. Collectively, these data suggest a novel mechanism by which Mo MDSCs mediate inflammation through SAA3-TLR2 signaling and thus exacerbate cancer progression by a STAT3-dependent mechanism.


Subject(s)
Myeloid Cells/immunology , Neoplasms, Experimental/immunology , STAT3 Transcription Factor/immunology , Serum Amyloid A Protein/immunology , Toll-Like Receptor 2/immunology , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Inflammation/immunology , Inflammation/pathology , Mice , Mice, Inbred BALB C , Myeloid Cells/pathology , Neoplasms, Experimental/pathology , Signal Transduction/immunology
11.
Cancer Res ; 73(18): 5669-81, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23867469

ABSTRACT

How myeloid-derived suppressor cells (MDSC) emerge in the tumor environment remains unclear. Here, we report that GM-CSF can convert natural killer (NK) cells into MDSCs. When transferred into tumor-bearing mice, adoptively transferred NK cells lost their NK phenotype and were converted into Ly6C(high)Ly6G(high) MDSC. This conversion was abolished by exposure to IL-2 either in vitro or in vivo. Notably, we found that of the 4 maturation stages based on CD11b/CD27 expression levels, only the CD11b(high)CD27(high) NK cells could be converted into CD11b(+)Gr1(+) MDSC ex vivo. Transfer of CD27(high) NK cells from tumor-bearing mice into tumor-bearing recipients was associated with conversion to MDSC in a manner associated with reduced numbers of CD11b(high)CD27(high) and CD11b(high)CD27(low) NK cell populations in the recipients. Our results identify a pathway of MDSC development from immature NK cells in tumor-bearing hosts, providing new insights into how tumor cells modulate their host immune microenvironment to escape immune surveillance.


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Killer Cells, Natural/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid Cells/immunology , Neoplasms/immunology , Tumor Microenvironment , Animals , Arginase/genetics , Arginase/metabolism , Blotting, Western , CD11b Antigen/genetics , CD11b Antigen/metabolism , Cell Differentiation , Cell Proliferation , Flow Cytometry , Humans , Immunologic Surveillance , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasms/metabolism , Neoplasms/pathology , Nitric Oxide/metabolism , RNA, Messenger/genetics , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
12.
Cancer Res ; 71(24): 7442-51, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22028323

ABSTRACT

Dendritic cells (DC) present α-galactosylceramide (αGalCer) to invariant T-cell receptor-expressing natural killer T cells (iNKT) activating these cells to secrete a variety of cytokines, which in turn results in DC maturation and activation of other cell types, including NK cells, B cells, and conventional T cells. In this study, we showed that αGalCer-pulsing of antigen-activated CD8 T cells before adoptive transfer to tumor-bearing mice caused a marked increase in donor T-cell proliferation, precursor frequency, and cytotoxic lymphocyte activity. This effect was interleukin (IL)-2 dependent and involved both natural killer T cells (NKT) and DCs, as mice lacking IL-2, NKTs, and DCs lacked any enhanced response to adoptively transferred αGalCer-loaded CD8 T cells. iNKT activation was mediated by transfer of αGalCer from the cell membrane of the donor CD8 T cells onto the αGalCer receptor CD1d which is present on host DCs. αGalCer transfer was increased by prior activation of the donor CD8 T cells and required AP-2-mediated endocytosis by host DCs. In addition, host iNKT cell activation led to strong IL-2 synthesis, thereby increasing expansion and differentiation of donor CD8 T cells. Transfer of these cells led to improved therapeutic efficacy against established solid tumors in mice. Thus, our findings illustrate how αGalCer loading of CD8 T cells after antigen activation in vitro may leverage the therapeutic potential of adoptive T-cell therapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Galactosylceramides/immunology , Neoplasms/immunology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Dendritic Cells/metabolism , Endocytosis/immunology , Flow Cytometry , Galactosylceramides/pharmacokinetics , Immunotherapy, Adoptive , Interferon-gamma/genetics , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-2/genetics , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Neoplasms/pathology , Neoplasms/therapy , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...