Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Nat Immunol ; 24(4): 612-624, 2023 04.
Article in English | MEDLINE | ID: mdl-36928415

ABSTRACT

Gamma delta (γδ) T cells reside within human tissues including tumors, but their function in mediating antitumor responses to immune checkpoint inhibition is unknown. Here we show that kidney cancers are infiltrated by Vδ2- γδ T cells, with equivalent representation of Vδ1+ and Vδ1- cells, that are distinct from γδ T cells found in normal human tissues. These tumor-resident Vδ2- T cells can express the transcriptional program of exhausted αß CD8+ T cells as well as canonical markers of terminal T-cell exhaustion including PD-1, TIGIT and TIM-3. Although Vδ2- γδ T cells have reduced IL-2 production, they retain expression of cytolytic effector molecules and co-stimulatory receptors such as 4-1BB. Exhausted Vδ2- γδ T cells are composed of three distinct populations that lack TCF7, are clonally expanded and express cytotoxic molecules and multiple Vδ2- T-cell receptors. Human tumor-derived Vδ2- γδ T cells maintain cytotoxic function and pro-inflammatory cytokine secretion in vitro. The transcriptional program of Vδ2- T cells in pretreatment tumor biopsies was used to predict subsequent clinical responses to PD-1 blockade in patients with cancer. Thus, Vδ2- γδ T cells within the tumor microenvironment can contribute to antitumor efficacy.


Subject(s)
CD8-Positive T-Lymphocytes , Kidney Neoplasms , Humans , CD8-Positive T-Lymphocytes/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Programmed Cell Death 1 Receptor/metabolism , Kidney Neoplasms/metabolism , T-Lymphocyte Subsets , Tumor Microenvironment
2.
Cell ; 181(7): 1612-1625.e13, 2020 06 25.
Article in English | MEDLINE | ID: mdl-32497499

ABSTRACT

Responses to anti-PD-1 immunotherapy occur but are infrequent in bladder cancer. The specific T cells that mediate tumor rejection are unknown. T cells from human bladder tumors and non-malignant tissue were assessed with single-cell RNA and paired T cell receptor (TCR) sequencing of 30,604 T cells from 7 patients. We find that the states and repertoires of CD8+ T cells are not distinct in tumors compared with non-malignant tissues. In contrast, single-cell analysis of CD4+ T cells demonstrates several tumor-specific states, including multiple distinct states of regulatory T cells. Surprisingly, we also find multiple cytotoxic CD4+ T cell states that are clonally expanded. These CD4+ T cells can kill autologous tumors in an MHC class II-dependent fashion and are suppressed by regulatory T cells. Further, a gene signature of cytotoxic CD4+ T cells in tumors predicts a clinical response in 244 metastatic bladder cancer patients treated with anti-PD-L1.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Urinary Bladder Neoplasms/genetics , Urinary Bladder Neoplasms/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Biomarkers, Pharmacological/analysis , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic/genetics , Genes, MHC Class II , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunotherapy , Lymphocytes, Tumor-Infiltrating , Programmed Cell Death 1 Receptor/genetics , Receptors, Antigen, T-Cell/genetics , Single-Cell Analysis/methods , T-Lymphocytes, Regulatory , Urinary Bladder Neoplasms/immunology
3.
Cell ; 168(3): 487-502.e15, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28111070

ABSTRACT

Immune responses involve coordination across cell types and tissues. However, studies in cancer immunotherapy have focused heavily on local immune responses in the tumor microenvironment. To investigate immune activity more broadly, we performed an organism-wide study in genetically engineered cancer models using mass cytometry. We analyzed immune responses in several tissues after immunotherapy by developing intuitive models for visualizing single-cell data with statistical inference. Immune activation was evident in the tumor and systemically shortly after effective therapy was administered. However, during tumor rejection, only peripheral immune cells sustained their proliferation. This systemic response was coordinated across tissues and required for tumor eradication in several immunotherapy models. An emergent population of peripheral CD4 T cells conferred protection against new tumors and was significantly expanded in patients responding to immunotherapy. These studies demonstrate the critical impact of systemic immune responses that drive tumor rejection.


Subject(s)
Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocyte Subsets/immunology , Animals , B7-H1 Antigen/antagonists & inhibitors , Bone Marrow/immunology , Cell Proliferation , Disease Models, Animal , Female , Humans , Immune Tolerance , Killer Cells, Natural/immunology , Lymphocyte Activation , Lymphoid Tissue/immunology , Male , Melanoma/immunology , Melanoma/therapy , Mice, Inbred BALB C , Mice, Inbred C57BL , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/therapy , Tumor Microenvironment
4.
Immunity ; 50(2): 477-492.e8, 2019 02 19.
Article in English | MEDLINE | ID: mdl-30737146

ABSTRACT

Resistance to checkpoint-blockade treatments is a challenge in the clinic. We found that although treatment with combined anti-CTLA-4 and anti-PD-1 improved control of established tumors, this combination compromised anti-tumor immunity in the low tumor burden (LTB) state in pre-clinical models as well as in melanoma patients. Activated tumor-specific T cells expressed higher amounts of interferon-γ (IFN-γ) receptor and were more susceptible to apoptosis than naive T cells. Combination treatment induced deletion of tumor-specific T cells and altered the T cell repertoire landscape, skewing the distribution of T cells toward lower-frequency clonotypes. Additionally, combination therapy induced higher IFN-γ production in the LTB state than in the high tumor burden (HTB) state on a per-cell basis, reflecting a less exhausted immune status in the LTB state. Thus, elevated IFN-γ secretion in the LTB state contributes to the development of an immune-intrinsic mechanism of resistance to combination checkpoint blockade, highlighting the importance of achieving the optimal magnitude of immune stimulation for successful combination immunotherapy strategies.


Subject(s)
Antibodies, Monoclonal/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Interferon-gamma/pharmacology , Neoplasms, Experimental/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes/drug effects , Animals , Antibodies, Monoclonal/immunology , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Cell Line, Tumor , Clonal Deletion/drug effects , Clonal Deletion/immunology , Drug Resistance, Neoplasm/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/immunology , Neoplasms, Experimental/metabolism , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Burden/drug effects , Tumor Burden/immunology
5.
Cancer Immunol Immunother ; 70(10): 3031-3040, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33864502

ABSTRACT

BACKGROUND: Soft-tissue sarcomas (STS) are a rare group of mesenchymal malignancies that account for approximately 1% of adult human cancer. Undifferentiated pleomorphic sarcoma (UPS) is one of the most common subtypes of adult STS. Clinical stratification of UPS patients has not evolved for decades and continues to rely on tumor-centric metrics including tumor size and depth. Our understanding of how the tumor microenvironment correlates to these clinicopathologic parameters remains limited. METHODS: Here, we performed single-cell flow cytometric immune-based profiling of 15 freshly resected UPS tumors and integrated this analysis with clinical, histopathologic, and outcomes data using both a prospective and retrospective cohort of UPS patients. RESULTS: We uncovered a correlation between physiologic and anatomic properties of UPS tumors and the composition of immune cells in the tumor microenvironment. Specifically, we identified an inverse correlation between tumor-infiltrating CD8 + T cells and UPS tumor size; and a positive correlation between tumor-infiltrating CD8 + T cells and overall survival. Moreover, we demonstrate an association between anatomical location (deep or superficial) and frequency of CD4 + PD1hi infiltrating T cells in UPS tumors. CONCLUSIONS: Our study provides an immune-based analysis of the tumor microenvironment in UPS patients and describes the different composition of tumor infiltrating lymphocytes based on size and tumor depth.


Subject(s)
Sarcoma/physiopathology , Soft Tissue Neoplasms/physiopathology , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Male , Middle Aged , Prospective Studies , Retrospective Studies , Tumor Microenvironment
6.
J Immunol ; 189(7): 3759-66, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22956585

ABSTRACT

CTLA-4 is a surface receptor on activated T cells that delivers an inhibitory signal, serving as an immune checkpoint. Treatment with anti-CTLA-4 Abs can induce clinical responses to different malignancies, but the nature of the induced Ag-specific recognition is largely unknown. Using microarrays spotted with >8000 human proteins, we assessed the diversity of Ab responses modulated by treatment with CTLA-4 blockade and GM-CSF. We find that advanced prostate cancer patients who clinically respond to treatment also develop enhanced Ab responses to a higher number of Ags than nonresponders. These induced Ab responses targeted Ags to which preexisting Abs are more likely to be present in the clinical responders compared with nonresponders. The majority of Ab responses are patient-specific, but immune responses against Ags shared among clinical responders are also detected. One of these shared Ags is PAK6, which is expressed in prostate cancer and to which CD4(+) T cell responses were also induced. Moreover, immunization with PAK6 can be both immunogenic and protective in mouse tumor models. These results demonstrate that immune checkpoint blockade modulates Ag-specific responses to both individualized and shared Ags, some of which can mediate anti-tumor responses.


Subject(s)
Antibody Diversity , Autoantigens/immunology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Epitopes, T-Lymphocyte/biosynthesis , Epitopes, T-Lymphocyte/immunology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Autoantibodies/biosynthesis , Autoantibodies/therapeutic use , Feasibility Studies , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Humans , Ipilimumab , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Neoplasm Transplantation/immunology , Neoplasm Transplantation/pathology , Prostatic Neoplasms/pathology , Tumor Cells, Cultured
7.
Cancer Immunol Res ; 12(4): 453-461, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38276989

ABSTRACT

Denosumab is a fully human mAb that binds receptor activator of NFκB ligand (RANKL). It is routinely administered to patients with cancer to reduce the incidence of new bone metastasis. RANK-RANKL interactions regulate bone turnover by controlling osteoclast recruitment, development, and activity. However, these interactions also can regulate immune cells including dendritic cells and medullary thymic epithelial cells. Inhibition of the latter results in reduced thymic negative selection of T cells and could enhance the generation of tumor-specific T cells. We examined whether administering denosumab could modify modulate circulating immune cells in patients with cancer. Blood was collected from 23 patients with prostate cancer and 3 patients with renal cell carcinoma, all of whom had advanced disease and were receiving denosumab, prior to and during denosumab treatment. Using high-dimensional mass cytometry, we found that denosumab treatment by itself induced modest effects on circulating immune cell frequency and activation. We also found minimal changes in the circulating T-cell repertoire and the frequency of new thymic emigrants with denosumab treatment. However, when we stratified patients by whether they were receiving chemotherapy and/or steroids, patients receiving these concomitant treatments showed significantly greater immune modulation, including an increase in the frequency of natural killer cells early and classical monocytes later. We also saw broad induction of CTLA-4 and TIM3 expression in circulating lymphocytes and some monocyte populations. These findings suggest that denosumab treatment by itself has modest immunomodulatory effects, but when combined with conventional cancer treatments, can lead to the induction of immunologic checkpoints. See related Spotlight by Nasrollahi and Davar, p. 383.


Subject(s)
Bone Neoplasms , Denosumab , Humans , Male , Bone Neoplasms/drug therapy , Bone Neoplasms/prevention & control , Bone Neoplasms/secondary , Denosumab/therapeutic use , Kidney Neoplasms/drug therapy , RANK Ligand/antagonists & inhibitors , Prostatic Neoplasms/drug therapy
8.
Blood Adv ; 2024 Apr 04.
Article in English | MEDLINE | ID: mdl-38574299

ABSTRACT

Multiple myeloma is characterized by frequent clinical relapses following conventional therapy. Recently, chimeric antigen receptor T (CAR-T) cells targeting B-cell maturation antigen (BCMA) has been established as a treatment option for patients with relapsed or refractory disease. However, while >70% of patients initially respond to this treatment, clinical relapse and disease progression occur in most cases. Recent studies showed persistent expression of BCMA at the time of relapse, indicating that immune intrinsic mechanisms may contribute to this resistance. While there were no pre-existing T cell features associated with clinical outcomes, we found that patients with a durable response to CAR-T cell treatment had greater persistence of their CAR-T cells compared to patients with transient clinical responses. They also possessed a significantly higher proportion of CD8+ T effector memory cells. In contrast, patients with short-lived responses to treatment have increased frequencies of cytotoxic CD4+ CAR-T cells. These cells expand in vivo early after infusion but express exhaustion markers (HAVCR2 and TIGIT) and remain polyclonal. Finally, we demonstrate that non-classical monocytes are enriched in the myeloma niche and may induce CAR-T cell dysfunction through mechanisms that include TGFß. These findings shed new light on the role of cytotoxic CD4+ T cells in disease progression after CAR-T cell therapy.

9.
Science ; 376(6589): eabf1970, 2022 04 08.
Article in English | MEDLINE | ID: mdl-35389781

ABSTRACT

Systemic lupus erythematosus (SLE) is a heterogeneous autoimmune disease. Knowledge of circulating immune cell types and states associated with SLE remains incomplete. We profiled more than 1.2 million peripheral blood mononuclear cells (162 cases, 99 controls) with multiplexed single-cell RNA sequencing (mux-seq). Cases exhibited elevated expression of type 1 interferon-stimulated genes (ISGs) in monocytes, reduction of naïve CD4+ T cells that correlated with monocyte ISG expression, and expansion of repertoire-restricted cytotoxic GZMH+ CD8+ T cells. Cell type-specific expression features predicted case-control status and stratified patients into two molecular subtypes. We integrated dense genotyping data to map cell type-specific cis-expression quantitative trait loci and to link SLE-associated variants to cell type-specific expression. These results demonstrate mux-seq as a systematic approach to characterize cellular composition, identify transcriptional signatures, and annotate genetic variants associated with SLE.


Subject(s)
Interferon Type I , Lupus Erythematosus, Systemic , CD8-Positive T-Lymphocytes/metabolism , Case-Control Studies , Humans , Interferon Type I/metabolism , Leukocytes, Mononuclear , Lupus Erythematosus, Systemic/genetics , RNA-Seq , Transcription, Genetic
10.
Cancer Res ; 77(6): 1322-1330, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28031229

ABSTRACT

While immune checkpoint blockade elicits efficacious responses in many patients with cancer, it also produces a diverse and unpredictable number of immune-related adverse events (IRAE). Mechanisms driving IRAEs are generally unknown. Because CTLA-4 blockade leads to proliferation of circulating T cells, we examined in this study whether ipilimumab treatment leads to clonal expansion of tissue-reactive T cells. Rather than narrowing the T-cell repertoire to a limited number of clones, ipilimumab induced greater diversification in the T-cell repertoire in IRAE patients compared with patients without IRAEs. Specifically, ipilimumab triggered increases in the numbers of clonotypes, including newly detected clones and a decline in overall T-cell clonality. Initial broadening in the repertoire occurred within 2 weeks of treatment, preceding IRAE onset. IRAE patients exhibited greater diversity of CD4+ and CD8+ T cells, but showed no differences in regulatory T-cell numbers relative to patients without IRAEs. Prostate-specific antigen responses to ipilimumab were also associated with increased T-cell diversity. Our results show how rapid diversification in the immune repertoire immediately after checkpoint blockade can be both detrimental and beneficial for patients with cancer. Cancer Res; 77(6); 1322-30. ©2016 AACR.


Subject(s)
Antibodies, Monoclonal/pharmacology , CTLA-4 Antigen/antagonists & inhibitors , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Prostatic Neoplasms, Castration-Resistant/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Humans , Ipilimumab , Male , Prognosis , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/pathology , T-Lymphocyte Subsets/drug effects , T-Lymphocytes, Regulatory/drug effects
11.
Oncoimmunology ; 5(4): e1101204, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27141383

ABSTRACT

We conducted a phase II clinical trial of anti-CTLA-4 antibody (ipilimumab) and granulocyte-macrophage colony-stimulating factor (GM-CSF) in 22 patients with metastatic melanoma and determined clinical outcomes and immunologic responses. The treatment consisted of a 3-mo induction with ipilimumab at 10 mg/kg administered every 3 weeks for four doses in combination with GM-CSF at 125 µg/m2 for 14 d beginning on the day of the ipilimumab infusion and then GM-CSF for 3 mo on the same schedule without ipilimumab. This was followed by maintenance therapy with the combination every 3 mo for up to 2 y or until disease progression or unacceptable toxicity. Blood samples for determination of immune subsets were obtained before treatment, at week 3 (end of cycle 1) and at week 6 (end of cycle 2). Blood samples were also obtained from seven subjects who were cancer-free. The immune response disease control (irDC) rate at 24 weeks was 41% and the overall response rate (ORR) was 32%. The median progression free-survival (PFS) was 3.5 mo and the median overall survival (OS) was 21.1 mo. 41% of the patients experienced Grade 3 to 4 adverse events. We conclude that this combination is safe and the results suggest the combination may be more effective than ipilimumab monotherapy. Further, the results suggest that lower levels of CD4+ effector T cells but higher levels of CD8+ T cells expressing PD-1 at pre-treatment could be a potential biomarker for disease control in patients who receive immunotherapy with ipilimumab and GM-CSF. Further trials of this combination are warranted.

12.
Cancer Immunol Res ; 3(9): 1008-16, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25968455

ABSTRACT

Cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) blockade can induce tumor regression and improved survival in cancer patients. This treatment can enhance adaptive immune responses without an exogenous vaccine, but the immunologic biomarkers associated with improved clinical outcome in cancer patients are not fully established. A phase Ib trial in patients with metastatic, castration-resistant prostate cancer was performed combining ipilimumab with sargramostim (GM-CSF). In addition to evaluating ipilimumab dose, patients were followed clinically for response and overall survival, and for immunomodulation of circulating T cells. PSA declines of ≥50% and radiographic responses were observed at doses of ≥3 mg/kg/dose. Timing of clinical responses could be either immediate or delayed. Durable responses were also observed off treatment. A subset of patients experienced long-term survival with or without objective clinical responses. The relationship between T-cell phenotype in peripheral blood and overall survival was examined retrospectively. We found that the treatment induced an increase in the levels of CD4(+) effector T (Teff) cells, regulatory T cells, PD-1(+) CD4 Teff cells, and PD-1(+) CD8 T cells. However, these increased levels were not associated with overall survival. Instead, low pretreatment baseline levels of PD-1(+) CD4 Teff cells were found to correlate with longer overall survival. Furthermore, baseline levels of PD-1(+) CD4 Teff cells from patients with shorter overall survival were higher than from cancer-free male control subjects. These results suggest that preexisting expression of immunologic checkpoint marker PD-1 on CD4 Teff cells may help identify patients that may benefit from ipilimumab treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , CD4-Positive T-Lymphocytes/immunology , Programmed Cell Death 1 Receptor/blood , Prostatic Neoplasms, Castration-Resistant/drug therapy , Aged , Aged, 80 and over , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , CTLA-4 Antigen/antagonists & inhibitors , Dose-Response Relationship, Drug , Follow-Up Studies , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Ipilimumab , Kallikreins/blood , Lymphocyte Count , Male , Middle Aged , Neoplasm Proteins/blood , Prognosis , Prostate-Specific Antigen/blood , Prostatic Neoplasms, Castration-Resistant/blood , Prostatic Neoplasms, Castration-Resistant/immunology , Recombinant Proteins/administration & dosage , Recombinant Proteins/adverse effects , T-Lymphocyte Subsets/immunology , Treatment Outcome
13.
Cancer Biol Ther ; 1(1): 39-44, 2002.
Article in English | MEDLINE | ID: mdl-12170763

ABSTRACT

Inactivation of p53 and expression of Bcl-2, frequently occurring during tumor progression, have different prognostic value: while inactivation of p53 is generally associated with unfavorable prognosis, expression of Bcl-2 often correlates with better clinical outcome and delays selection of metastatic variants of experimental tumors. To analyze the mechanisms underlying the "anti-progression" function of Bcl-2, we engineered tumor cell variants differing in their p53 status and Bcl-2 expression and compared their expansion in experimental tumors. Although neither p53 suppression nor Bcl-2-expression altered cell growth properties in vitro, both variants showed rapid accumulation in growing tumors in vivo, presumably due to their resistance to hypoxia. However, no expansion of p53-deficient variants occurred in the tumors formed by Bcl-2-overexpressing cells, indicating that p53 deficiency has no selective advantages in the Bcl-2-expressing environment. Importantly, expression of Bcl-2, unlike p53 suppression, did not lead to genomic instability as judged by the frequencies of gene amplification. Thus, acquisition of Bcl-2 expression is as advantageous for tumor cell growth in vivo as is p53 inactivation but does not affect genomic stability and creates the environment restrictive for the expansion of genetically unstable and potentially malignant p53-deficient cells, causing a delay in tumor progression and explaining the different prognostic value of Bcl-2 and p53.


Subject(s)
Apoptosis/genetics , Genes, bcl-2 , Mutation/genetics , Neoplasm Proteins/physiology , Neoplasms/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Tumor Suppressor Protein p53/deficiency , Animals , Cell Division , Cell Line, Transformed/drug effects , Cell Line, Transformed/pathology , Cricetinae , Disease Progression , Gene Expression Regulation, Neoplastic , Genes, Dominant , Genes, Reporter , Genes, p53 , Humans , Male , Mesocricetus , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms/pathology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Prognosis , Recombinant Fusion Proteins/physiology , Selection, Genetic , Tumor Suppressor Protein p53/physiology
14.
Nat Rev Cancer ; 12(4): 289-97, 2012 03 01.
Article in English | MEDLINE | ID: mdl-22378189

ABSTRACT

Although cancer cells can be immunogenic, tumour progression is associated with the evasion of immunosurveillance, the promotion of tumour tolerance and even the production of pro-tumorigenic factors by immune cells. Cytotoxic T lymphocyte-associated antigen 4 (CTLA4) represents a crucial immune checkpoint, the blockade of which can potentiate anti-tumour immunity. CTLA4-blocking antibodies are now an established therapeutic approach for malignant melanoma, and clinical trials with CTLA4-specific antibodies in prostate cancer have also shown clinical activity. This treatment may provide insights into the targets that the immune system recognizes to drive tumour regression, and could potentially improve both outcome and toxicity for patients with prostate cancer.


Subject(s)
Antibodies/therapeutic use , CTLA-4 Antigen/antagonists & inhibitors , Prostatic Neoplasms/therapy , Animals , Antibodies/pharmacology , Antigens, Neoplasm/immunology , CTLA-4 Antigen/immunology , CTLA-4 Antigen/metabolism , Clinical Trials as Topic , Humans , Immunomodulation , Immunotherapy , Lymphocyte Activation , Male , Mice , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism
15.
Cancer Res ; 69(2): 609-15, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-19147575

ABSTRACT

CTL-associated antigen 4 (CTLA4) is a costimulatory molecule expressed on activated T cells that delivers an inhibitory signal to these T cells. CTLA4 blockade with antibody treatment has been shown to augment antitumor immunity in animal models and is being developed as a treatment for cancer patients. As has been seen in preclinical models, combining CTLA4 blockade and granulocyte macrophage colony-stimulating factor (GM-CSF)-based immunotherapies can enhance the antitumor efficacy of this approach. We therefore examined whether CTLA4 blockade could be combined with GM-CSF administration. We treated 24 patients with metastatic, castration-resistant prostate cancer in a phase I trial where sequential cohorts were treated with increasing doses of ipilimumab, a fully human anti-CTLA4 antibody. Study subjects also received s.c. injections of GM-CSF at a fixed dose. Of the six patients treated at the highest dose level, three had confirmed PSA declines of >50%, including one patient that had a partial response in visceral metastases. Expansion of activated, circulating CD25(+) CD69(+) CD8(+) T cells occurred more frequently at higher doses of treatment and was greater in magnitude than was seen in patients who received the same doses of either ipilimumab or GM-CSF alone. By screening sera with protein arrays, we showed that our treatment can induce antibody responses to NY-ESO-1. These results show that this combination immunotherapy can induce the expansion not only of activated effector CD8 T cells in vivo but also of T cells that are specific for known tumor-associated antigens from the endogenous immune repertoire.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antigens, CD/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunotherapy/methods , Prostatic Neoplasms/therapy , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antigens, Neoplasm/immunology , Antigens, Neoplasm/pharmacology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen , Granulocyte-Macrophage Colony-Stimulating Factor/adverse effects , Humans , Immunotherapy/adverse effects , Ipilimumab , Lymphocyte Activation , Male , Middle Aged , Prostatic Neoplasms/immunology
SELECTION OF CITATIONS
SEARCH DETAIL