Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Blood ; 138(24): 2583-2588, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34424962

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains a potential curative option for treating a variety of hematologic diseases, but acute and chronic graft-versus-host disease (GVHD) remain major barriers limiting efficacy. Acute gut GVHD occurs with marked increases in proinflammatory cytokines (including TNF and IL-6), which we recently demonstrated was exacerbated in obesity resulting in severe gastrointestinal pathology. Given the pleiotropic and overlapping effects of these 2 cytokines, we assessed the impact of dual TNF and IL-6R blockade on GVHD as well as graft-versus tumor (GVT) effects in different mouse GVHD models. Early administration of combined blockade resulted in greater protection and survival from acute gut GVHD compared with single blockade regimens and even development of later chronic skin GVHD. Importantly, double cytokine blockade preserved GVT effects reinforcing that GVT and GVHD can be delineated and may result in greater efficacy in allo-HSCT.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Receptors, Interleukin-6/antagonists & inhibitors , Tumor Necrosis Factor Inhibitors/therapeutic use , Animals , Antibodies, Monoclonal/therapeutic use , Disease Models, Animal , Etanercept/therapeutic use , Female , Graft vs Tumor Effect/drug effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Mice, Inbred BALB C , Mice, Inbred C57BL , Transplantation, Homologous/methods
2.
Liver Transpl ; 25(8): 1251-1264, 2019 08.
Article in English | MEDLINE | ID: mdl-31152624

ABSTRACT

Liver transplantation (LT) is currently considered an important method in treating hepatocellular carcinoma (HCC) and an alternative treatment for other liver malignancies. Here, we demonstrated that the graft-versus-tumor (GVT) effect exists in allogeneic liver transplantation (allo LT). Recipient-derived T cells played a critical role in the GVT process of allo LT, as demonstrated by extensive infiltration and significant activation of recipient T cells in the tumor after surgery. Moreover, this process was related to donor-derived T/B cells by improving the immune microenvironment in the tumor, as demonstrated by elevated levels of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), interleukin-2 (IL-2), IL-6, IL-16, chemokine (C-X-C motif) ligand 10 (CXCL10), and CXCL11 and decreased levels of IL-10 and IL-4 at tumor sites. Additionally, tacrolimus (FK506) treatment inhibited the GVT effect on allo LT. Donor liver-derived T/B cells infiltrate extrahepatic tumors to trigger a strong T-cell-mediated immune response and thus improve the tumor immune microenvironment.


Subject(s)
Carcinoma, Hepatocellular/surgery , Graft vs Tumor Effect/immunology , Liver Neoplasms/surgery , Liver Transplantation , Tumor Microenvironment/immunology , Allografts/immunology , Animals , Carcinoma, Hepatocellular/immunology , Cytokines/immunology , Cytokines/metabolism , Disease Models, Animal , Graft vs Tumor Effect/drug effects , Humans , Immunosuppressive Agents/administration & dosage , Isoantigens/immunology , Liver/immunology , Liver Neoplasms/immunology , Male , Mice , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tacrolimus/administration & dosage , Transplantation, Homologous , Tumor Microenvironment/drug effects
3.
Leuk Lymphoma ; 60(6): 1528-1539, 2019 06.
Article in English | MEDLINE | ID: mdl-30501537

ABSTRACT

Graft-versus-host disease (GVHD) remains the least curable complication after allogeneic bone marrow transplantation (BMT). Myeloid differentiation factor 88 (MyD88) is an adaptor molecule critically involved in the toll-like receptor (TLR) signaling pathway. The Toll/IL-1 receptor (TIR) domains of MyD88 and TLR are interactional modules responsible for sorting and signaling via direct or indirect TIR-TIR interactions, which can contribute to all phases of GVHD progression. Here, we describe the mechanisms of the novel MyD88 inhibitor, TJ-M2010-5, and the discovery of its immunosuppressive properties in the context of GVHD and the graft-versus-tumor (GVT) effect in a fully MHC-mismatched murine model. TJ-M2010-5 potentially interrupted the conformation of the TIR domain through its predicted DD loops, BB loops, and Poc site, and inhibited the homodimerization of MyD88, the LPS-stimulated activation of dendritic cells, and the priming of donor allogeneic T cell proliferation in a dose-dependent manner. Oral administration of TJ-M2010-5 ameliorated the inflammatory environment, decreased the number of apoptotic cells, increased tissue repair in GVHD target organs, and suppressed lethal GVHD. Further, protection against GVHD by TJ-M2010-5 did not abrogate a GVT effect against SP2/0, a myeloma cell line. Our data define the mechanisms of actions and provide novel insight into the potential clinical uses of TJ-M2010-5 for GVHD prevention.


Subject(s)
Bone Marrow Transplantation/adverse effects , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/drug effects , Immunosuppressive Agents/pharmacology , Multiple Myeloma/therapy , Piperazines/pharmacology , Thiazoles/pharmacology , Administration, Oral , Animals , Bone Marrow Cells , Bone Marrow Transplantation/methods , Cell Line, Tumor/transplantation , Cell Proliferation , Dendritic Cells/immunology , Disease Models, Animal , Female , Graft vs Host Disease/immunology , HEK293 Cells , Humans , Immunosuppressive Agents/therapeutic use , Lymphocyte Activation/drug effects , Male , Mice , Multiple Myeloma/immunology , Myeloid Differentiation Factor 88/antagonists & inhibitors , Myeloid Differentiation Factor 88/immunology , Myeloid Differentiation Factor 88/metabolism , Piperazines/therapeutic use , Primary Cell Culture , Protein Domains/drug effects , Protein Domains/immunology , Protein Multimerization/drug effects , Protein Multimerization/immunology , Thiazoles/therapeutic use , Transplantation, Homologous/adverse effects , Transplantation, Homologous/methods
4.
Oncotarget ; 8(27): 44366-44378, 2017 Jul 04.
Article in English | MEDLINE | ID: mdl-28574833

ABSTRACT

Interleukin-15 (IL-15) is a potent cytokine that increases CD8+ T and NK cell numbers and function in experimental models. However, obstacles remain in using IL-15 therapeutically, specifically its low potency and short in vivo half-life. To help overcome this, a new IL-15 superagonist complex comprised of an IL-15N72D mutation and IL-15RαSu/Fc fusion (IL-15SA, also known as ALT-803) was developed. IL-15SA exhibits a significantly longer serum half-life and increased in vivo activity against various tumors. Herein, we evaluated the effects of IL-15SA in recipients of allogeneic hematopoietic stem cell transplantation. Weekly administration of IL-15SA to transplant recipients significantly increased the number of CD8+ T cells (specifically CD44+ memory/activated phenotype) and NK cells. Intracellular IFN-γ and TNF-α secretion by CD8+ T cells increased in the IL-15SA-treated group. IL-15SA also upregulated NKG2D expression on CD8+ T cells. Moreover, IL-15SA enhanced proliferation and cytokine secretion of adoptively transferred CFSE-labeled T cells in syngeneic and allogeneic models by specifically stimulating the slowly proliferative and nonproliferative cells into actively proliferating cells.We then evaluated IL-15SA's effects on anti-tumor activity against murine mastocytoma (P815) and murine B cell lymphoma (A20). IL-15SA enhanced graft-versus-tumor (GVT) activity in these tumors following T cell infusion. Interestingly, IL-15 SA administration provided GVT activity against A20 lymphoma cells in the murine donor leukocyte infusion (DLI) model without increasing graft versus host disease. In conclusion, IL-15SA could be a highly potent T- cell lymphoid growth factor and novel immunotherapeutic agent to complement stem cell transplantation and adoptive immunotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Graft vs Tumor Effect/drug effects , Proteins/pharmacology , Adoptive Transfer , Animals , Cell Line, Tumor , Disease Models, Animal , Female , Hematopoietic Stem Cell Transplantation , Interleukin-15/metabolism , Interleukin-15 Receptor alpha Subunit/agonists , Interleukin-15 Receptor alpha Subunit/metabolism , Lymphocyte Count , Mice , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Recombinant Fusion Proteins , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation, Homologous , Xenograft Model Antitumor Assays
5.
Cancer Immunol Res ; 5(8): 710-715, 2017 08.
Article in English | MEDLINE | ID: mdl-28637876

ABSTRACT

Although allogeneic stem cell transplantation (allo-SCT) can elicit graft-versus-tumor (GVT) immunity, patients often relapse due to residual tumor cells. As essential orchestrators of the immune system, vaccination with dendritic cells (DC) is an appealing strategy to boost the GVT response. Nevertheless, durable clinical responses after DC vaccination are still limited, stressing the need to improve current DC vaccines. Aiming to empower DC potency, we engineered monocyte-derived DCs to deprive them of ligands for the immune checkpoint regulated by programmed death 1 (PD-1). We also equipped them with interleukin (IL)-15 "transpresentation" skills. Transfection with short interfering (si)RNA targeting the PD-1 ligands PD-L1 and PD-L2, in combination with IL15 and IL15Rα mRNA, preserved their mature DC profile and rendered the DCs superior in inducing T-cell proliferation and IFNγ and TNFα production. Translated into an ex vivo hematological disease setting, DCs deprived of PD-1 ligands (PD-L), equipped with IL15/IL15Rα expression, or most effectively, both, induced superior expansion of minor histocompatibility antigen-specific CD8+ T cells from transplanted cancer patients. These data support the combinatorial approach of in situ suppression of the PD-L inhibitory checkpoints with DC-mediated IL15 transpresentation to promote antigen-specific T-cell responses and, ultimately, contribute to GVT immunity. Cancer Immunol Res; 5(8); 710-5. ©2017 AACR.


Subject(s)
Cancer Vaccines/administration & dosage , Dendritic Cells/transplantation , Interleukin-15/genetics , Programmed Cell Death 1 Receptor/genetics , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Dendritic Cells/immunology , Graft vs Tumor Effect/drug effects , Graft vs Tumor Effect/immunology , Humans , Interleukin-15/antagonists & inhibitors , Monocytes/immunology , Monocytes/transplantation , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , RNA, Small Interfering/genetics , Stem Cell Transplantation , Transfection , Transplantation, Homologous , Vaccination
6.
Br J Haematol ; 175(3): 505-516, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27447780

ABSTRACT

Given that donor T cells from a transplant contribute both the desired graft-versus-tumour (GVT) effect and detrimental graft-versus-host disease (GVHD), strategies to separate GVHD and GVT activity are a major clinical goal. We have previously demonstrated that in vivo administration of a recombinant (r)IL-7/HGFß hybrid cytokine, consisting of interleukin-7 (IL-7, IL7) and the ß-chain of hepatocyte growth factor (HGFß), significantly inhibits the growth of cancer cells in murine tumour models. The antit-umour effect of rIL-7/HGFß is related to a marked infiltration T cells in the tumour tissues. We have also shown that GVHD was not induced in rIL-7/HGFß-treated T cell-depleted allogeneic haematopoietic stem cell transplantation (HSCT) recipients. We show here that, in T cell-replete allogeneic HSCT murine models, rIL-7/HGFß attenuated acute GVHD (aGVHD), while promoting GVT activity. This was related to an alteration of donor T cell trafficking, with an increased infiltration of donor T cells into tumour tissues and the lympho-haematopoietic system but decreased number of the T cells in the GVHD target organs. Therefore, rIL-7/HGFß may offer a new tool to alleviate aGVHD while prompting GVT, and to study the molecular regulation of T cell trafficking.


Subject(s)
Chemotaxis/drug effects , Graft vs Host Disease/immunology , Graft vs Tumor Effect/drug effects , Hepatocyte Growth Factor/pharmacology , Interleukin-7/pharmacology , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Tissue Donors , Animals , Cell Line, Tumor , Chemokines/metabolism , Chemotaxis/immunology , Cytokines/metabolism , Disease Models, Animal , Graft vs Host Disease/drug therapy , Graft vs Host Disease/mortality , Graft vs Tumor Effect/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Hepatocyte Growth Factor/genetics , Interleukin-7/genetics , Mice , Neoplasms/complications , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Recombinant Fusion Proteins/genetics , T-Lymphocytes/metabolism , Tumor Burden/drug effects
7.
Blood ; 125(23): 3655-63, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25788701

ABSTRACT

In allogeneic hematopoietic stem cell transplantation (HSCT), controlling graft-versus-host disease (GVHD) while maintaining graft-versus-tumor (GVT) responses is of critical importance. Using a mouse model of allogeneic HSCT, we hereby demonstrate that NKG2D expression by CD8(+) T cells plays a major role in mediating GVHD and GVT effects by promoting the survival and cytotoxic function of CD8(+) T cells. The expression of NKG2D ligands was not induced persistently on normal tissues of allogeneic HSCT-recipient mice treated with anti-NKG2D antibody, suggesting that transient NKG2D blockade might be sufficient to attenuate GVHD and allow CD8(+) T cells to regain their GVT function. Indeed, short-term treatment with anti-NKG2D antibody restored GVT effects while maintaining an attenuated GVHD state. NKG2D expression was also detected on CD8(+) T cells from allogeneic HSCT patients and trended to be higher in those with active GVHD. Together, these data support a novel role for NKG2D expression by CD8(+) T cells during allogeneic HSCT, which could be potentially therapeutically exploited to separate GVHD from GVT effects.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation/immunology , Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Hematopoietic Stem Cell Transplantation , NK Cell Lectin-Like Receptor Subfamily K/immunology , Allografts , Animals , Antibodies, Neutralizing/pharmacology , CD8-Positive T-Lymphocytes/pathology , Disease Models, Animal , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Graft vs Tumor Effect/drug effects , Graft vs Tumor Effect/genetics , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/genetics
8.
Exp Hematol ; 42(10): 862-6.e3, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24971697

ABSTRACT

Indoleamine 2,3-dioxygenase (IDO) is a rate-limiting enzyme in tryptophan catabolism that plays an important role in the induction of immune tolerance. Its role in graft-versus-tumor effect after allogeneic stem cell transplantation (allo-SCT) remains unclear. Using a murine graft-versus-tumor model of reduced-intensity allo-HSCT followed by donor leukocyte infusion (DLI), we examined the role of IDO inhibition. Two stereoisomers of 1-methyl tryptophan (1-MT), a small-molecule inhibitor of IDO, reduced the growth of inoculated tumor in the mice that received DLI and had higher expression of IDO1 and IFNγ. However, L-1MT, but not D-1MT, mitigated tumor growth in mice that did not receive DLI and did not express IDO1 and IFNγ. Accordingly, both stereoisomers reduced plasma kynurenine concentrations early after DLI and enhanced in vitro cytotoxic lymphocyte function after allogeneic mixed lymphocyte reaction. Furthermore, L-1MT was more efficient in causing direct cytotoxic effects than D-1MT. Our results suggest that IDO inhibition can benefit anti-tumor therapy in the setting of reduced-intensity allo-SCT using DLI.


Subject(s)
Graft vs Tumor Effect/physiology , Immunologic Factors/pharmacology , Indoleamine-Pyrrole 2,3,-Dioxygenase/antagonists & inhibitors , Mastocytoma/therapy , Neoplasm Proteins/antagonists & inhibitors , Tryptophan/analogs & derivatives , Allografts , Animals , Bone Marrow Transplantation , Cell Line, Tumor , Cytotoxicity, Immunologic , Drug Evaluation, Preclinical , Enzyme Induction , Graft vs Tumor Effect/drug effects , Immunologic Factors/therapeutic use , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Indoleamine-Pyrrole 2,3,-Dioxygenase/physiology , Interferon-gamma/biosynthesis , Kynurenine/biosynthesis , Kynurenine/blood , Leukocyte Transfusion , Lymph Nodes/enzymology , Lymphocyte Culture Test, Mixed , Mastocytoma/drug therapy , Mastocytoma/enzymology , Mastocytoma/immunology , Mice , Mice, Inbred C57BL , Neoplasm Proteins/physiology , Radiation Chimera , Spleen/enzymology , Stereoisomerism , Time Factors , Transplantation Chimera , Tryptophan/chemistry , Tryptophan/metabolism , Tryptophan/pharmacology , Tryptophan/therapeutic use
9.
Br J Haematol ; 160(2): 133-45, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23121307

ABSTRACT

A major challenge to transplant immunologists and physicians remains the separation of harmful graft-versus-host disease (GvHD) and beneficial graft-versus-tumour (GvT) effects after allogeneic haematopoietic stem cell transplantation. Recent advances in our understanding of the allogeneic immune response provide potential new opportunities to achieve this goal. Three potential new approaches that capitalize on this new knowledge are considered in depth; the manipulation of organ-specific cytokines and other pro-inflammatory signals, the selective manipulation of donor effector T cell migration, and the development of cell-mediated immunosuppressive strategies using donor-derived regulatory T cells. These new approaches could provide strategies for local control of allogeneic immune responses, a new paradigm to separate GvHD and GvT effects. Although these strategies are currently in their infancy and have challenges to successful translation to clinical practice, all have exciting potential for the future.


Subject(s)
Graft Enhancement, Immunologic/methods , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect , Hematopoietic Stem Cell Transplantation , Transplantation, Homologous/immunology , Adaptive Immunity , Animals , Antigens, Neoplasm/administration & dosage , Cell Movement , Clinical Trials as Topic , Cytokines/metabolism , Graft vs Host Disease/immunology , Graft vs Tumor Effect/drug effects , Graft vs Tumor Effect/immunology , Humans , Immunity, Innate , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Inflammation/immunology , Inflammation/prevention & control , Lymphocyte Transfusion , Mice , Models, Immunological , Organ Specificity , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/transplantation , Translational Research, Biomedical/trends , Transplantation Conditioning/adverse effects , Transplantation Conditioning/methods , Vaccination
10.
Biol Blood Marrow Transplant ; 19(1): 12-21, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22771839

ABSTRACT

Graft-versus-host disease (GVHD) remains a major cause of morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). Measures developed that have significantly reduced GVHD were also frequently associated with an increased risk of relapse. GVHD and graft-versus-tumor (GVT) effects are tightly linked, and balance between both reactions is difficult to achieve. To have an impact on the outcome and quality of life after HSCT, improvements in current strategies to prevent and treat GVHD while preserving the GVT effect are clearly needed. Sirolimus (rapamycin) is a lipophilic macrocytic lactone with immunosuppressive, antitumor, and antiviral properties. Because of its multiple modes of activities, it is being increasingly used in the management of GVHD. This review aims to summarize its mechanisms of action and potential advantages over other immunosuppressors and to analyze the most relevant studies investigating its role in both prevention and treatment of GVHD.


Subject(s)
Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Immunosuppressive Agents/therapeutic use , Sirolimus/therapeutic use , Female , Graft vs Host Disease/mortality , Graft vs Host Disease/pathology , Graft vs Tumor Effect/drug effects , Humans , Male , Recurrence , Transplantation, Homologous
11.
Exp Hematol ; 41(2): 134-142.e3, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23085463

ABSTRACT

Lenalidomide may prevent relapses after allogeneic stem cell transplantation by promoting the immune-mediated graft-versus-tumor effect. We performed a prospective phase I/II study to define the dose-limiting toxicity and the immunologic effects of lenalidomide given early (day 100-180) after allograft for four cycles in patients with multiple myeloma. According to the Fibonacci design, 24 patients with a median age of 53 years were included. Dose-limiting toxicity was organ toxicity owing to graft-versus-host disease, and the maximum tolerable dose was 5 mg. The incidence of graft-versus-host disease after lenalidomide was 38%, occurring after a median of 22 days, and was beside organ toxicity, a leading cause to discontinue the study in 29% of the patients. Immune monitoring revealed a significant increase in peripheral γ-interferon-secreting CD4(+) and CD8(+) T cells within the first week of lenalidomide treatment followed by a delayed increase in T regulatory cells. Furthermore, natural killer (NK) cells isolated from the peripheral blood of patients evidenced a significantly improved antimyeloma activity after lenalidomide treatment. The immune effect might have contributed to the increased CR rate from 24-42% after lenalidomide treatment because nonresponding patients showed significantly less natural killer and T cell activation. (Study registered under: NCT 00778752.).


Subject(s)
Graft vs Host Disease/etiology , Graft vs Tumor Effect/drug effects , Immunologic Factors/therapeutic use , Killer Cells, Natural/drug effects , Multiple Myeloma/surgery , Peripheral Blood Stem Cell Transplantation , T-Lymphocyte Subsets/drug effects , Thalidomide/analogs & derivatives , Adult , Aged , Combined Modality Therapy , Disease Progression , Dose-Response Relationship, Immunologic , Female , Graft vs Host Disease/immunology , Humans , Immunologic Factors/administration & dosage , Immunologic Factors/adverse effects , Immunologic Factors/pharmacology , Lenalidomide , Lymphocyte Activation/drug effects , Lymphocyte Count , Maintenance Chemotherapy , Male , Middle Aged , Multiple Myeloma/drug therapy , Multiple Myeloma/immunology , Prospective Studies , Remission Induction , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Thalidomide/administration & dosage , Thalidomide/adverse effects , Thalidomide/pharmacology , Thalidomide/therapeutic use , Transplantation Conditioning , Transplantation, Homologous
12.
J Immunol ; 189(10): 4719-27, 2012 Nov 15.
Article in English | MEDLINE | ID: mdl-23045613

ABSTRACT

Allogeneic hematopoietic cell transplantation is an established treatment for hematologic and nonhematologic malignancies. Donor-derived immune cells can identify and attack host tumor cells, producing a graft-versus-tumor (GVT) effect that is crucial to the effectiveness of the transplantation therapy. CBLB502 is a novel agonist for TLR5 derived from Salmonella flagellin. On the basis of TLR5-mediated immunomodulatory function, we examined the effect of CBLB502 on GVT activity. Using two tumor models that do not express TLR5, and thereby do not directly respond to CBLB502, we found that CBLB502 treatment significantly enhanced allogeneic CD8(+) T cell-mediated GVT activity, which was evidenced by decreased tumor burden and improved host survival. Importantly, histopathologic analyses showed that CBLB502 treatment did not exacerbate the moderate graft-versus-host disease condition caused by the allogeneic CD8(+) T cells. Moreover, mechanistic analyses showed that CBLB502 stimulates CD8(+) T cell proliferation and enhances their tumor killing activity mainly indirectly through a mechanism that involves the IL-12 signaling pathway and the CD11c(+) and CD11b(+) populations in the bone marrow cells. This study demonstrates a new beneficial effect of CBLB502, and suggests that TLR5-mediated immune modulation may be a promising approach to improve GVT immunity without exacerbating graft-versus-host disease.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Flagellin/pharmacology , Graft vs Tumor Effect/drug effects , Hematopoietic Stem Cell Transplantation , Immunity, Cellular/drug effects , Neoplasms, Experimental/therapy , Salmonella/chemistry , Toll-Like Receptor 5/agonists , Animals , CD8-Positive T-Lymphocytes/pathology , Cell Proliferation/drug effects , Flagellin/chemistry , Flagellin/immunology , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Tumor Effect/immunology , Immunity, Cellular/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Toll-Like Receptor 5/immunology , Transplantation, Homologous
13.
Biol Blood Marrow Transplant ; 18(9): 1329-40, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22326303

ABSTRACT

Transforming growth factor (TGF)-ß is a pleiotropic cytokine with widespread and profound effects on immune cells. Consequently, it has generated considerable interest in relation to the immunologic outcomes after allogeneic hematopoietic cell transplantation. The TGF-ß pathway has been shown to be an important modulator of alloimmunity, with direct consequences on graft-versus-host disease pathophysiology and graft-versus-tumor response. The TGF-ß-related effects can be both beneficial and detrimental to the host, underscoring the complexity of TGF-ß biology. This article reviews the evidence linking TGF-ß to alloimmune responses in allogeneic hematopoietic cell transplantation and highlights foreseeable strategies that would maximize the beneficial effects of TGF-ß pathway modulation on both graft-versus-host disease pathophysiology and the graft-versus-tumor effect.


Subject(s)
Graft vs Host Disease/immunology , Graft vs Tumor Effect/immunology , Hematopoietic Stem Cell Transplantation , Lymphocytes/immunology , Transforming Growth Factor beta/immunology , Animals , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/drug effects , Humans , Immune Tolerance/drug effects , Immunomodulation/drug effects , Lymphocytes/drug effects , Mice , Signal Transduction/drug effects , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Transplantation, Homologous
14.
Biol Blood Marrow Transplant ; 18(1 Suppl): S125-31, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22226096

ABSTRACT

Despite improvements to hematopoietic stem cell transplantation over the past several decades, further advances are necessary to achieve: improved control of toxicities like graft-versus-host disease; enhanced immunologic reconstitution posttransplantation; and reduction in relapse risk via enhancement of graft-versus-tumor responses. Achieving these disparate hematopoietic stem cell transplantation goals will likely require the introduction of novel therapeutic agents to the current armamentarium. In this article, we outline preclinical and early-phase clinical data indicating the potential of proteasome-inhibitor therapy (bortezomib), hypomethylating agent therapy (azacytidine), and histone deacetylase-inhibitor therapy (vorinostat) to help improve hematopoietic stem cell transplantation outcomes.


Subject(s)
Antineoplastic Agents/therapeutic use , Azacitidine/therapeutic use , Boronic Acids/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Tumor Effect/drug effects , Hematopoietic Stem Cell Transplantation , Hydroxamic Acids/therapeutic use , Pyrazines/therapeutic use , Bortezomib , Graft vs Host Disease/etiology , Humans , Proteasome Inhibitors , Vorinostat
15.
Expert Rev Clin Immunol ; 7(4): 515-25; quiz 526-7, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21787195

ABSTRACT

Disease recurrence is the single most common cause of death after allogeneic or autologous hematopoietic stem cell transplantation (HSCT). Disease status and chemosensitivity at the time of transplantation, as well as the development of graft-versus-host disease (GVHD), are factors known to influence the risk of relapse post-HSCT. Both acute and chronic GVHD have been associated with decreased relapse rates; however, owing to toxicity, overall survival is not consistently improved in these patients. Furthermore, there is a transient period of immunodeficiency after HSCT, which may permit residual malignant cells to proliferate early in the post-transplant course, before the donor immune system can establish a graft-versus-tumor response. Patients who fail an initial HSCT have an extremely poor outcome; therefore, maneuvers to prevent, identify and treat recurrent disease as early as possible in these situations are necessary. Strategies to distinguish graft-versus-tumor from GVHD, to enhance both general and disease-specific immune reconstitution after transplantation, and to increase donor-mediated anti-host immune reactions are being investigated in clinical trials. Single agent nontoxic post-HSCT chemotherapy, cellular therapies and second allogeneic HSCT using reduced intensity regimens are among the modalities under investigation.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation , Neoplasm Recurrence, Local/therapy , Animals , Clinical Trials as Topic , Graft vs Tumor Effect/drug effects , Graft vs Tumor Effect/immunology , Hematologic Neoplasms/pathology , Hematologic Neoplasms/physiopathology , Humans , Immunomodulation , Reoperation , Transplantation, Homologous , Treatment Failure
16.
Biol Blood Marrow Transplant ; 17(5): 682-92, 2011 May.
Article in English | MEDLINE | ID: mdl-20713164

ABSTRACT

Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) is a key negative regulator of T cell activation and proliferation. Ipilimumab is a human monoclonal antibody that specifically blocks the binding of CTLA-4 to its ligand. To test the hypothesis that blockade of CTLA-4 by ipilimumab could augment graft-versus-malignancy (GVM) effects without a significant impact on graft-versus-host disease (GVHD), we conducted a phase I clinical trial of ipilimumab infusion in patients with relapsed malignancy following allogeneic hematopoietic stem cell transplantation (allo-HSCT). Here, we report the analysis of peripheral blood T lymphocyte reconstitution, T regulatory cell (Treg) expression, and T cell activation markers after a single dose of ipilimumab in 29 patients. Peripheral blood samples were collected from all patients before and after ipilimumab infusion. Lymphocyte immunophenotyes, including levels of CD4(+)CD25(high) cells and T cell activation markers, were analyzed in all cases. Levels of CD4(+)CD25(high)Foxp3(+) cells and intracellular CTLA-4 in CD4(+) T cells also were evaluated in the last 11 cases. We found lower baseline levels of CD4(+) and CD45RO(+) T cells in patients compared with normal controls. More than 50% of the patients had abnormally low lymphocyte counts (CD4 or/and CD8 T cells), and some had no circulating B lymphocytes. The percentages of both CD4(+)CD25(high) and CD4(+)CD25(high)Foxp3(+) T cells were significantly higher in patients before ipilimumab infusion than in healthy donors. Twenty of 29 patients exhibited an elevated level of CD4(+)CD25(low) activated T cells at baseline, compared with only 3 of 26 healthy donors. Both CD4(+) and CD8(+) T lymphocyte counts were significantly increased after ipilimumab infusion. There was no consistent change in absolute lymphocyte count or in the number of T cells expressing the activation marker CD69. However, increases in CD4(+)CD25(low) T cells were seen in 20 of 29 patients and increases in CD4(+)HLA-DR(+) T cells were seen in the last 10 patients in the first 60 days after ipilimumab infusion. Although the percentages of both CD4(+)CD25(high) and CD4(+)CD25(high)Foxp3(+) T cells decreased significantly during the observation period, the absolute cell counts did not change. Intracellular CTLA-4 expression in CD4(+)CD25(lo/-) T cells increased significantly after ipilimumab infusion. We conclude that CTLA-4 blockade by a single infusion of ipilimumab increased CD4(+) and CD4(+)HLA-DR(+) T lymphocyte counts and intracellular CTLA-4 expression at the highest dose level. There was no significant change in Treg cell numbers after ipilimumab infusion. These data demonstrate that significant changes in T cell populations occur on exposure to a single dose of ipilimumab. Further studies with multiple doses are needed to explore this phenomenon further and to correlate changes in lymphocyte subpopulations with clinical events.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, CD , Breast Neoplasms/immunology , Graft vs Tumor Effect , Leukemia/immunology , Lymphoproliferative Disorders/immunology , T-Lymphocytes, Regulatory/immunology , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/therapeutic use , Antigens, CD/analysis , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/analysis , Breast Neoplasms/physiopathology , Breast Neoplasms/prevention & control , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Case-Control Studies , Cell Count , Female , Flow Cytometry , Graft vs Host Disease/immunology , Graft vs Tumor Effect/drug effects , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Injections, Intravenous , Interleukin-2 Receptor alpha Subunit/analysis , Ipilimumab , Lectins, C-Type/analysis , Leukemia/physiopathology , Leukemia/prevention & control , Leukemia/therapy , Leukocyte Common Antigens/analysis , Lymphocyte Activation/immunology , Lymphoproliferative Disorders/physiopathology , Lymphoproliferative Disorders/prevention & control , Lymphoproliferative Disorders/therapy , Male , Recurrence , Transplantation, Homologous
17.
Expert Rev Hematol ; 3(3): 301-14, 2010 Jun.
Article in English | MEDLINE | ID: mdl-21082981

ABSTRACT

This article focuses on the recent evolution of novel conditioning regimens in combination with adoptive cellular therapy in the allogeneic transplant setting for hematologic malignancies. Building on data from animal models, the field of allogeneic transplantation is undergoing a paradigm shift toward immunosuppressive regimens with less toxicity that allow donor hematopoietic engraftment in order to provide a graft-versus-tumor effect as the primary goal of transplantation, rather than chemoablation. In addition, the strategies described in this article, including the use of T-cell subsets as adoptive therapy, will apply to a much broader pool of patients than traditional transplant approaches, thereby allowing more patients with life-limiting illnesses, previously deemed ineligible, to pursue therapy with curative intent.


Subject(s)
Ablation Techniques/methods , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/drug effects , Graft vs Tumor Effect/immunology , Hematologic Neoplasms/therapy , Hematopoietic Stem Cell Transplantation/methods , Immunosuppressive Agents/pharmacology , Immunotherapy, Adoptive , Transplantation Conditioning , Animals , Clinical Trials as Topic , Hematologic Neoplasms/immunology , Humans , Immunotherapy, Adoptive/methods , Immunotherapy, Adoptive/trends , Mice , Transplantation Conditioning/methods , Transplantation, Homologous
18.
Biol Blood Marrow Transplant ; 16(10): 1463-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20685260

ABSTRACT

We retrospectively analyzed outcomes among 1206 patients with hematologic malignancies who had hematopoietic cell transplantation (HCT) from HLA-identical siblings (n = 630) or HLA-matched unrelated donors (n = 576) at a single institution between 2001 and 2007 for a correlation between recipient statin use and risk of graft-versus-host disease (GVHD). Among recipients with cyclosporine-based postgrafting immunosuppression (n = 821), statin use at the time of transplant (6%) was associated with a decreased risk of extensive chronic GVHD (cGVHD) (multivariate hazard ratio [HR], 0.62; 95% confidence interval [CI], 0.4-1.0; P = .05) and an increased risk of recurrent malignancy (HR, 1.75; 95% CI, 1.0-3.0; P = .04). Recipient statin use, however, had no apparent impact on the risks of cGVHD and recurrent malignancy among recipients given tacrolimus-based immunosuppression (n = 385; 8% statin treated). Risks of acute GVHD, nonrelapse mortality, and overall mortality were not significantly affected by recipient statin use. Hence, recipient statin treatment at the time of allogeneic HCT may decrease the risk of cGVHD in patients with cyclosporine-based immunosuppression, but at the expense of a compromised graft-versus-tumor effect.


Subject(s)
Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/drug effects , Hematopoietic Stem Cell Transplantation , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , T-Lymphocytes/drug effects , Adult , Aged , Chronic Disease , Cyclosporine/therapeutic use , Drug Interactions , Female , Graft vs Host Disease/drug therapy , Graft vs Host Disease/epidemiology , Graft vs Host Disease/etiology , Hematologic Neoplasms/mortality , Hematologic Neoplasms/surgery , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Immunosuppressive Agents/therapeutic use , Incidence , Male , Middle Aged , Recurrence , Retrospective Studies , Risk , Transplantation, Homologous/adverse effects , Young Adult
19.
Biol Blood Marrow Transplant ; 16(6): 739-50, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20338256

ABSTRACT

The occurrence of acute graft-versus-host disease (aGVHD) and tumor relapse represent the two major obstacles impeding the efficacy of allogeneic bone marrow transplantation (BMT) in cancer. We have previously shown that the synthetic triterpenoid 2-cyano-3, 12-dioxooleana-1, 9-dien-28-oic acid (CDDO) can inhibit murine early aGVHD, but antitumor effects were not assessed. In the current study, we found that a new derivative of CDDO, CDDO-Me, had an increased ability to inhibit allogeneic T cell responses and induce cell death of alloreactive T cells in vitro. Administration of CDDO-Me to mice following allogeneic BMT resulted in significant and increased protection from lethal aGVHD compared to CDDO. This correlated with reduced TNF-alpha production, reduced donor T cell proliferation, and decreased adhesion molecule (alpha(4)beta(7) integrin) expression on the donor T cells. CDDO-Me was also superior to CDDO in inhibiting leukemia growth in vitro. When CDDO-Me was administered following an allogeneic BMT to leukemia-bearing mice, significant increases in survival were observed. These findings suggest that CDDO-Me is superior to CDDO in delaying aGVHD, while preserving or possibly even augmenting GVT effects.


Subject(s)
Bone Marrow Transplantation/methods , Graft vs Host Disease/prevention & control , Graft vs Tumor Effect/immunology , Oleanolic Acid/analogs & derivatives , Animals , Apoptosis/drug effects , Apoptosis/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Colon/pathology , Female , Graft vs Host Disease/blood , Graft vs Host Disease/pathology , Graft vs Tumor Effect/drug effects , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Integrins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/therapy , Liver/pathology , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oleanolic Acid/administration & dosage , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Spleen/cytology , Spleen/immunology , Survival Analysis , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Transplantation, Homologous , Tumor Necrosis Factor-alpha/blood
20.
J Immunol ; 182(9): 5846-54, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19380833

ABSTRACT

Posttransplant immunodeficiency, specifically a lack of T cell reconstitution, is a major complication of allogeneic bone marrow transplantation. This immunosuppression results in an increase in morbidity and mortality from infections and very likely contributes to relapse. In this study, we demonstrate that sex steroid ablation using leuprolide acetate, a luteinizing hormone-releasing hormone agonist (LHRHa), increases the number of lymphoid and myeloid progenitor cells in the bone marrow and developing thymocytes in the thymus. Although few differences are observed in the peripheral myeloid compartments, the enhanced thymic reconstitution following LHRHa treatment and allogeneic bone marrow transplantation leads to enhanced peripheral T cell recovery, predominantly in the naive T cell compartment. This results in an increase in T cell function in vivo and in vitro. Graft-versus-host-disease is not exacerbated by LHRHa treatment and graft-versus-tumor activity is maintained. Because LHRHa allows for reversible (and temporary) sex steroid ablation, has a strong safety profile, and has been clinically approved for diseases such as prostate and breast cancer, this drug treatment represents a novel therapeutic approach to reversal of thymic atrophy and enhancement of immunity following immunosuppression.


Subject(s)
Bone Marrow Transplantation/immunology , Gonadotropin-Releasing Hormone/administration & dosage , T-Lymphocytes/drug effects , T-Lymphocytes/transplantation , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Transplantation/pathology , Cell Differentiation/drug effects , Cell Differentiation/immunology , Female , Gonadotropin-Releasing Hormone/agonists , Graft vs Host Disease/immunology , Graft vs Host Disease/pathology , Graft vs Host Disease/therapy , Graft vs Tumor Effect/drug effects , Graft vs Tumor Effect/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/pathology , Humans , Isoantigens/administration & dosage , Isoantigens/genetics , Leuprolide/administration & dosage , Lymphopenia/immunology , Lymphopenia/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Spleen/cytology , Spleen/drug effects , Spleen/immunology , T-Lymphocytes/pathology , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...