Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Sci Adv ; 5(7): eaaw5096, 2019 07.
Article in English | MEDLINE | ID: mdl-31281894

ABSTRACT

Chimeric antigen receptor (CAR) T cell therapy for hematologic malignancies is fraught with several unknowns, including number of functional T cells that engage target tumor, durability and subsequent expansion and contraction of that engagement, and whether toxicity can be managed. Non-invasive, serial imaging of CAR T cell therapy using a reporter transgene can address those issues quantitatively. We have transduced anti-CD19 CAR T cells with the prostate-specific membrane antigen (PSMA) because it is a human protein with restricted normal tissue expression and has an expanding array of positron emission tomography (PET) and therapeutic radioligands. We demonstrate that CD19-tPSMA(N9del) CAR T cells can be tracked with [18F]DCFPyL PET in a Nalm6 model of acute lymphoblastic leukemia. Divergence between the number of CD19-tPSMA(N9del) CAR T cells in peripheral blood and bone marrow and those in tumor was evident. These findings underscore the need for non-invasive repeatable monitoring of CAR T cell disposition clinically.


Subject(s)
Antigens, Surface/metabolism , Glutamate Carboxypeptidase II/metabolism , Immunotherapy, Adoptive , Positron-Emission Tomography/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/diagnostic imaging , Animals , Antigens, CD19/metabolism , Antigens, Surface/genetics , Glutamate Carboxypeptidase II/genetics , Humans , Leukemia, Experimental/diagnostic imaging , Leukemia, Experimental/pathology , Lysine/analogs & derivatives , Mice, Inbred NOD , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/physiology , Urea/analogs & derivatives
2.
J Exp Med ; 215(3): 859-876, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29436396

ABSTRACT

Depletion of immunosuppressive tumor-associated macrophages (TAMs) or reprogramming toward a proinflammatory activation state represent different strategies to therapeutically target this abundant myeloid population. In this study, we report that inhibition of colony-stimulating factor-1 receptor (CSF-1R) signaling sensitizes TAMs to profound and rapid reprogramming in the presence of a CD40 agonist before their depletion. Despite the short-lived nature of macrophage hyperactivation, combined CSF-1R+CD40 stimulation of macrophages is sufficient to create a proinflammatory tumor milieu that reinvigorates an effective T cell response in transplanted tumors that are either responsive or insensitive to immune checkpoint blockade. The central role of macrophages in regulating preexisting immunity is substantiated by depletion experiments, transcriptome analysis of ex vivo sorted TAMs, and gene expression profiling of whole tumor lysates at an early treatment time point. This approach enabled the identification of specific combination-induced changes among the pleiotropic activation spectrum of the CD40 agonist. In patients, CD40 expression on human TAMs was detected in mesothelioma and colorectal adenocarcinoma.


Subject(s)
Immunity , Macrophages/immunology , Neoplasms/immunology , Neoplasms/pathology , Animals , CD40 Antigens/agonists , CD40 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , Female , Humans , Inflammation/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Biological , Phenotype , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/metabolism
3.
Cancer Res ; 75(1): 51-61, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25388285

ABSTRACT

Traditionally, cell-mediated immune responses to vaccination in animal models are evaluated by invasive techniques such as biopsy and organ extraction. We show here that by combining two noninvasive imaging technologies, MRI and bioluminescence imaging (BLI), we can visualize both the afferent and efferent arms of cellular events following vaccination longitudinally. To this end, we evaluated the immune response elicited by a novel Toll-like receptor 4 agonist vaccine adjuvant, glucopyranosyl lipid A (GLA), using a whole-cell tumor vaccine. After magnetovaccination, MRI was used to visualize antigen-presenting cell-mediated antigen capture and subsequent migration to draining lymph nodes (DLN). Paradoxically, we observed that the incorporation of GLA in the vaccine reduced these critical parameters of the afferent immune response. For the efferent arm, the magnitude of the ensuing antigen-specific T-cell response in DLN visualized using BLI correlated with antigen delivery to the DLN as measured by MRI. These findings were confirmed using flow cytometry. In spite of the GLA-associated reduction in antigen delivery to the DLN, however, the use of GLA as a vaccine adjuvant led to a massive proliferation of vaccine primed antigen-specific T cells in the spleen. This was accompanied by an enhanced tumor therapeutic effect of the vaccine. These findings suggest that GLA adjuvant changes the temporal and anatomical features of both the afferent and efferent arms of the vaccine response and illustrates the utility of quantitative noninvasive imaging as a tool for evaluating these parameters during vaccine optimization.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Lymph Nodes/immunology , Melanoma, Experimental/therapy , Toll-Like Receptor 4/agonists , Toll-Like Receptor 4/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/immunology , Female , Humans , Luminescent Measurements/methods , Magnetic Resonance Imaging/methods , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Multimodal Imaging/methods
4.
Cancer Cell ; 25(6): 846-59, 2014 Jun 16.
Article in English | MEDLINE | ID: mdl-24898549

ABSTRACT

Macrophage infiltration has been identified as an independent poor prognostic factor in several cancer types. The major survival factor for these macrophages is macrophage colony-stimulating factor 1 (CSF-1). We generated a monoclonal antibody (RG7155) that inhibits CSF-1 receptor (CSF-1R) activation. In vitro RG7155 treatment results in cell death of CSF-1-differentiated macrophages. In animal models, CSF-1R inhibition strongly reduces F4/80(+) tumor-associated macrophages accompanied by an increase of the CD8(+)/CD4(+) T cell ratio. Administration of RG7155 to patients led to striking reductions of CSF-1R(+)CD163(+) macrophages in tumor tissues, which translated into clinical objective responses in diffuse-type giant cell tumor (Dt-GCT) patients.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Colonic Neoplasms/therapy , Macrophages/drug effects , Macrophages/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/immunology , Animals , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Monoclonal, Humanized , Cell Differentiation/physiology , Cell Line, Tumor , Clinical Trials, Phase I as Topic , Cohort Studies , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Female , Humans , Macaca fascicularis , Macrophages/cytology , Macrophages/metabolism , Male , Mice, Inbred C57BL , Models, Molecular , Receptor, Macrophage Colony-Stimulating Factor/metabolism
5.
BMC Genomics ; 14: 869, 2013 Dec 10.
Article in English | MEDLINE | ID: mdl-24325565

ABSTRACT

BACKGROUND: Although transposable element (TE) derived DNA accounts for more than half of mammalian genomes and initiates a significant proportion of RNA transcripts, high throughput methods are rarely leveraged specifically to detect expression from interspersed repeats. RESULTS: To characterize the contribution of transposons to mammalian transcriptomes, we developed a custom microarray platform with probes covering known human and mouse transposons in both sense and antisense orientations. We termed this platform the "TE-array" and profiled TE repeat expression in a panel of normal mouse tissues. Validation with nanoString® and RNAseq technologies demonstrated that TE-array is an effective method. Our data show that TE transcription occurs preferentially from the sense strand and is regulated in highly tissue-specific patterns. CONCLUSIONS: Our results are consistent with the hypothesis that transposon RNAs frequently originate within genomic TE units and do not primarily accumulate as a consequence of random 'read-through' from gene promoters. Moreover, we find TE expression is highly dependent on the tissue context. This suggests that TE expression may be related to tissue-specific chromatin states or cellular phenotypes. We anticipate that TE-array will provide a scalable method to characterize transposable element RNAs.


Subject(s)
DNA Transposable Elements , Genomics/methods , Oligonucleotide Array Sequence Analysis/methods , Transcription, Genetic , Animals , Cell Line , Female , Gene Expression , Gene Expression Profiling/methods , Gene Expression Regulation , Humans , Male , Mice , Oligonucleotide Array Sequence Analysis/standards , Organ Specificity/genetics , Reproducibility of Results , Transfection
6.
J Clin Invest ; 123(6): 2447-63, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23728179

ABSTRACT

Activation of TLR9 by direct injection of unmethylated CpG nucleotides into a tumor can induce a therapeutic immune response; however, Tregs eventually inhibit the antitumor immune response and thereby limit the power of cancer immunotherapies. In tumor-bearing mice, we found that Tregs within the tumor preferentially express the cell surface markers CTLA-4 and OX40. We show that intratumoral coinjection of anti-CTLA-4 and anti-OX40 together with CpG depleted tumor-infiltrating Tregs. This in situ immunomodulation, which was performed with low doses of antibodies in a single tumor, generated a systemic antitumor immune response that eradicated disseminated disease in mice. Further, this treatment modality was effective against established CNS lymphoma with leptomeningeal metastases, sites that are usually considered to be tumor cell sanctuaries in the context of conventional systemic therapy. These results demonstrate that antitumor immune effectors elicited by local immunomodulation can eradicate tumor cells at distant sites. We propose that, rather than using mAbs to target cancer cells systemically, mAbs could be used to target the tumor infiltrative immune cells locally, thereby eliciting a systemic immune response.


Subject(s)
Brain Neoplasms/therapy , Lymphoma/therapy , Meningeal Neoplasms/therapy , T-Lymphocytes, Regulatory/immunology , Adjuvants, Immunologic/administration & dosage , Adjuvants, Immunologic/pharmacology , Animals , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Combined Modality Therapy , Female , Humans , Immunologic Factors/administration & dosage , Immunomodulation , Immunotherapy , Injections, Intralesional , Lymphocyte Depletion , Lymphoma/immunology , Lymphoma/pathology , Meningeal Neoplasms/immunology , Meningeal Neoplasms/secondary , Mice , Mice, Inbred BALB C , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Oligodeoxyribonucleotides/administration & dosage , Oligodeoxyribonucleotides/pharmacology , Receptors, OX40/immunology , T-Lymphocytes, Regulatory/metabolism , Toll-Like Receptor 9/agonists , Tumor Burden
7.
Clin Dev Immunol ; 2012: 124187, 2012.
Article in English | MEDLINE | ID: mdl-22778760

ABSTRACT

The past decade has witnessed the evolvement of cancer immunotherapy as an increasingly effective therapeutic modality, evidenced by the approval of two immune-based products by the FDA, that is, the cancer vaccine Provenge (sipuleucel-T) for prostate cancer and the antagonist antibody against cytotoxic T-lymphocyte antigen-4 (CTLA-4) ipilimumab for advanced melanoma. In addition, the clinical evaluations of a variety of promising immunotherapy drugs are well under way. Benefiting from more efficacious immunotherapeutic agents and treatment strategies, a number of recent clinical studies have achieved unprecedented therapeutic outcomes in some patients with certain types of cancers. Despite these advances, however, the efficacy of most cancer immunotherapies currently under clinical development has been modest. A recurring scenario is that therapeutic maneuvers initially led to measurable antitumor immune responses in cancer patients but ultimately failed to improve patient outcomes. It is increasingly recognized that tumor cells can antagonize therapy-induced immune attacks through a variety of counterregulation mechanisms, which represent a fundamental barrier to the success of cancer immunotherapy. Herein we summarize the findings from some recent preclinical and clinical studies, focusing on how tumor cells advance their survival and expansion by hijacking therapy-induced immune effector mechanisms that would otherwise mediate their destruction.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , Tumor Escape/immunology , Combined Modality Therapy , Humans , Immunomodulation
8.
Cancer J ; 17(5): 309-24, 2011.
Article in English | MEDLINE | ID: mdl-21952281

ABSTRACT

Hematologic malignancies were the first diseases in clinical oncology for which the potential of harnessing the immune system as targeted therapy was unequivocally demonstrated. Unfortunately, the use of this highly efficacious modality has been limited to only a subset of patients and diseases because of immune-mediated toxicities resulting from incomplete specificity, and disease-specific determinants of sensitivity versus resistance to immune effector mechanisms. Recent studies, however, have begun to elucidate the molecular basis of the observed clinical effects allowing the rational development of next generation of immunotherapeutic combinations. We discuss here cancer antigen targets in hematologic malignancies and the specific approaches to induce immunity being pursued, the importance of modulating the host immunoregulatory environment, and the special features of immunological monitoring in clinical investigation. The hematologic malignancies represent an ideal setting for the development of immunotherapy due to logistical, clinical monitoring, and disease biology factors and may represent an exemplar for immune-based treatment in other cancer types.


Subject(s)
Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Immunotherapy/methods , Antigens, Neoplasm/immunology , Biomarkers, Tumor/immunology , Cancer Vaccines/immunology , Humans , Immunologic Factors/therapeutic use
9.
Leuk Lymphoma ; 52(11): 2076-81, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21756035

ABSTRACT

Relapse after autologous stem cell transplant for low grade B-cell lymphoma is common secondary to ineffective conditioning and/or tumor autograft contamination. We investigated high-dose cyclophosphamide and rituximab without stem cell rescue as first-line or salvage therapy in lymphomas. After establishing safety, accrual was increased to evaluate event-free survival (EFS). Eighty-one adults received rituximab (375 mg/m(2) days 1, 4, 8, 11, 45, 52), cyclophosphamide (50 mg/kg days 15-18), and pegfilgrastim (day 20). Forty-two patients had low grade B-cell lymphoma [grade I/II follicular (69%), transformed lymphoma (17%), other (15%)]: 45% were treated without measurable disease. Thirty-nine patients had mantle cell lymphoma: 82% were treated without measurable disease. All achieved hematopoietic recovery; 46% required brief hospitalizations. The 5-year EFS and overall survival (OS) for patients with low grade B-cell and transformed lymphoma were 40% and 72%, respectively. The 5-year EFS and OS for patients with MCL were 39% and 62%, respectively. This low-toxicity therapeutic approach obviates the need for stem cell products and establishes a platform for future therapies.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, B-Cell/drug therapy , Lymphoma, Mantle-Cell/drug therapy , Adult , Aged , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Antibodies, Monoclonal, Murine-Derived/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cyclophosphamide/administration & dosage , Cyclophosphamide/adverse effects , Disease-Free Survival , Dose-Response Relationship, Drug , Drug Administration Schedule , Feasibility Studies , Fever/chemically induced , Hospitalization/statistics & numerical data , Humans , Lymphoma, B-Cell/pathology , Lymphoma, Mantle-Cell/pathology , Middle Aged , Neoplasm Grading , Neutropenia/chemically induced , Rituximab , Treatment Outcome , Young Adult
10.
J Immunol ; 186(4): 2148-55, 2011 Feb 15.
Article in English | MEDLINE | ID: mdl-21242518

ABSTRACT

Activated T cells can acquire membrane molecules from APCs through a process termed trogocytosis. The functional consequence of this event has been a subject of debate. Focusing on transfer of peptide-MHC class II (MHC-II) complexes from APCs to CD4(+) T cells after activation, in this study we investigated the molecule acquisition potential of naturally occurring regulatory T cells (Tregs) and CD4(+) Th cells. We show that acquisition of membrane molecules from APCs is an inherent feature of CD4(+) T cell activation. Triggering of the TCR enables CD4(+) T cells to acquire their agonist ligands as well as other irrelevant membrane molecules from the interacting APCs or bystander cells in a contact-dependent manner. Notably, trogocytosis is a continuous process during cell cycle progression, and Th cells and Tregs have comparable capacity for trogocytosis both in vitro and in vivo. The captured peptide-MHC-II molecules, residing in sequestered foci on the host cell surface, endow the host cells with Ag-presenting capability. Presentation of acquired peptide-MHC-II ligands by Th cells or Tregs has either stimulatory or regulatory effect on naive CD4(+) T cells, respectively. Furthermore, Th cells with captured peptide-MHC-II molecules become effector cells that manifest better recall responses, and Tregs with captured ligands exhibit enhanced suppression activity. These findings implicate trogocytosis in different subsets of CD4(+) T cells as an intrinsic mechanism for the fine tuning of Ag-specific CD4(+) T cell response.


Subject(s)
Antigen Presentation/immunology , CD4 Antigens/physiology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation/immunology , Peptides/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , CD4 Antigens/biosynthesis , Cell Line, Tumor , Chickens , Histocompatibility Antigens Class II/immunology , Immunologic Memory , Ligands , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Peptides/immunology , Protein Interaction Mapping , Sarcoma, Experimental/immunology , Sarcoma, Experimental/metabolism , Sarcoma, Experimental/pathology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
11.
Blood ; 117(2): 608-17, 2011 Jan 13.
Article in English | MEDLINE | ID: mdl-20935254

ABSTRACT

Few published studies characterize early lymphocyte recovery after intensive chemotherapy for acute myelogenous leukemia (AML). To test the hypothesis that lymphocyte recovery mirrors ontogeny, we characterized early lymphocyte recovery in 20 consecutive patients undergoing induction timed sequential chemotherapy for newly diagnosed AML. Recovering T lymphocytes were predominantly CD4(+) and included a greatly expanded population of CD3(+)CD4(+)CD25(+)Foxp3(+) T cells. Recovering CD3(+)CD4(+)CD25(+)Foxp3(+) T cells were phenotypically activated regulatory T cells and showed suppressive activity on cytokine production in a mixed lymphocyte reaction. Despite an initial burst of thymopoiesis, most recovering regulatory T cells were peripherally derived. Furthermore, regulatory T cells showed marked oligoclonal skewing, suggesting that their peripheral expansion was antigen-driven. Overall, lymphocyte recovery after chemotherapy differs from ontogeny, specifically identifying a peripherally expanded oligoclonal population of activated regulatory T lymphocytes. These differences suggest a stereotyped immunologic recovery shared by patients with newly diagnosed AML after induction timed sequential chemotherapy. Further insight into this oligoclonal regulatory T-cell population will be fundamental toward developing effective immunomodulatory techniques to improve survival for patients with AML.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Regulatory/cytology , Adult , Aged , Cell Separation , Cytarabine/administration & dosage , Cytosine/administration & dosage , Daunorubicin/administration & dosage , Etoposide/administration & dosage , Female , Flavonoids/administration & dosage , Flow Cytometry , Gene Expression Profiling , Humans , Immunophenotyping , Male , Middle Aged , Mitoxantrone/administration & dosage , Piperidines/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Vidarabine/analogs & derivatives , Young Adult
12.
Am J Hematol ; 86(1): 31-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21132730

ABSTRACT

Although tyrosine kinase inhibitors have redefined the care of chronic myeloid leukemia (CML), these agents have not proved curative, likely due to resistance of the leukemia stem cells (LSC). While a number of potential therapeutic targets have emerged in CML, their expression in the LSC remains largely unknown. We therefore isolated subsets of CD34(+) stem/progenitor cells from normal donors and from patients with chronic phase or blast crisis CML. These cell subsets were then characterized based on ability to engraft immunodeficient mice and expression of candidate therapeutic targets. The CD34(+)CD38(-) CML cell population with high aldehyde dehydrogenase (ALDH) activity was the most enriched for immunodeficient mouse engrafting capacity. The putative targets: PROTEINASE 3, SURVIVIN, and hTERT were expressed only at relatively low levels by the CD34(+)CD38(-)ALDH(high) CML cells, similar to the normal CD34(+)CD38(-)ALDH(high) cells and less than in the total CML CD34(+) cells. In fact, the highest expression of these antigens was in normal, unfractionated CD34(+) cells. In contrast, PRAME and WT1 were more highly expressed by all CML CD34(+) subsets than their normal counterparts. Thus, ALDH activity appears to enrich for CML stem cells, which display an expression profile that is distinct from normal stem/progenitor cells and even the CML progenitors. Indeed, expression of a putative target by the total CD34(+) population in CML does not guarantee expression by the LSC. These expression patterns suggest that PROTEINASE 3, SURVIVIN, and hTERT are not optimal therapeutic targets in CML stem cells; whereas PRAME and WT1 seem promising.


Subject(s)
Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , ADP-ribosyl Cyclase 1/biosynthesis , Adult , Aged , Aldehyde Dehydrogenase/biosynthesis , Animals , Antigens, CD34/biosynthesis , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/genetics , Female , Hematopoietic Stem Cells/immunology , Humans , Inhibitor of Apoptosis Proteins , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Microtubule-Associated Proteins/biosynthesis , Microtubule-Associated Proteins/genetics , Middle Aged , Myeloblastin/biosynthesis , Myeloblastin/genetics , Neoplasm Transplantation , RNA, Messenger/biosynthesis , Survivin , Telomerase/biosynthesis , Telomerase/genetics , WT1 Proteins/biosynthesis , WT1 Proteins/genetics
13.
Blood ; 115(16): 3224-30, 2010 Apr 22.
Article in English | MEDLINE | ID: mdl-20124511

ABSTRACT

Because of its potent immunosuppressive yet stem cell-sparing activity, high-dose cyclophosphamide was tested as sole prophylaxis of graft-versus-host disease (GVHD) after myeloablative allogeneic bone marrow transplantation (alloBMT). We treated 117 patients (median age, 50 years; range, 21-66 years) with advanced hematologic malignancies; 78 had human leukocyte antigen (HLA)-matched related donors and 39 had HLA-matched unrelated donors. All patients received conventional myeloablation with busulfan/cyclophosphamide (BuCy) and T cell-replete bone marrow followed by 50 mg/kg/d of cyclophosphamide on days 3 and 4 after transplantation. The incidences of acute grades II through IV and grades III through IV GVHD for all patients were 43% and 10%, respectively. The nonrelapse mortality at day 100 and 2 years after transplantation were 9% and 17%, respectively. The actuarial overall survival and event-free survivals at 2 years after transplantation were 55% and 39%, respectively, for all patients and 63% and 54%, respectively, for patients who underwent transplantation while in remission. With a median follow-up of 26.3 months among surviving patients, the cumulative incidence of chronic GVHD is 10%. These results suggest that high-dose posttransplantation cyclophosphamide is an effective single-agent prophylaxis of acute and chronic GVHD after BuCy conditioning and HLA-matched BMT (clinicaltrials.gov no. NCT00134017).


Subject(s)
Bone Marrow Transplantation/adverse effects , Cyclophosphamide/administration & dosage , Graft vs Host Disease/prevention & control , Hematologic Neoplasms/therapy , Immunosuppressive Agents/administration & dosage , Adult , Aged , Bone Marrow Transplantation/mortality , Disease-Free Survival , Dose-Response Relationship, Drug , Female , Hematologic Neoplasms/mortality , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Transplantation Conditioning/methods , Transplantation, Homologous , Treatment Outcome , Young Adult
14.
Clin Cancer Res ; 16(1): 338-47, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20048335

ABSTRACT

PURPOSE: Chronic myeloid leukemia (CML) can be responsive to T-cell-mediated immunity. K562/granulocyte macrophage-colony stimulating factor (GM-CSF) is a GM-CSF producing vaccine derived from a CML cell line that expresses several CML-associated antigens. A pilot study was developed to determine if K562/GM-CSF immunotherapy could improve clinical responses to imatinib mesylate (IM) in patients with chronic myeloid leukemia. EXPERIMENTAL DESIGN: Patients with chronic phase CML who achieved at least a major cytogeneic response but remained with persistent, measurable disease despite one or more years on imatinib mesylate were eligible. Each was given a series of four vaccines administered in three-week intervals, with or without topical imiquimod, while remaining on a stable dose of imatinib mesylate. CML disease burden was measured serially before and after vaccination. RESULTS: Nineteen patients were vaccinated, with a median duration of previous imatinib mesylate therapy of 37 (13-53) months. Mean PCR measurements of BCR-ABL for the group declined significantly following the vaccines (P = 0.03). Thirteen patients had a progressive decline in disease burden, 8 of whom had increasing disease burden before vaccination. Twelve patients achieved their lowest tumor burden measurements to date following vaccine, including seven subjects who became PCR-undetectable. CONCLUSIONS: K562/GM-CSF vaccine appears to improve molecular responses in patients on imatinib mesylate, including achieving complete molecular remissions, despite long durations of previous imatinib mesylate therapy.


Subject(s)
Cancer Vaccines/therapeutic use , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/therapy , Neoplasm, Residual/therapy , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Adult , Aged , Aminoquinolines/administration & dosage , Benzamides , Female , Fusion Proteins, bcr-abl/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Imatinib Mesylate , Imiquimod , Immunotherapy , K562 Cells , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Leukemia, Myeloid, Chronic-Phase , Male , Middle Aged , Neoplasm, Residual/drug therapy , Pilot Projects , Tumor Burden
15.
Immunity ; 31(4): 527-8, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19833080

ABSTRACT

Immunotherapy, especially therapeutic vaccination, has a great deal of potential in the treatment of cancer and certain infectious diseases such as HIV (Allison et al., 2006; Fauci et al., 2008; Feldmann and Steinman, 2005). Numerous vaccine candidates have been tested in patients with a variety of tumor types and chronic viral diseases. Often, the best way to assess the clinical potential of these vaccines is to monitor the induced T cell response, and yet there are currently no standards for reporting these results. This letter is an effort to address this problem.


Subject(s)
Cancer Vaccines/therapeutic use , Immunoassay/standards , Monitoring, Immunologic/standards , Neoplasms/therapy , Practice Guidelines as Topic/standards , T-Lymphocytes/immunology , Viral Vaccines/therapeutic use , Virus Diseases/therapy , Cancer Vaccines/immunology , Humans , Immunotherapy , Viral Vaccines/immunology
16.
Blood ; 114(9): 1736-45, 2009 Aug 27.
Article in English | MEDLINE | ID: mdl-19556425

ABSTRACT

Preclinical models have demonstrated the efficacy of granulocyte-macrophage colony-stimulating factor-secreting cancer immunotherapies (GVAX platform) accompanied by immunotherapy-primed lymphocytes after autologous stem cell transplantation in hematologic malignancies. We conducted a phase 2 study of this combination in adult patients with acute myeloid leukemia. Immunotherapy consisted of autologous leukemia cells admixed with granulocyte-macrophage colony-stimulating factor-secreting K562 cells. "Primed" lymphocytes were collected after a single pretransplantation dose of immunotherapy and reinfused with the stem cell graft. Fifty-four subjects were enrolled; 46 (85%) achieved a complete remission, and 28 (52%) received the pretransplantation immunotherapy. For all patients who achieved complete remission, the 3-year relapse-free survival (RFS) rate was 47.4% and overall survival was 57.4%. For the 28 immunotherapy-treated patients, the RFS and overall survival rates were 61.8% and 73.4%, respectively. Posttreatment induction of delayed-type hypersensitivity reactions to autologous leukemia cells was associated with longer 3-year RFS rate (100% vs 48%). Minimal residual disease was monitored by quantitative analysis of Wilms tumor-1 (WT1), a leukemia-associated gene. A decrease in WT1 transcripts in blood was noted in 69% of patients after the first immunotherapy dose and was also associated with longer 3-year RFS (61% vs 0%). In conclusion, immunotherapy in combination with primed lymphocytes and autologous stem cell transplantation shows encouraging signals of potential activity in acute myeloid leukemia (ClinicalTrials.gov: NCT00116467).


Subject(s)
Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunotherapy/methods , Leukemia, Myeloid, Acute/therapy , Stem Cell Transplantation/methods , Transplantation, Autologous/methods , Adult , Cell Line, Tumor , Disease-Free Survival , Female , Humans , Male , Middle Aged , Remission Induction , Treatment Outcome
17.
J Immunol ; 183(1): 696-705, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19535642

ABSTRACT

Previous work done in our laboratory, using mouse models, showed that soluble Fas ligand (sFasL) can efficiently delete donor anti-host T cells during their activation against irradiated host cells in MLCs. In the mouse models, this ex vivo sFasL treatment abrogated graft-vs-host disease (GVHD) while sparing donor T cells with antitumor reactivity. The present work was performed with human cells, to extend our work toward reduction of clinical GVHD. PBMC responders from a given individual (first party) were stimulated in vitro with irradiated PBMC stimulators from a second person (second party), in the presence of sFasL. In control MLCs without sFasL, alloreacting T cells began to up-regulate Fas (CD95) detectably and became sensitive to Fas-mediated apoptosis by as early as day 1-2. In MLCs containing sFasL, there were greatly reduced numbers of alloreacting CD3(+)CFSE(lo) cells, activation Ag-expressing CD4(hi) and CD8(hi) cells, IFN-gamma-producing CD4(+) and CD8(+) cells, and CD8(+)CD107a(+) CTLs. Furthermore, mice transplanted with the ex vivo sFasL/MLR-treated cells had prolonged time to fatal GVHD in an in vivo xenogeneic GVHD model. Responder cells harvested from primary MLCs containing sFasL had reduced proliferation in response to second party cells, but proliferated in response to CMV Ags, PHA, and third party cells. In addition, sFasL/MLR-treated cell populations contained influenza-specific T cells, CD4(+)FOXP3(+) T cells, and CD4(+)CD25(+) T cells. These data indicate that this ex vivo sFasL/MLR depletion of alloreacting human donor anti-host T cells was efficient and selective.


Subject(s)
Fas Ligand Protein/therapeutic use , Graft vs Host Disease/immunology , Graft vs Host Disease/prevention & control , Lymphocyte Depletion/methods , T-Lymphocyte Subsets/immunology , Animals , Cell Proliferation , Cells, Cultured , Female , Graft vs Host Disease/pathology , Humans , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mice, Transgenic , Solubility , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/pathology
18.
Cancer Res ; 69(7): 3180-7, 2009 Apr 01.
Article in English | MEDLINE | ID: mdl-19276358

ABSTRACT

A major parameter limiting immune responses to vaccination is the number of activated antigen-presenting cells (APC) that capture antigen and migrate to draining lymph nodes (LN). Currently, a quantitative noninvasive technique for monitoring in vivo antigen capture and delivery is lacking. The use of cellular magnetic resonance (MR) imaging (MRI) is a promising approach for this purpose; however, cellular imaging currently requires ex vivo prelabeling of cells with contrast agents followed by reintroduction of cells into the subject being monitored. Here, we describe an in vivo labeling method, which relies upon cell-to-cell transfer of superparamagnetic iron oxide (SPIO) from tumor cells to endogenous APCs, in situ, to quantify APC delivery to LNs in a tumor vaccine model. Mice were immunized with a tumor cell-based vaccine that was irradiated and labeled with SPIO. APCs that had captured SPIO were imaged over time as they accumulated in LNs. We show here that MRI is capable of monitoring, in vivo, the trafficking of magnetically labeled APCs inducing a tumor-specific immune response, and that these cells can be magnetically recovered ex vivo. Excellent correlation was observed between in vivo and ex vivo quantification of APCs, with resolution sufficient to detect increased APC trafficking elicited by an adjuvant. This study shows the potential of magnetovaccination and MRI cell tracking to systematically evaluate a key parameter relevant to the optimization of vaccine therapies through noninvasive MRI-based quantification of APC numbers.


Subject(s)
Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Ferric Compounds/administration & dosage , Lymph Nodes/immunology , Magnetic Resonance Imaging/methods , Magnetics/methods , Adjuvants, Immunologic/pharmacology , Aminoquinolines/pharmacology , Animals , Antigen Presentation , Cancer Vaccines/immunology , Female , Ferric Compounds/analysis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Image Processing, Computer-Assisted , Imiquimod , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL
19.
Proc Natl Acad Sci U S A ; 105(38): 14342-6, 2008 Sep 23.
Article in English | MEDLINE | ID: mdl-18794521

ABSTRACT

Antigen specific T cell migration to sites of infection or cancer is critical for an effective immune response. In mouse models of cancer, the number of lymphocytes reaching the tumor is typically only a few hundred, yet technology capable of imaging these cells using bioluminescence has yet to be achieved. A combination of codon optimization, removal of cryptic splice sites and retroviral modification was used to engineer an enhanced firefly luciferase (ffLuc) vector. Compared with ffLuc, T cells expressing our construct generated >100 times more light, permitting detection of as few as three cells implanted s.c. while maintaining long term coexpression of a reporter gene (Thy1.1). Expression of enhanced ffLuc in mouse T cells permitted the tracking of <3 x 10(4) adoptively transferred T cells infiltrating sites of vaccination and preestablished tumors. Penetration of light through deep tissues, including the liver and spleen, was also observed. Finally, we were able to enumerate infiltrating mouse lymphocytes constituting <0.3% of total tumor cellularity, representing a significant improvement over standard methods of quantitation including flow cytometry.


Subject(s)
Immunocompetence , Luciferases, Firefly/metabolism , Neoplasms/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Line, Tumor , Cell Movement , Cells, Cultured , Dendritic Cells/immunology , Disease Models, Animal , Gene Expression Regulation, Enzymologic , Genetic Vectors/genetics , Luciferases, Firefly/genetics , Luminescent Measurements , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Protein Engineering , Retroviridae/genetics , T-Lymphocytes/metabolism , Transduction, Genetic
20.
Biol Blood Marrow Transplant ; 14(5): 499-509, 2008 May.
Article in English | MEDLINE | ID: mdl-18410892

ABSTRACT

The "allogeneic effect" refers to the induction of host B cell antibody synthesis or host T cell cytotoxicity, including tumoricidal activity, by an infusion of allogeneic lymphocytes. We show that treatment of mice with cyclophosphamide (Cy) followed by CD8(+) T cell-depleted allogeneic donor lymphocyte infusion (Cy + CD8(-) DLI) induces regression of established tumors with minimal toxicity in models of both hematologic and solid cancers, even though the donor cells are eventually rejected by the host immune system. The optimal antitumor effect of Cy + CD8(-) DLI required the presence of donor CD4(+) T cells, host CD8(+) T cells, and alloantigen expression by normal host but not tumor tissue. The results support a model in which a donor CD4(+) T cell-mediated graft-versus-host (GVH) reaction effectively awakens antitumor immunity among Cy-resistant host CD8(+) T cells. These events provide the cellular mechanism of the "allogeneic effect" in antitumor immunity. Cy + CD8(-) DLI may be an effective and minimally toxic strategy for awakening the host immune response to advanced cancers.


Subject(s)
Graft vs Host Reaction/immunology , Lymphocyte Transfusion/methods , Animals , CD4-Positive T-Lymphocytes/transplantation , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cyclophosphamide/therapeutic use , Lymphocyte Depletion , Mice , Neoplasms/immunology , Neoplasms/therapy , Transplantation, Homologous , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...