Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 668307, 2021.
Article in English | MEDLINE | ID: mdl-34489927

ABSTRACT

Management for high-risk neuroblastoma (NBL) has included autologous hematopoietic stem cell transplant (HSCT) and anti-GD2 immunotherapy, but survival remains around 50%. The aim of this study was to determine if allogeneic HSCT could serve as a platform for inducing a graft-versus-tumor (GVT) effect against NBL with combination immunocytokine and NK cells in a murine model. Lethally irradiated C57BL/6 (B6) x A/J recipients were transplanted with B6 bone marrow on Day +0. On day +10, allogeneic HSCT recipients were challenged with NXS2, a GD2+ NBL. On days +14-16, mice were treated with the anti-GD2 immunocytokine hu14.18-IL2. In select groups, hu14.18-IL2 was combined with infusions of B6 NK cells activated with IL-15/IL-15Rα and CD137L ex vivo. Allogeneic HSCT alone was insufficient to control NXS2 tumor growth, but the addition of hu14.18-IL2 controlled tumor growth and improved survival. Adoptive transfer of ex vivo CD137L/IL-15/IL-15Rα activated NK cells with or without hu14.18-IL2 exacerbated lethality. CD137L/IL-15/IL-15Rα activated NK cells showed enhanced cytotoxicity and produced high levels of TNF-α in vitro, but induced cytokine release syndrome (CRS) in vivo. Infusing Perforin-/- CD137L/IL-15/IL-15Rα activated NK cells had no impact on GVT, whereas TNF-α-/- CD137L/IL-15/IL-15Rα activated NK cells improved GVT by decreasing peripheral effector cell subsets while preserving tumor-infiltrating lymphocytes. Depletion of Ly49H+ NK cells also improved GVT. Using allogeneic HSCT for NBL is a viable platform for immunocytokines and ex vivo activated NK cell infusions, but must be balanced with induction of CRS. Regulation of TNFα or activating NK subsets may be needed to improve GVT effects.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Cytokines/pharmacology , Gangliosides/antagonists & inhibitors , Graft vs Tumor Effect , Hematopoietic Stem Cell Transplantation , Immunotherapy, Adoptive , Killer Cells, Natural/drug effects , Killer Cells, Natural/transplantation , Lymphocyte Activation/drug effects , Neuroblastoma/therapy , Animals , Cell Line, Tumor , Combined Modality Therapy , Female , Gangliosides/immunology , Gangliosides/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neuroblastoma/immunology , Neuroblastoma/metabolism , Neuroblastoma/pathology
2.
Leuk Lymphoma ; 62(5): 1167-1177, 2021 05.
Article in English | MEDLINE | ID: mdl-33843403

ABSTRACT

Targeting the JAK/STAT and BCL2 pathways in patients with relapsed/refractory T cell acute lymphoblastic leukemia (T-ALL) may provide an alternative approach to achieve clinical remissions. Ruxolitinib and venetoclax show a dose-dependent effect on T-ALL individually, but combination treatment reduces survival and proliferation of T-ALL in vitro. Using a xenograft model, the combination treatment fails to improve survival, with death from hind limb paralysis. Despite on-target inhibition by the drugs, histopathology demonstrates increased leukemic infiltration into the central nervous system (CNS) as compared to liver or bone marrow. Liquid chromatography-tandem mass spectroscopy shows that ruxolitinib and venetoclax insufficiently cross into the CNS. The addition of the CXCR4 inhibitor plerixafor with ruxolitinib and venetoclax reduces clinical scores and enhances survival. While combination therapy with ruxolitinib and venetoclax shows promise for treating T-ALL, additional inhibition of the CXCR4-CXCL12 axis may be needed to maximize the possibility of complete remission.


Subject(s)
Precursor T-Cell Lymphoblastic Leukemia-Lymphoma , Receptors, CXCR4 , Benzylamines , Bridged Bicyclo Compounds, Heterocyclic , Central Nervous System , Cyclams , Hematopoietic Stem Cell Mobilization , Humans , Janus Kinase 1 , Nitriles , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proto-Oncogene Proteins c-bcl-2/genetics , Pyrazoles , Pyrimidines , Sulfonamides
3.
Cytotherapy ; 22(8): 450-457, 2020 08.
Article in English | MEDLINE | ID: mdl-32536506

ABSTRACT

BACKGROUND AIMS: Several methods to expand and activate (EA) NK cells ex vivo have been developed for the treatment of relapsed or refractory cancers. Infusion of fresh NK cells is generally preferred to the infusion of cryopreserved/thawed (C/T) NK cells because of concern that cryopreservation diminishes NK cell activity. However, there has been little head-to-head comparison of the functionality of fresh versus C/T NK cell products. METHODS: We evaluated activity of fresh and C/T EA NK cells generated by interleukin (IL)-15, IL-2 and CD137L expansion. RESULTS: Analysis of C/T NK cell products demonstrated decreased recovery of viable CD56+ cells, but the proportion of NK cells in the C/T EA NK cell product did not decrease compared with the fresh EA NK cell product. Fresh and C/T EA NK cells demonstrated increased granzyme B compared with NK cells pre-expansion, but only fresh EA NK cells showed increased NKG2D. Compared with fresh EA NK cells, cytotoxic ability of C/T EA NK cells was reduced, but C/T EA NK cells remained potently cytotoxic against tumor cells via both antibody-independent and antibody-dependent mechanisms within 4 h post-thaw. Fresh EA NK cells generated high levels of gamma interferon (IFN-γ), which was abrogated by JAK1/JAK2 inhibition with ruxolitinib, but C/T EA NK cells showed lower IFN-γ unaffected by JAK1/JAK2 inhibition. DISCUSSION: Usage of C/T EA NK cells may be an option to provide serial "boost" NK cell infusions from a single apheresis to maximize NK cell persistence and potentially improve NK-induced responses to refractory cancer.


Subject(s)
Cell Culture Techniques/methods , Cryopreservation , Killer Cells, Natural/cytology , Lymphocyte Activation/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Cytotoxicity, Immunologic/drug effects , Granzymes/metabolism , Humans , Interferon-gamma/metabolism , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Lymphocyte Activation/drug effects , Lymphocyte Count , Lymphocyte Subsets/cytology , Lymphocyte Subsets/drug effects , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nitriles , Pyrazoles/pharmacology , Pyrimidines
4.
Mol Cancer Ther ; 19(3): 835-846, 2020 03.
Article in English | MEDLINE | ID: mdl-31879362

ABSTRACT

Inhibitors of adaptive immune checkpoints have shown promise as cancer treatments. CD47 is an innate immune checkpoint receptor broadly expressed on normal tissues and overexpressed on many tumors. Binding of tumor CD47 to signal regulatory protein alpha (SIRPα) on macrophages and dendritic cells triggers a "don't eat me" signal that inhibits phagocytosis enabling escape of innate immune surveillance. Blocking CD47/SIRPα interaction promotes phagocytosis reducing tumor burden in numerous xenograft and syngeneic animal models. We have developed a next-generation humanized anti-CD47 antibody, AO-176, that not only blocks the CD47/SIRPα interaction to induce tumor cell phagocytosis, but also induces tumor cytotoxicity in hematologic and solid human tumor cell lines, but not normal noncancerous cells, by a cell autonomous mechanism (not ADCC). AO-176 also binds preferentially to tumor versus many normal cell types. In particular, AO-176 binds negligibly to RBCs in contrast to tumor cells, even at high concentrations up to 200 µg/mL and does not agglutinate RBCs up to 1 mg/mL in vitro These properties are expected not only to decrease the antigen sink, but also to minimize on-target clinical adverse effects observed following treatment with other reported RBC-binding anti-CD47 antibodies. When tested in cynomolgus monkeys, AO-176 was well tolerated with no adverse effects. Finally, we show that AO-176 demonstrates dose-dependent antitumor activity in tumor xenograft models. Taken together, the unique properties and antitumor activity of our next-generation anti-CD47 antibody, AO-176, distinguishes it from other CD47/SIRPα axis targeting agents in clinical development.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , CD47 Antigen/antagonists & inhibitors , Erythrocytes/metabolism , Immunity, Innate/immunology , Neoplasms/drug therapy , Phagocytosis , Receptors, Immunologic/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/immunology , Antigens, Differentiation/immunology , Apoptosis , CD47 Antigen/immunology , Cell Proliferation , Female , Humans , Immunity, Innate/drug effects , Macaca fascicularis , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/immunology , Neoplasms/pathology , Receptors, Immunologic/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
5.
Front Immunol ; 9: 150, 2018.
Article in English | MEDLINE | ID: mdl-29456538

ABSTRACT

Recent advances in cellular therapies for patients with cancer, including checkpoint blockade and ex vivo-expanded, tumor-specific T cells, have demonstrated that targeting the immune system is a powerful approach to the elimination of tumor cells. Clinical efforts have also demonstrated limitations, however, including the potential for tumor cell antigenic drift and neoantigen formation, which promote tumor escape and recurrence, as well as rapid onset of T cell exhaustion in vivo. These findings suggest that antigen unrestricted cells, such as natural killer (NK) cells, may be beneficial for use as an alternative to or in combination with T cell based approaches. Although highly effective in lysing transformed cells, to date, few clinical trials have demonstrated antitumor function or persistence of transferred NK cells. Several recent studies describe methods to expand NK cells for adoptive transfer, although the effects of ex vivo expansion are not fully understood. We therefore explored the impact of a clinically validated 12-day expansion protocol using a K562 cell line expressing membrane-bound IL-15 and 4-1BB ligand with high-dose soluble IL-2 on the phenotype and functions of NK cells from healthy donors. Following expansions using this protocol, we found expression of surface proteins that implicate preferential expansion of NK cells that are not fully mature, as is typically associated with highly cytotoxic NK cell subsets. Despite increased expression of markers associated with functional exhaustion in T cells, we found that ex vivo-expanded NK cells retained cytokine production capacity and had enhanced tumor cell cytotoxicity. The preferential expansion of an NK cell subset that is phenotypically immature and functionally pleiotropic suggests that adoptively transferred cells may persist better in vivo when compared with previous methods using this approach. Ex vivo expansion does not quell killer immunoglobulin-like receptor diversity, allowing responsiveness to various factors in vivo that may influence activation and inhibition. Collectively, our data suggest that in addition to robust NK cell expansion that has been described using this method, expanded NK cells may represent an ideal cell therapy that is longer lived, highly potent, and responsive to an array of activating and inhibitory signals.


Subject(s)
Killer Cells, Natural/immunology , 4-1BB Ligand/immunology , Humans , Interleukin-15/immunology , Interleukin-2/immunology , K562 Cells , Phenotype
6.
Biol Blood Marrow Transplant ; 23(6): 897-905, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28257800

ABSTRACT

Mesenchymal stem cells (MSCs) have immunosuppressive and tissue repair properties, but clinical trials using MSCs to prevent or treat graft-versus-host disease (GVHD) have shown mixed results. Macrophages (MØs) are important regulators of immunity and can promote tissue regeneration and remodeling. We have previously shown that MSCs can educate MØs toward a unique anti-inflammatory immunophenotype (MSC-educated MØs [MEMs]); however, their implications for in vivo models of inflammation have not been studied yet. We now show that in comparison with MØs, MEMs have increased expression of the inhibitory molecules PD-L1, PD-L2, in addition to markers of alternatively activated MØs: CD206 and CD163. RNA-Seq analysis of MEMs, as compared with MØs, show a distinct gene expression profile that positively correlates with multiple pathways important in tissue repair. MEMs also show increased expression of IL-6, transforming growth factor-ß, arginase-1, CD73, and decreased expression of IL-12 and tumor necrosis factor-α. We show that IL-6 secretion is controlled in part by the cyclo-oxygenase-2, arginase, and JAK1/STAT1 pathway. When tested in vivo, we show that human MEMs significantly enhance survival from lethal GVHD and improve survival of mice from radiation injury. We show these effects could be mediated in part through suppression of human T cell proliferation and may have attenuated host tissue injury in part by enhancing murine fibroblast proliferation. MEMs are a unique MØ subset with therapeutic potential for the management of GVHD and/or protection from radiation-induced injury.


Subject(s)
Cell Communication/immunology , Cell- and Tissue-Based Therapy/methods , Graft vs Host Disease/therapy , Macrophages/immunology , Mesenchymal Stem Cells/immunology , Radiation Injuries/therapy , Animals , Humans , Inflammation/immunology , Interleukin-6/biosynthesis , Macrophage Activation/immunology , Macrophages/cytology , Mesenchymal Stem Cells/cytology , Mice
7.
Oncoimmunology ; 5(5): e1143996, 2016 May.
Article in English | MEDLINE | ID: mdl-27467963

ABSTRACT

The availability of clinical-grade cytokines and artificial antigen-presenting cells has accelerated interest in using natural killer (NK) cells as adoptive cellular therapy (ACT) for cancer. One of the technological shortcomings of translating therapies from animal models to clinical application is the inability to effectively and non-invasively track these cells after infusion in patients. We have optimized the nonradioactive isotope fluorine-19 ((19)F) as a means to label and track NK cells in preclinical models using magnetic resonance imaging (MRI). Human NK cells were expanded with interleukin (IL)-2 and labeled in vitro with increasing concentrations of (19)F. Doses as low as 2 mg/mL (19)F were detected by MRI. NK cell viability was only decreased at 8 mg/mL (19)F. No effects on NK cell cytotoxicity against K562 leukemia cells were observed with 2, 4 or 8 mg/mL (19)F. Higher doses of (19)F, 4 mg/mL and 8 mg/mL, led to an improved (19)F signal by MRI with 3 × 10(11) (19)F atoms per NK cell. The 4 mg/mL (19)F labeling had no effect on NK cell function via secretion of granzyme B or interferon gamma (IFNγ), compared to NK cells exposed to vehicle alone. (19)F-labeled NK cells were detectable immediately by MRI after intratumoral injection in NSG mice and up to day 8. When (19)F-labeled NK cells were injected subcutaneously, we observed a loss of signal through time at the site of injection suggesting NK cell migration to distant organs. The (19)F perfluorocarbon is a safe and effective reagent for monitoring the persistence and trafficking of NK cell infusions in vivo, and may have potential for developing novel imaging techniques to monitor ACT for cancer.

8.
Cancer Immunol Immunother ; 64(12): 1541-52, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26423422

ABSTRACT

We have demonstrated that immunostimulatory therapies such as interleukin-2 (IL-2) and anti-CD40 (αCD40) can be combined to deliver synergistic anti-tumor effects. While this strategy has shown success, efficacy varies depending on a number of factors including tumor type and severe toxicities can be seen. We sought to determine whether blockade of negative regulators such as cytotoxic T lymphocyte antigen-4 (CTLA-4) could simultaneously prolong CD8(+) T cell responses and augment T cell anti-tumor effects. We devised a regimen in which anti-CTLA-4 was administered late so as to delay contraction and minimize toxicities. This late administration both enhanced and prolonged CD8 T cell activation without the need for additional IL-2. The quality of the T cell response was improved with increased frequency of effector/effector memory phenotype cells along with improved lytic ability and bystander expansion. This enhanced CD8 response translated to improved anti-tumor responses both at the primary and metastatic sites. Importantly, toxicities were not exacerbated with combination. This study provides a platform for rational design of immunotherapy combinations to maximize anti-tumor immunity while minimizing toxicities.


Subject(s)
CD8 Antigens/metabolism , CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy , Neoplasms/immunology , Recombinant Fusion Proteins/pharmacology , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Flow Cytometry , Humans , Interleukin-2/pharmacology , Mice , Mice, Inbred C57BL , Neoplasms/mortality , Neoplasms/therapy , Recombinant Fusion Proteins/administration & dosage
9.
Immunity ; 43(2): 240-50, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26231116

ABSTRACT

Primary T cell activation involves the integration of three distinct signals delivered in sequence: (1) antigen recognition, (2) costimulation, and (3) cytokine-mediated differentiation and expansion. Strong immunostimulatory events such as immunotherapy or infection induce profound cytokine release causing "bystander" T cell activation, thereby increasing the potential for autoreactivity and need for control. We show that during strong stimulation, a profound suppression of primary CD4(+) T-cell-mediated immune responses ensued and was observed across preclinical models and patients undergoing high-dose interleukin-2 (IL-2) therapy. This suppression targeted naive CD4(+) but not CD8(+) T cells and was mediated through transient suppressor of cytokine signaling-3 (SOCS3) inhibition of the STAT5b transcription factor signaling pathway. These events resulted in complete paralysis of primary CD4(+) T cell activation, affecting memory generation and induction of autoimmunity as well as impaired viral clearance. These data highlight the critical regulation of naive CD4(+) T cells during inflammatory conditions.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Herpesviridae Infections/therapy , Immunotherapy/methods , Melanoma/therapy , Muromegalovirus/immunology , Skin Neoplasms/therapy , Animals , Antigens/immunology , Cell Differentiation/genetics , Cell Proliferation/genetics , Clonal Anergy , Female , Herpesviridae Infections/immunology , Humans , Immunity, Cellular , Immunologic Memory , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-2/administration & dosage , Melanoma/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microarray Analysis , Randomized Controlled Trials as Topic , Signal Transduction , Skin Neoplasms/immunology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/genetics , Suppressor of Cytokine Signaling Proteins/metabolism , Viral Load/immunology
10.
J Exp Med ; 211(12): 2373-83, 2014 Nov 17.
Article in English | MEDLINE | ID: mdl-25366964

ABSTRACT

Aging is a contributing factor in cancer occurrence. We recently demonstrated that systemic immunotherapy (IT) administration in aged, but not young, mice resulted in induction of rapid and lethal cytokine storm. We found that aging was accompanied by increases in visceral fat similar to that seen in young obese (ob/ob or diet-induced obese [DIO]) mice. Yet, the effects of aging and obesity on inflammatory responses to immunotherapeutics are not well defined. We determine the effects of adiposity on systemic IT tolerance in aged compared with young obese mice. Both young ob/ob- and DIO-generated proinflammatory cytokine levels and organ pathologies are comparable to those in aged ad libitum mice after IT, culminating in lethality. Young obese mice exhibited greater ratios of M1/M2 macrophages within the peritoneal and visceral adipose tissues and higher percentages of TNF(+) macrophages in response to αCD40/IL-2 as compared with young lean mice. Macrophage depletion or TNF blockade in conjunction with αCD40/IL-2 prevented cytokine storms in young obese mice and protected from lethality. Calorie-restricted aged mice contain less visceral fat and displayed reduced cytokine levels, protection from organ pathology, and protection from lethality upon αCD40/IL-2 administration. Our data demonstrate that adiposity is a critical factor in the age-associated pathological responses to systemic anti-cancer IT.


Subject(s)
Adiposity/immunology , Aging/immunology , Cytokines/immunology , Immunotherapy/methods , Animals , Caloric Restriction , Cytokines/blood , Female , Flow Cytometry , Humans , Inflammation Mediators/blood , Inflammation Mediators/immunology , Intra-Abdominal Fat/immunology , Intra-Abdominal Fat/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Obese , Neoplasms/immunology , Neoplasms/therapy , Obesity/genetics , Obesity/immunology , Obesity/metabolism , Tumor Necrosis Factor-alpha/immunology , Tumor Necrosis Factor-alpha/metabolism
11.
J Immunol ; 193(4): 1709-16, 2014 Aug 15.
Article in English | MEDLINE | ID: mdl-25000978

ABSTRACT

Because of increasing interest in the removal of immunosuppressive pathways in cancer, the combination of IL-2 with Abs to neutralize TGF-ß, a potent immunosuppressive cytokine, was assessed. Combination immunotherapy resulted in significantly greater antitumor effects. These were correlated with significant increases in the numbers and functionality of NK cells, NK cell progenitors, and activated CD8 T cells, resulting in the observed antitumor effects. Combination immunotherapy also was accompanied by lesser toxicities than was IL-2 therapy alone. Additionally, we observed a dual competition between NK cells and activated CD8 T cells such that, after immunotherapy, the depletion of either effector population resulted in the increased total expansion of the other population and compensatory antitumor effects. This study demonstrates the efficacy of this combination immunotherapeutic regimen as a promising cancer therapy and illustrates the existence of potent competitive regulatory pathways between NK cells and CD8 T cells in response to systemic activation.


Subject(s)
Antibodies, Neutralizing/therapeutic use , CD8-Positive T-Lymphocytes/immunology , Interleukin-2/therapeutic use , Killer Cells, Natural/immunology , Neoplasms/therapy , Transforming Growth Factor beta/antagonists & inhibitors , Animals , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Fas Ligand Protein/immunology , Female , Immunotherapy/methods , Killer Cells, Natural/transplantation , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily A/biosynthesis , NK Cell Lectin-Like Receptor Subfamily C/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , T-Lymphocytes, Regulatory/immunology , fas Receptor/immunology
12.
J Exp Med ; 210(11): 2223-37, 2013 Oct 21.
Article in English | MEDLINE | ID: mdl-24081947

ABSTRACT

Cancer commonly occurs in the elderly and immunotherapy (IT) is being increasingly applied to this population. However, the majority of preclinical mouse tumor models assessing potential efficacy and toxicities of therapeutics use young mice. We assessed the impact of age on responses to systemic immune stimulation. In contrast to young mice, systemic cancer IT regimens or LPS given to aged mice resulted in rapid and lethal toxicities affecting multiple organs correlating with heightened proinflammatory cytokines systemically and within the parenchymal tissues. This inflammatory response and increased morbidity with age was independent of T cells or NK cells. However, prior in vivo depletion of macrophages in aged mice resulted in lesser cytokine levels, increased survival, and decreased liver histopathology. Furthermore, macrophages from aged mice and normal human elderly volunteers displayed heightened TNF and IL-6 production upon in vitro stimulation. Treatment of both TNF knockout mice and in vivo TNF blockade in aged mice resulted in significant increases in survival and lessened pathology. Importantly, TNF blockade in tumor-bearing, aged mice receiving IT displayed significant anti-tumor effects. These data demonstrate the critical role of macrophages in the age-associated hyper-inflammatory cytokine responses to systemic immunostimulation and underscore the importance of performing preclinical assessments in aged mice.


Subject(s)
Aging/immunology , Aging/pathology , Immunotherapy , Inflammation/pathology , Neoplasms/immunology , Neoplasms/pathology , Animals , CD40 Antigens/immunology , Cytokines/toxicity , Dose-Response Relationship, Drug , Female , Humans , Inflammation Mediators/metabolism , Lipopolysaccharides/pharmacology , Liver/drug effects , Liver/enzymology , Liver/pathology , Macrophages/drug effects , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Neoplasms/therapy , Survival Analysis , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism
13.
Oncoimmunology ; 2(12): e27186, 2013 Dec 01.
Article in English | MEDLINE | ID: mdl-24498569

ABSTRACT

Cancer immunotherapy holds great promise, yet its efficacy and applicability can be hampered by the rise of systemic toxicities. We have recently shown that the lethal side effects of cancer immunotherapy are markedly exacerbated with aging. Blocking tumor necrosis factor α or macrophages can alleviate the systemic toxicity of immunotherapy while preserving its antineoplastic effects.

14.
PLoS One ; 7(10): e48049, 2012.
Article in English | MEDLINE | ID: mdl-23133545

ABSTRACT

The primary tumor represents a potential source of antigens for priming immune responses for disseminated disease. Current means of debulking tumors involves the use of cytoreductive conditioning that impairs immune cells or removal by surgery. We hypothesized that activation of the immune system could occur through the localized release of tumor antigens and induction of tumor death due to physical disruption of tumor architecture and destruction of the primary tumor in situ. This was accomplished by intratumor injection of magneto-rheological fluid (MRF) consisting of iron microparticles, in Balb/c mice bearing orthotopic 4T1 breast cancer, followed by local application of a magnetic field resulting in immediate coalescence of the particles, tumor cell death, slower growth of primary tumors as well as decreased tumor progression in distant sites and metastatic spread. This treatment was associated with increased activation of DCs in the draining lymph nodes and recruitment of both DCs and CD8(+)T cells to the tumor. The particles remained within the tumor and no toxicities were observed. The immune induction observed was significantly greater compared to cryoablation. Further anti-tumor effects were observed when MRF/magnet therapy was combined with systemic low dose immunotherapy. Thus, mechanical disruption of the primary tumor with MRF/magnetic field application represents a novel means to induce systemic immune activation in cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Iron/pharmacology , Mammary Neoplasms, Animal/radiotherapy , Animals , CD8-Positive T-Lymphocytes/cytology , Dendritic Cells/cytology , Disease Progression , Female , Flow Cytometry/methods , Immune System , Immunotherapy/methods , Magnetic Fields , Magnetics , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/therapy , Mice , Mice, Inbred BALB C , Necrosis , Neoplasm Metastasis , Neoplasms/immunology , Stem Cells
15.
Blood ; 119(13): 3073-83, 2012 Mar 29.
Article in English | MEDLINE | ID: mdl-22251483

ABSTRACT

Memory T cells exhibit tremendous antigen specificity within the immune system and accumulate with age. Our studies reveal an antigen-independent expansion of memory, but not naive, CD8(+) T cells after several immunotherapeutic regimens for cancer resulting in a distinctive phenotype. Signaling through T-cell receptors (TCRs) or CD3 in both mouse and human memory CD8(+) T cells markedly up-regulated programmed death-1 (PD-1) and CD25 (IL-2 receptor α chain), and led to antigen-specific tumor cell killing. In contrast, exposure to cytokine alone in vitro or with immunotherapy in vivo did not up-regulate these markers but resulted in expanded memory CD8(+) T cells expressing NKG2D, granzyme B, and possessing broadly lytic capabilities. Blockade of NKG2D in mice also resulted in significantly diminished antitumor effects after immunotherapy. Treatment of TCR-transgenic mice bearing nonantigen expressing tumors with immunotherapy still resulted in significant antitumor effects. Human melanoma tissue biopsies obtained from patients after topically applied immunodulatory treatment resulted in increased numbers of these CD8(+) CD25(-) cells within the tumor site. These findings demonstrate that memory CD8(+) T cells can express differential phenotypes indicative of adaptive or innate effectors based on the nature of the stimuli in a process conserved across species.


Subject(s)
CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Cytokines/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , T-Cell Antigen Receptor Specificity/immunology , Animals , Cells, Cultured , Double-Blind Method , Humans , Immunologic Memory/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Placebos , Randomized Controlled Trials as Topic , T-Cell Antigen Receptor Specificity/physiology , Time Factors
16.
Immunotherapy ; 2(3): 399-418, 2010 May.
Article in English | MEDLINE | ID: mdl-20635904

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) is a particularly important treatment for hematologic malignancies. Unfortunately, following allogeneic HSCT, graft-versus-host disease, immunosuppression and susceptibility to opportunistic infections remain among the most substantial problems restricting the efficacy and use of this procedure, particularly for cancer. Adoptive immunotherapy and/or manipulation of the graft offer ways to attack residual cancer as well as other transplant-related complications. Recent exciting discoveries have demonstrated that HSCT could be expanded to solid tissue cancers with profound effects on the effectiveness of adoptive immunotherapy. This review will provide a background regarding HSCT, discuss the complications that make it such a complex treatment procedure following up with current immunotherapeutic strategies and discuss emerging approaches in applying immunotherapy in HSCT for cancer.


Subject(s)
Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/methods , Immunotherapy, Adoptive/methods , Neoplasms/therapy , Animals , Combined Modality Therapy , Disease Models, Animal , Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Humans , Neoplasms/immunology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...