Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cell ; 179(4): 829-845.e20, 2019 Oct 31.
Article in English | MEDLINE | ID: mdl-31675496

ABSTRACT

The immune microenvironment of hepatocellular carcinoma (HCC) is poorly characterized. Combining two single-cell RNA sequencing technologies, we produced transcriptomes of CD45+ immune cells for HCC patients from five immune-relevant sites: tumor, adjacent liver, hepatic lymph node (LN), blood, and ascites. A cluster of LAMP3+ dendritic cells (DCs) appeared to be the mature form of conventional DCs and possessed the potential to migrate from tumors to LNs. LAMP3+ DCs also expressed diverse immune-relevant ligands and exhibited potential to regulate multiple subtypes of lymphocytes. Of the macrophages in tumors that exhibited distinct transcriptional states, tumor-associated macrophages (TAMs) were associated with poor prognosis, and we established the inflammatory role of SLC40A1 and GPNMB in these cells. Further, myeloid and lymphoid cells in ascites were predominantly linked to tumor and blood origins, respectively. The dynamic properties of diverse CD45+ cell types revealed by this study add new dimensions to the immune landscape of HCC.


Subject(s)
Carcinoma, Hepatocellular/immunology , Cation Transport Proteins/genetics , Inflammation/immunology , Liver Neoplasms/immunology , Membrane Glycoproteins/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Communication/genetics , Cell Communication/immunology , Cell Lineage/genetics , Cell Lineage/immunology , Dendritic Cells/immunology , Dendritic Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Inflammation/genetics , Inflammation/pathology , Leukocyte Common Antigens/immunology , Liver/immunology , Liver/pathology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocytes/immunology , Lymphocytes/pathology , Lysosomal Membrane Proteins/genetics , Macrophages/immunology , Macrophages/pathology , Myeloid Cells/immunology , Myeloid Cells/pathology , Neoplasm Proteins/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Transcriptome/genetics , Transcriptome/immunology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
2.
J Mol Diagn ; 21(5): 796-807, 2019 09.
Article in English | MEDLINE | ID: mdl-31255795

ABSTRACT

Clinical biomarker studies are often hindered by the scarcity or suboptimal quality of biological specimens. EdgeSeq, a transcriptomics analysis platform, combines quantitative nuclease protection assay technology with next-generation sequencing, using small amounts of starting material and delivering reproducible gene expression profiles from challenging material, such as formalin-fixed, paraffin-embedded (FFPE) tissue. To evaluate EdgeSeq for analysis of archives of stained FFPE tissue, EdgeSeq was performed on unstained, hematoxylin and eosin (H&E)-stained, and immunohistochemistry-stained slides from patients with small-cell and non-small-cell lung cancer. Pairwise comparisons of gene expression profiles from stained and unstained slides showed higher Pearson correlation coefficients with H&E staining (0.86 to 0.97) than with immunohistochemistry staining (0.21 to 0.56). A 25-gene interferon-γ signature score from unstained slides showed a Pearson correlation coefficient of 0.92 with H&E-stained slides and a significant Spearman correlation (P = 0.0025) with immune scores. To test gene expression profiling in small samples, FFPE sample equivalents were examined from 5.0 to 0.08 mm2 of a section (5 µm thick); sample equivalents ≥0.31 mm2 showed alignment rates >69% and pairwise Pearson correlation coefficients ≥0.87. EdgeSeq can, thus, be used to profile small and H&E-stained FFPE tumor specimens to obtain biomarker data from limited tissue in oncology clinical trials and enable research into tumor microenvironment and immune cell engagement with tumors at the locoregional level.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Gene Expression Profiling/methods , High-Throughput Nucleotide Sequencing/methods , Lung Neoplasms/genetics , Paraffin Embedding/methods , Small Cell Lung Carcinoma/genetics , Colorectal Neoplasms/diagnosis , Eosine Yellowish-(YS)/chemistry , Formaldehyde/chemistry , Gene Library , Hematoxylin/chemistry , Humans , Lung Neoplasms/diagnosis , Small Cell Lung Carcinoma/diagnosis , Tissue Fixation
3.
J Immunother ; 39(4): 181-7, 2016 May.
Article in English | MEDLINE | ID: mdl-27070449

ABSTRACT

Two cancer testis antigens, the New York esophageal squamous cell carcinoma-1 (NY-ESO-1) and the melanoma-antigen family A (MAGE-A), represent promising immunotherapy targets due to the low expression of these antigens in nonmalignant tissue. To assess overexpression patterns in various cancers, we performed a systematic immunohistochemical analysis for NY-ESO-1 and MAGE-A on tissue array samples of 3668 common epithelial carcinomas (CA) and germ cell tumors of high prevalence and mortality. Here, we find significantly higher expression of MAGE-A (>50% on tumor cells) compared with NY-ESO-1 in several CAs including cutaneous squamous cell carcinomas (SCC) (52.8%/2.8%), esophageal SCC (50%/0%), head and neck SCC (41.1%/<1%), bladder urothelial CA (40.4%/8.3%), cervical/anal SCC (37.5%/0%), lung SCC (34%/3.8%), lung adenocarcinomas (27.6%/3.9%), ovarian CA (26.4%/3.6%), endometrial CA (26.3%/1.3%), lung small cell CA (24.4%/2.4%), gastric adenocarcinomas (20%/4%), breast mucinous CA (19.3%/0%), hepatocellular CA (18.8%/1.2%), breast infiltrating ductal CA (16.4%/1.8%), colorectal adenocarcinomas (10.7%/<1%), cholangiocarcinomas (9.8%/0%), thymic CA (9%/4.5%), and mesotheliomas (7.9%/<1%). Furthermore, high expression of MAGE-A, but not NY-ESO-1, was seen in whole slide evaluations of an independent cohort of metastatic SCC (45.5%/3.6%) and metastatic CA (13.5%/0%) of various primaries with significantly higher expression of MAGE-A in metastatic SCC compared with other metastatic CA. MAGE-A is also more highly expressed in germ cell tumors, seminomas (69%/3.5%) and nonseminomas (40.1%/4.7%). In summary, MAGE-A is more highly expressed than NY-ESO-1 in a majority of human malignancies, and targeting MAGE-A may benefit a large number of patients.


Subject(s)
Antigens, Neoplasm/metabolism , Carcinoma, Squamous Cell/metabolism , Esophageal Neoplasms/metabolism , Head and Neck Neoplasms/metabolism , Melanoma-Specific Antigens/metabolism , Membrane Proteins/metabolism , Neoplasms, Germ Cell and Embryonal/metabolism , Skin Neoplasms/metabolism , Carcinoma, Squamous Cell/pathology , Cohort Studies , Esophageal Neoplasms/pathology , Female , Head and Neck Neoplasms/pathology , Humans , Immunohistochemistry , Male , Neoplasm Metastasis , Neoplasms, Germ Cell and Embryonal/pathology , Skin Neoplasms/pathology , Tissue Array Analysis
4.
J Immunother ; 39(1): 1-7, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26641256

ABSTRACT

Melanoma-associated antigen-A (MAGE-A) and New York esophageal squamous cell cancer-1 (NY-ESO-1) are 2 cancer testis antigens (CTA) demonstrating potential for use in targeted immunotherapy. Clinical trials in melanoma and synovial sarcomas targeting these antigens in immune-based therapies have demonstrated durable tumor regression. Although protein expression of NY-ESO-1 has been assessed in a variety of cancer types, the expression of MAGE-A has not been studied in depth. In this study we analyzed MAGE-A and NY-ESO-1 expression in 314 melanoma specimens from 301 melanoma patients, 38 patients with squamous cell cancers and 111 patients with adenocarcinomas. Our results demonstrated higher expression of MAGE-A compared with NY-ESO-1 in melanomas (32% vs. 13%) and squamous cell carcinomas (45% vs. 7.9%), and higher expression of both CTAs in metastatic versus primary tumors. CTA expression in adenocarcinomas was low (MAGE-A: 10%, NY-ESO-1: 0.9%). In addition, we looked at concordance of expression among metastatic melanoma lesions within the same patient and found concordant expression in 38 of 47 patients for MAGE-A and 43 of 47 patients for NY-ESO-1. Our study demonstrated that the MAGE-A family may be of greater utility than NY-ESO-1 for targeted immunotherapy in a variety of cancer histologies, in particular metastatic melanomas and squamous cell carcinomas.


Subject(s)
Antigens, Neoplasm/metabolism , Membrane Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biomarkers, Tumor , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Female , Gene Expression , Humans , Immunohistochemistry , Male , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Membrane Proteins/genetics , Membrane Proteins/immunology , Neoplasm Metastasis , Neoplasm Staging , Neoplasms/genetics
5.
Semin Cancer Biol ; 35 Suppl: S199-S223, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25865775

ABSTRACT

Cancer arises in the context of an in vivo tumor microenvironment. This microenvironment is both a cause and consequence of tumorigenesis. Tumor and host cells co-evolve dynamically through indirect and direct cellular interactions, eliciting multiscale effects on many biological programs, including cellular proliferation, growth, and metabolism, as well as angiogenesis and hypoxia and innate and adaptive immunity. Here we highlight specific biological processes that could be exploited as targets for the prevention and therapy of cancer. Specifically, we describe how inhibition of targets such as cholesterol synthesis and metabolites, reactive oxygen species and hypoxia, macrophage activation and conversion, indoleamine 2,3-dioxygenase regulation of dendritic cells, vascular endothelial growth factor regulation of angiogenesis, fibrosis inhibition, endoglin, and Janus kinase signaling emerge as examples of important potential nexuses in the regulation of tumorigenesis and the tumor microenvironment that can be targeted. We have also identified therapeutic agents as approaches, in particular natural products such as berberine, resveratrol, onionin A, epigallocatechin gallate, genistein, curcumin, naringenin, desoxyrhapontigenin, piperine, and zerumbone, that may warrant further investigation to target the tumor microenvironment for the treatment and/or prevention of cancer.


Subject(s)
Carcinogenesis/drug effects , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , Tumor Microenvironment/genetics , Antineoplastic Agents/therapeutic use , Carcinogenesis/genetics , Cell Proliferation/drug effects , Humans , Molecular Targeted Therapy , Neoplasms/genetics , Neoplasms/prevention & control , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Signal Transduction , Tumor Microenvironment/drug effects
6.
Clin Cancer Res ; 21(10): 2278-88, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25695689

ABSTRACT

PURPOSE: Infusion of interleukin-12 (IL12) can mediate antitumor immunity in animal models, yet its systemic administration to patients with cancer results in minimal efficacy and severe toxicity. Here, we evaluated the antitumor activity of adoptively transferred human tumor-infiltrating lymphocytes (TILs) genetically engineered to secrete single-chain IL12 selectively at the tumor site. EXPERIMENTAL DESIGN: Thirty-three patients with metastatic melanoma were treated in a cell dose-escalation trial of autologous TILs transduced with a gene encoding a single-chain IL12 driven by a nuclear factor of the activated T cells promoter (NFAT.IL12). No IL2 was administered. RESULTS: The administration of 0.001 to 0.1 × 10(9) NFAT.IL12-transduced TILs to 17 patients resulted in a single, objective response (5.9%). However, at doses between 0.3 and 3 × 10(9) cells, 10 of 16 patients (63%) exhibited objective clinical responses. The responses tended to be short, and the administered IL12-producing cells rarely persisted at 1 month. Increasing cell doses were associated with high serum levels of IL12 and IFNγ as well as clinical toxicities, including liver dysfunction, high fevers, and sporadic life-threatening hemodynamic instability. CONCLUSIONS: In this first-in-man trial, administration of TILs transduced with an inducible IL12 gene mediated tumor responses in the absence of IL2 administration using cell doses 10- to 100-fold lower than conventional TILs. However, due to toxicities, likely attributable to the secreted IL12, further refinement will be necessary before this approach can be safely used in the treatment of cancer patients.


Subject(s)
Interleukin-12/genetics , Lymphocytes, Tumor-Infiltrating/physiology , Melanoma/therapy , Skin Neoplasms/therapy , Adult , Aged , Cells, Cultured , Female , Genetic Engineering , Humans , Immunotherapy , Interleukin-12/biosynthesis , Lymphocytes, Tumor-Infiltrating/transplantation , Male , Melanoma/immunology , Melanoma/secondary , Middle Aged , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Transcriptional Activation , Treatment Outcome , Young Adult
7.
Cancer Res ; 75(2): 296-305, 2015 Jan 15.
Article in English | MEDLINE | ID: mdl-25432172

ABSTRACT

Adoptive cell therapy (ACT) using autologous tumor-infiltrating lymphocytes (TIL) results in complete regression of advanced cancer in some patients, but the efficacy of this potentially curative therapy may be limited by poor persistence of TIL after adoptive transfer. Pharmacologic inhibition of the serine/threonine kinase Akt has recently been shown to promote immunologic memory in virus-specific murine models, but whether this approach enhances features of memory (e.g., long-term persistence) in TIL that are characteristically exhausted and senescent is not established. Here, we show that pharmacologic inhibition of Akt enables expansion of TIL with the transcriptional, metabolic, and functional properties characteristic of memory T cells. Consequently, Akt inhibition results in enhanced persistence of TIL after adoptive transfer into an immunodeficient animal model and augments antitumor immunity of CD8 T cells in a mouse model of cell-based immunotherapy. Pharmacologic inhibition of Akt represents a novel immunometabolomic approach to enhance the persistence of antitumor T cells and improve the efficacy of cell-based immunotherapy for metastatic cancer.


Subject(s)
Immunotherapy, Adoptive/methods , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma, Experimental/therapy , Melanoma/therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Animals , Humans , Immunologic Memory , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/pathology , Melanoma/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Transgenic , Proto-Oncogene Proteins c-akt/immunology , Random Allocation , Tumor Cells, Cultured
8.
Int J Surg Case Rep ; 5(10): 677-80, 2014.
Article in English | MEDLINE | ID: mdl-25194603

ABSTRACT

INTRODUCTION: Pythiosis is a serious life- and limb-threatening infection endemic to Thailand, but rarely seen in the Western hemisphere. Here, we present a unique case of vascular pythiosis initially managed with limb-sparing vascular bypass grafts complicated by a pseudoaneurysm in our repair. PRESENTATION OF CASE: The patient is a 17 year-old Jamaican male with severe aplastic anemia. He sustained a minor injury to his left leg while fishing in Jamaica, which evolved to become an exquisitely tender inguinal swelling. His physical exam and imaging were significant for arteriovenous fistula with limb ischemia. Pathology obtained during surgery for an extra-anatomic vascular bypass showed extensive invasion by Pythium insidiosum. He later developed a pseudoaneurysm at the site of proximal anastomosis and required urgent intervention. DISCUSSION: This patient presented with a rare, but classic case of vascular pythiosis, which was unrecognized at the time of presentation. A variety of therapeutic modalities have been used to treat this disease, including antibiotics, antifungals, and immunotherapy, but the ultimate management of vascular pythiosis is surgical source control. CONCLUSION: A high index of suspicion in susceptible patients is needed for timely diagnosis of vascular pythiosis to achieve optimal source control.

9.
Immunology ; 143(1): 96-108, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24678989

ABSTRACT

A growing body of evidence suggests that inflammatory cytokines have a dualistic role in immunity. In this study, we sought to determine the direct effects of interferon-γ (IFN-γ) on the differentiation and maturation of human peripheral blood monocyte-derived dendritic cells (moDC). Here, we report that following differentiation of monocytes into moDC with granulocyte-macrophage colony-stimulating factor and interleukin-4, IFN-γ induces moDC maturation and up-regulates the co-stimulatory markers CD80/CD86/CD95 and MHC Class I, enabling moDC to effectively generate antigen-specific CD4(+) and CD8(+) T-cell responses for multiple viral and tumour antigens. Early exposure of monocytes to high concentrations of IFN-γ during differentiation promotes the formation of macrophages. However, under low concentrations of IFN-γ, monocytes continue to differentiate into dendritic cells possessing a unique gene-expression profile, resulting in impairments in subsequent maturation by IFN-γ or lipopolysaccharide and an inability to generate effective antigen-specific CD4(+) and CD8(+) T-cell responses. These findings demonstrate that IFN-γ imparts differential programmes on moDC that shape the antigen-specific T-cell responses they induce. Timing and intensity of exposure to IFN-γ can therefore determine the functional capacity of moDC.


Subject(s)
Cell Differentiation/immunology , Dendritic Cells/immunology , Interferon-gamma/immunology , Monocytes/cytology , Monocytes/immunology , Cell Line , Cytokines/biosynthesis , Dendritic Cells/cytology , Flow Cytometry , Humans , Transcriptome
10.
Front Immunol ; 4: 304, 2013 Sep 27.
Article in English | MEDLINE | ID: mdl-24098300

ABSTRACT

T lymphocytes first carried foreign genes safely into humans over two decades ago. Since these pioneering studies, scientific techniques to better understand the genomic landscape of cells has directly led to a more sophisticated appreciation of the diversity, functional complexity, and therapeutic potential of T cells. Through the use of mouse models, we now know the function of the many genes that are critical for T cells to recognize foreign, mutated, or self-antigens and the factors responsible for the lineage diversification of T cells that lead to inhibitory or stimulatory immune responses. This knowledge combined with well-established modalities to introduce genes into T cells allows for the design of effector and memory CD8 and CD4 T lymphocytes specific for viral, fungal, bacterial, parasitic, and tumor-antigens and to design regulatory lymphocytes specific for the self-antigens responsible for autoimmune and inflammatory diseases. Here, I review strategies for designing the ideal T cell by introducing genes controlling (1) the secretion of cytokines/chemokines and their receptors, (2) T-cell receptor specificity, (3) chimeric-antigen receptors that enable for the recognition of surface antigens in an MHC-independent fashion, (4) co-stimulatory/inhibitory surface molecules, and (5) disease defining single-gene factors.

11.
Mol Ther ; 21(7): 1369-77, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23568260

ABSTRACT

Engineering CD8⁺ T cells to deliver interleukin 12 (IL-12) to the tumor site can lead to striking improvements in the ability of adoptively transferred T cells to induce the regression of established murine cancers. We have recently shown that IL-12 triggers an acute inflammatory environment that reverses dysfunctional antigen presentation by myeloid-derived cells within tumors and leads to an increase in the infiltration of adoptively transferred antigen-specific CD8⁺ T cells. Here, we find that local delivery of IL-12 increased the expression of Fas within tumor-infiltrating macrophages, dendritic cells, and myeloid-derived suppressor cells (MDSC), and that these changes were abrogated in mice deficient in IL-12-receptor signaling. Importantly, upregulation of Fas in host mice played a critical role in the proliferation and antitumor activity of adoptively transferred IL-12-modified CD8⁺ T cells. We also observed higher percentages of myeloid-derived cell populations within tumors in Fas-deficient mice, indicating that tumor stromal destruction was dependent on the Fas death receptor. Taken together, these results describe the likely requirement for costimulatory reverse signaling through Fasl on T cells that successfully infiltrate tumors, a mechanism triggered by the induction of Fas expression on myeloid-derived cells by IL-12 and the subsequent collapse of the tumor stroma.


Subject(s)
Interleukin-12/therapeutic use , Melanoma, Experimental/metabolism , fas Receptor/metabolism , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells/metabolism , Female , Flow Cytometry , Interleukin-12/administration & dosage , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , fas Receptor/genetics
12.
Cancer Res ; 72(13): 3125-30, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22721837

ABSTRACT

Established tumors are complex masses that contain not only neoplastic cells but also nontransformed cellular elements such as stromal cells, the neovasculature, and the full gamut of immune cells. However, evidence suggests that, unlike cells found in lymphoid organs that productively respond to acute infections, immune cells in tumors are dysregulated and functionally impaired. Tumor masses can contain regulatory lymphocytes, myeloid-derived suppressor cells, alternatively activated macrophages, and dendritic cells. Ablation or reprogramming of this aberrant microenvironment might dramatically augment cancer therapies, and this strategy is currently being deployed in a variety of clinical trials. A better understanding of the cellular constituents of tumors and the mechanisms involved in immune evasion may help guide the next generation of innovative cancer immunotherapies.


Subject(s)
Tumor Escape , Tumor Microenvironment , Humans , Immunotherapy , Interleukin-12/genetics , Macrophage Activation , Neoplasms/immunology , Neoplasms/therapy
13.
Blood ; 119(18): 4096-7, 2012 May 03.
Article in English | MEDLINE | ID: mdl-22555659

ABSTRACT

In this issue of Blood, Pegram and colleagues describe the ability to improve adoptive T-cell therapies by engineering T cells to secrete the potent proinflammatory cytokine IL-12.1 These exciting findings significantly extend previous observations made in a murine melanoma model targeting naturally occurring tumor antigens.2,3

14.
Clin Cancer Res ; 18(6): 1672-83, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22291136

ABSTRACT

PURPOSE: We investigated the feasibility of delivering the proinflammatory cytokine interleukin (IL)-12 into tumor using T cells genetically engineered to express a chimeric antigen receptor (CAR) against the VEGF receptor-2 (VEGFR-2). EXPERIMENTAL DESIGN: Two different strains of mice bearing five different established subcutaneous tumors were treated with syngeneic T cells cotransduced with an anti-VEGFR-2 CAR and a constitutively expressed single-chain murine IL-12 or an inducible IL-12 gene after host lymphodepletion. Tumor regression, survival of mice, and persistence of the transferred cells were evaluated. RESULTS: Adoptive transfer of syngeneic T cells cotransduced with an anti-VEGFR-2 CAR and a constitutively expressing single-chain IL-12 resulted in the regression of five different established tumors of different histologies without the need for IL-2 administration. T cells transduced with either anti-VEGFR-2 CAR or single-chain IL-12 alone did not alter the tumor growth indicating that both of them had to be expressed in the same cell to mediate tumor regression. Anti-VEGFR-2 CAR and IL-12-cotransduced T cells infiltrated the tumors, expanded, and persisted for prolonged periods. The antitumor effect did not require the presence of host T and B cells but was dependent on host IL-12R-expressing cells. The anti-VEGFR-2 CAR changed the immunosuppressive tumor environment by altering/reducing both the systemic and the intratumoral CD11b(+)Gr1(+) myeloid suppressor cell subsets that expressed VEGFR-2. CONCLUSIONS: These results suggest that targeted delivery of IL-12 into the tumor environment with T cells redirected against VEGFR-2 is a promising approach for treating patients with a variety of solid tumor types.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-12/administration & dosage , Interleukin-12/genetics , Neoplasms/therapy , T-Lymphocytes/immunology , Transduction, Genetic , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , Feasibility Studies , Female , Humans , Interleukin-12/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasms/blood supply , Receptors, Antigen, T-Cell/immunology , Recombinant Proteins/metabolism
15.
Immunity ; 35(6): 972-85, 2011 Dec 23.
Article in English | MEDLINE | ID: mdl-22177921

ABSTRACT

Th17 cells have been described as short lived, but this view is at odds with their capacity to trigger protracted damage to normal and transformed tissues. We report that Th17 cells, despite displaying low expression of CD27 and other phenotypic markers of terminal differentiation, efficiently eradicated tumors and caused autoimmunity, were long lived, and maintained a core molecular signature resembling early memory CD8(+) cells with stem cell-like properties. In addition, we found that Th17 cells had high expression of Tcf7, a direct target of the Wnt and ß-catenin signaling axis, and accumulated ß-catenin, a feature observed in stem cells. In vivo, Th17 cells gave rise to Th1-like effector cell progeny and also self-renewed and persisted as IL-17A-secreting cells. Multipotency was required for Th17 cell-mediated tumor eradication because effector cells deficient in IFN-γ or IL-17A had impaired activity. Thus, Th17 cells are not always short lived and are a less-differentiated subset capable of superior persistence and functionality.


Subject(s)
Stem Cells/metabolism , Th17 Cells/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Survival/genetics , Gene Expression Profiling , Interleukin-17/biosynthesis , Mice , Mice, Transgenic , Neoplasms/immunology , Stem Cells/cytology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Th1 Cells/cytology , Th1 Cells/immunology , Th17 Cells/cytology , Th17 Cells/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/genetics , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
16.
J Clin Invest ; 121(12): 4746-57, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22056381

ABSTRACT

Solid tumors are complex masses with a local microenvironment, or stroma, that supports tumor growth and progression. Among the diverse tumor-supporting stromal cells is a heterogeneous population of myeloid-derived cells. These cells are alternatively activated and contribute to the immunosuppressive environment of the tumor; overcoming their immunosuppressive effects may improve the efficacy of cancer immunotherapies. We recently found that engineering tumor-specific CD8(+) T cells to secrete the inflammatory cytokine IL-12 improved their therapeutic efficacy in the B16 mouse model of established melanoma. Here, we report the mechanism underlying this finding. Surprisingly, direct binding of IL-12 to receptors on lymphocytes or NK cells was not required. Instead, IL-12 sensitized bone marrow-derived tumor stromal cells, including CD11b(+)F4/80(hi) macrophages, CD11b(+)MHCII(hi)CD11c(hi) dendritic cells, and CD11b(+)Gr-1(hi) myeloid-derived suppressor cells, causing them to enhance the effects of adoptively transferred CD8(+) T cells. This reprogramming of myeloid-derived cells occurred partly through IFN-γ. Surprisingly, direct presentation of antigen to the transferred CD8(+) T cells by tumor was not necessary; however, MHCI expression on host cells was essential for IL-12-mediated antitumor enhancements. These results are consistent with a model in which IL-12 enhances the ability of CD8(+) T cells to collapse large vascularized tumors by triggering programmatic changes in otherwise suppressive antigen-presenting cells within tumors and support the use of IL-12 as part of immunotherapy for the treatment of solid tumors.


Subject(s)
CD8-Positive T-Lymphocytes/transplantation , Gene Expression Regulation, Neoplastic/drug effects , Immunotherapy, Adoptive , Interleukin-12/physiology , Melanoma, Experimental/therapy , Myeloid Cells/physiology , Tumor Escape/immunology , Tumor Microenvironment/immunology , Animals , Antigen Presentation , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/pathology , Antigens, Neoplasm/immunology , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Inflammation/genetics , Interferon-gamma/physiology , Interleukin-12/genetics , Interleukin-12/metabolism , Lymphocyte Subsets/immunology , Lymphocyte Subsets/pathology , Macrophages/physiology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Radiation Chimera , Recombinant Fusion Proteins/physiology , Stromal Cells/pathology , Stromal Cells/physiology
17.
Clin Cancer Res ; 17(16): 5343-52, 2011 Aug 15.
Article in English | MEDLINE | ID: mdl-21737507

ABSTRACT

PURPOSE: Adoptive cell transfer (ACT) of tumor infiltrating or genetically engineered T cells can cause durable responses in patients with metastatic cancer. Multiple clinically modifiable parameters can comprise this therapy, including cell dose and phenotype, in vivo antigen restimulation, and common gamma-chain (γ(c)) cytokine support. However, the relative contributions of each these individual components to the magnitude of the antitumor response have yet to be quantified. EXPERIMENTAL DESIGN: To systematically and quantitatively appraise each of these variables, we employed the Pmel-1 mouse model treating large, established B16 melanoma tumors. In addition to cell dose and magnitude of in vivo antigen restimulation, we also evaluated the relative efficacy of central memory (T(CM)), effector memory (T(EM)), and stem cell memory (T(SCM)) subsets on the strength of tumor regression as well as the dose and type of clinically available γ(c) cytokines, including IL-2, IL-7, IL-15, and IL-21. RESULTS: We found that cell dose, T-cell differentiation status, and viral vaccine titer each were correlated strongly and significantly with the magnitude of tumor regression. Surprisingly, although the total number of IL-2 doses was correlated with tumor regression, no significant benefit to prolonged (≥6 doses) administration was observed. Moreover, the specific type and dose of γ(c) cytokine only moderately correlated with response. CONCLUSION: Collectively, these findings elucidate some of the key determinants of successful ACT immunotherapy for the treatment of cancer in mice and further show that γ(c) cytokines offer a similar ability to effectively drive antitumor T-cell function in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Immunotherapy, Adoptive/methods , Melanoma, Experimental/immunology , Animals , CD8-Positive T-Lymphocytes/transplantation , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cell Differentiation/immunology , Cell Line, Tumor , Cytokines/therapeutic use , Dose-Response Relationship, Drug , Immunologic Memory/immunology , Interleukin-15/immunology , Interleukin-15/pharmacology , Interleukin-2/immunology , Interleukin-2/pharmacology , Interleukin-7/immunology , Interleukin-7/pharmacology , Interleukins/immunology , Interleukins/pharmacology , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Regression Analysis , Treatment Outcome , Tumor Burden/drug effects , Tumor Burden/immunology
18.
J Immunother ; 34(4): 343-52, 2011 May.
Article in English | MEDLINE | ID: mdl-21499127

ABSTRACT

T-cell receptor (TCR) gene therapy enables for the rapid creation of antigen-specific T cells from mice of any strain and represents a valuable tool for preclinical immunotherapy studies. Here, we describe the superiority of γ-retroviral vectors compared with lentiviral vectors for transduction of murine T cells and surprisingly illustrate robust gene-transfer into phenotypically naive/memory-stem cell like (TN/TSCM; CD62L(hi)/CD44(low)) and central memory (TCM; CD62L(hi)/CD44(hi)) CD8+ T cells using murine stem cell-based γ-retroviral vectors (MSGV1). We created MSGV1 vectors for a major histocompatibility complex-class I-restricted TCR specific for the melanocyte-differentiation antigen, glycoprotein 100 (MSGV1-pmel-1), and a major histocompatibility complex-class II-restricted TCR specific for tyrosinase-related protein-1 (MSGV1-TRP-1), and found that robust gene expression required codon optimization of TCR sequences for the pmel-1 TCR. To test for functionality, we adoptively transferred TCR-engineered T cells into mice bearing B16 melanomas and observed delayed growth of established tumors with pmel-1 TCR engineered CD8+ T cells and significant tumor regression with TRP-1 TCR transduced CD4 T cells. We simultaneously created lentiviral vectors encoding the pmel-1 TCR, but found that these vectors mediated low TCR expression in murine T cells, but robust gene expression in other murine and human cell lines. These results indicate that preclinical murine models of adoptive immunotherapies are more practical using γ-retroviral rather than lentiviral vectors.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Genetic Engineering , Immunotherapy, Adoptive , Animals , Gene Transfer Techniques , Genetic Vectors/genetics , HEK293 Cells , Humans , Jurkat Cells , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Retroviridae/genetics , Transduction, Genetic , gp100 Melanoma Antigen/genetics , gp100 Melanoma Antigen/immunology
19.
Mol Ther ; 19(4): 751-9, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21285960

ABSTRACT

Interleukin-12 (IL-12) is an important immunostimulatory cytokine, yet its clinical application has been limited by the systemic toxicity associated with its administration. In this work, we developed a strategy to selectively deliver IL-12 to the tumor environment using genetically engineered lymphocytes. However, peripheral blood lymphocytes (PBLs) transduced with a γ-retroviral vector, which constitutively expressed IL-12, failed to expand in culture due to apoptosis. To circumvent this problem, a vector was designed where IL-12 expression was directed by a composite promoter-containing binding motifs for nuclear factor of activated T-cells (NFAT.hIL12.PA2). The NFAT-responsive promoter was activated to drive IL-12 expression upon the recognition of tumor-specific antigen mediated by a T cell receptor (TCR) that was engineered into the same lymphocytes. We tested the efficacy of the inducible IL-12 vector in vivo in a murine melanoma model. Adoptive transfer of pmel-1 T cells genetically engineered with NFAT-murineIL12 (NFAT.mIL12.PA2) significantly enhanced regression of large established B16 melanoma. Notably, this targeted and controlled IL-12 treatment was without toxicity. Taken together, our results suggest that using the NFAT.hIL12.PA2 vector might be a promising approach to enhance adoptive cancer immunotherapy.


Subject(s)
Immunotherapy, Adoptive/methods , Interleukin-12/metabolism , Neoplasms/therapy , Animals , Cell Line , Cells, Cultured , Flow Cytometry , Genetic Vectors/genetics , Humans , Interleukin-12/genetics , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Retroviridae/genetics
20.
Blood ; 117(3): 808-14, 2011 Jan 20.
Article in English | MEDLINE | ID: mdl-20971955

ABSTRACT

Cluster of differentiation (CD)8(+) T cells exist as naive, central memory, and effector memory subsets, and any of these populations can be genetically engineered into tumor-reactive effector cells for adoptive immunotherapy. However, the optimal subset from which to derive effector CD8(+) T cells for patient treatments is controversial and understudied. We investigated human CD8(+) T cells and found that naive cells were not only the most abundant subset but also the population most capable of in vitro expansion and T-cell receptor transgene expression. Despite increased expansion, naive-derived cells displayed minimal effector differentiation, a quality associated with greater efficacy after cell infusion. Similarly, the markers of terminal differentiation, killer cell lectin-like receptor G1 and CD57, were expressed at lower levels in cells of naive origin. Finally, naive-derived effector cells expressed higher CD27 and retained longer telomeres, characteristics that suggest greater proliferative potential and that have been linked to greater efficacy in clinical trials. Thus, these data suggest that naive cells resist terminal differentiation, or "exhaustion," maintain high replicative potential, and therefore may be the superior subset for use in adoptive immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Immunotherapy, Adoptive/methods , T-Lymphocyte Subsets/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Differentiation/immunology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cells, Cultured , Cytokines/metabolism , Flow Cytometry , Gene Expression , Genetic Vectors/genetics , Humans , Retroviridae/genetics , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/transplantation , Telomere/genetics , Transduction, Genetic , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...