Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
1.
BMC Cancer ; 18(1): 1274, 2018 Dec 19.
Article in English | MEDLINE | ID: mdl-30567529

ABSTRACT

BACKGROUND: HDIL-2 is approved for advanced melanoma based on its durable antitumor activity. MAGE-A3 cancer immunotherapeutic (MAGE-A3 CI) is a recombinant MAGE-A3 protein combined with an immunostimulant adjuvant system and has shown antitumor activity in melanoma. We assessed the safety and anti-tumor activity of HDIL-2 combined with MAGE-A3 CI in advanced melanoma. METHODS: Patients with unresectable Stage III or Stage IV MAGE-A3-positive melanoma were enrolled in this phase II study. Treatment included an induction phase of MAGE-A3 CI plus HDIL-2 for 8 cycles followed by a maintenance phase of MAGE-A3 CI monotherapy. The primary endpoints were safety and objective response assessed per RECIST v1.1. Immune biomarker and correlative studies on tumor and peripheral blood were performed. RESULTS: Eighteen patients were enrolled. Seventeen patients were evaluable for safety and sixteen for response. Responses occurred in 4/16 (25%) patients with 3 complete responses, and stable disease in 6/16 (38%) patients with a disease control rate of 63%. The median duration of response was not reached at median follow-up of 36.8 months. Induction therapy of HDIL-2 + MAGE-A3 CI had similar toxicities to those reported with HDIL-2 alone. Maintenance MAGE-A3 monotherapy was well-tolerated. Increased immune checkpoint receptor expression by circulating T regulatory cells was associated with poor clinical outcomes; and responders tended to have increased tumor infiltrating T cells in the baseline tumor samples. CONCLUSIONS: The safety profile of HDIL-2 + MAGE-A3 CI was similar to HDIL-2 monotherapy. Maintenance MAGE-A3 CI provides robust anti-tumor activity in patients who achieved disease control with induction therapy. Immune monitoring data suggest that MAGE-A3 CI plus checkpoint inhibitors could be a promising treatment for MAGE-A3-positive melanoma. TRIAL REGISTRATION: ClinicalTrials.gov, NCT01266603 . Registered 12/24/2010, https://clinicaltrials.gov/ct2/show/NCT01266603.


Subject(s)
Antigens, Neoplasm/administration & dosage , Interleukin-2/administration & dosage , Melanoma/drug therapy , Neoplasm Proteins/administration & dosage , Recombinant Proteins/administration & dosage , Adjuvants, Immunologic , Adult , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Female , Humans , Immunotherapy , Interleukin-2/genetics , Interleukin-2/immunology , Male , Melanoma/genetics , Melanoma/immunology , Melanoma/pathology , Middle Aged , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Staging , Recombinant Proteins/genetics , Recombinant Proteins/immunology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Treatment Outcome
2.
J Clin Oncol ; 36(14): 1419-1427, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29432077

ABSTRACT

Purpose DNX-2401 (Delta-24-RGD; tasadenoturev) is a tumor-selective, replication-competent oncolytic adenovirus. Preclinical studies demonstrated antiglioma efficacy, but the effects and mechanisms of action have not been evaluated in patients. Methods A phase I, dose-escalation, biologic-end-point clinical trial of DNX-2401 was conducted in 37 patients with recurrent malignant glioma. Patients received a single intratumoral injection of DNX-2401 into biopsy-confirmed recurrent tumor to evaluate safety and response across eight dose levels (group A). To investigate the mechanism of action, a second group of patients (group B) underwent intratumoral injection through a permanently implanted catheter, followed 14 days later by en bloc resection to acquire post-treatment specimens. Results In group A (n = 25), 20% of patients survived > 3 years from treatment, and three patients had a ≥ 95% reduction in the enhancing tumor (12%), with all three of these dramatic responses resulting in > 3 years of progression-free survival from the time of treatment. Analyses of post-treatment surgical specimens (group B, n = 12) showed that DNX-2401 replicates and spreads within the tumor, documenting direct virus-induced oncolysis in patients. In addition to radiographic signs of inflammation, histopathologic examination of immune markers in post-treatment specimens showed tumor infiltration by CD8+ and T-bet+ cells, and transmembrane immunoglobulin mucin-3 downregulation after treatment. Analyses of patient-derived cell lines for damage-associated molecular patterns revealed induction of immunogenic cell death in tumor cells after DNX-2401 administration. Conclusion Treatment with DNX-2401 resulted in dramatic responses with long-term survival in recurrent high-grade gliomas that are probably due to direct oncolytic effects of the virus followed by elicitation of an immune-mediated antiglioma response.


Subject(s)
Adenoviridae , Brain Neoplasms/drug therapy , Glioma/drug therapy , Neoplasm Recurrence, Local/drug therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses , Adult , Biopsy , Female , Humans , Injections, Intralesional , Male , Middle Aged , Survival Rate , Treatment Outcome
3.
Clin Cancer Res ; 24(5): 1011-1018, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29246938

ABSTRACT

Purpose: Prevention or treatment of relapsed lymphoid malignancies after hematopoietic stem cell transplantation (HSCT) requires novel strategies. We hypothesized that antitumor-cell responses could be enhanced by the addition of lenalidomide to the cytotoxic T-lymphocyte-associated protein 4 inhibitor ipilimumab.Experimental Design: We conducted a phase II investigator-initiated trial to assess the safety and activity of ipilimumab and lenalidomide in patients with lymphoid malignancies that relapsed after allogeneic HSCT and in high-risk patients after autologous HSCT. Patients received 10 mg of oral lenalidomide daily for 21 days followed by intravenous ipilimumab at 3 mg/kg bodyweight. The regimen was repeated 4 weeks later for a total of four treatments.Results: We enrolled 17 patients (10 allogeneic and seven autologous transplant recipients). Immune-mediated toxicity was limited to one patient with asymptomatic hypothyroidism and one with dermatitis in the allogeneic and autologous groups, respectively. One allogeneic transplant recipient had a flare of prior GVHD while taking lenalidomide that precluded further treatment. All others finished treatment without GVHD. Four of 10 patients in the allogeneic group had complete responses (three of which were durable at 19+, 21+, and 32+ months), and three had partial responses. The disease in six of seven patients in the autologous group remains in remission. The groups had similar immune responses, including a two- to threefold increase in inducible ICOS+CD4+FoxP3- T-cell number.Conclusions: Our early-phase data suggested that ipilimumab plus lenalidomide is well tolerated after HSCT. Adverse events did not differ significantly between the allogeneic and autologous groups. Clin Cancer Res; 24(5); 1011-8. ©2017 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Hematopoietic Stem Cell Transplantation/methods , Ipilimumab/administration & dosage , Lenalidomide/administration & dosage , Lymphoma/therapy , Neoplasm Recurrence, Local/therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Graft vs Host Disease/chemically induced , Graft vs Host Disease/epidemiology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Ipilimumab/adverse effects , Lenalidomide/adverse effects , Lymphoma/immunology , Lymphoma/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Pilot Projects , Transplantation, Autologous/adverse effects , Transplantation, Autologous/methods , Transplantation, Homologous/adverse effects , Transplantation, Homologous/methods , Treatment Outcome
4.
Clin Cancer Res ; 23(20): 6151-6164, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28754817

ABSTRACT

Purpose: Adoptive T-cell therapy using autologous tumor-infiltrating lymphocytes (TIL) has shown an overall clinical response rate 40%-50% in metastatic melanoma patients. BTLA (B-and-T lymphocyte associated) expression on transferred CD8+ TILs was associated with better clinical outcome. The suppressive function of the ITIM and ITSM motifs of BTLA is well described. Here, we sought to determine the functional characteristics of the CD8+BTLA+TIL subset and define the contribution of the Grb2 motif of BTLA in T-cell costimulation.Experimental Design: We determined the functional role and downstream signal of BTLA in both human CD8+ TILs and mouse CD8+ T cells. Functional assays were used including single-cell analysis, reverse-phase protein array (RPPA), antigen-specific vaccination models with adoptively transferred TCR-transgenic T cells as well as patient-derived xenograft (PDX) model using immunodeficient NOD-scid IL2Rgammanull (NSG) tumor-bearing mice treated with autologous TILs.Results: CD8+BTLA- TILs could not control tumor growth in vivo as well as their BTLA+ counterpart and antigen-specific CD8+BTLA- T cells had impaired recall response to a vaccine. However, CD8+BTLA+ TILs displayed improved survival following the killing of a tumor target and heightened "serial killing" capacity. Using mutants of BTLA signaling motifs, we uncovered a costimulatory function mediated by Grb2 through enhancing the secretion of IL-2 and the activation of Src after TCR stimulation.Conclusions: Our data portrays BTLA as a molecule with the singular ability to provide both costimulatory and coinhibitory signals to activated CD8+ T cells, resulting in extended survival, improved tumor control, and the development of a functional recall response. Clin Cancer Res; 23(20); 6151-64. ©2017 AACR.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Animals , Cell Line, Tumor , Cytokines/metabolism , Cytotoxicity, Immunologic , Gene Expression , Heterografts , Humans , Immunologic Memory , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/metabolism , Melanoma/mortality , Melanoma/pathology , Melanoma, Experimental , Mice , Mice, Knockout , Models, Biological , Mutation , NF-kappa B , Neoplasm Metastasis , Prognosis , Signal Transduction , src-Family Kinases
5.
Cancer Discov ; 7(10): 1088-1097, 2017 10.
Article in English | MEDLINE | ID: mdl-28733428

ABSTRACT

Genomic intratumor heterogeneity (ITH) may be associated with postsurgical relapse of localized lung adenocarcinomas. Recently, mutations, through generation of neoantigens, were shown to alter tumor immunogenicity through T-cell responses. Here, we performed sequencing of the T-cell receptor (TCR) in 45 tumor regions from 11 localized lung adenocarcinomas and observed substantial intratumor differences in T-cell density and clonality with the majority of T-cell clones restricted to individual tumor regions. TCR ITH positively correlated with predicted neoantigen ITH, suggesting that spatial differences in the T-cell repertoire may be driven by distinct neoantigens in different tumor regions. Finally, a higher degree of TCR ITH was associated with an increased risk of postsurgical relapse and shorter disease-free survival, suggesting a potential clinical significance of T-cell repertoire heterogeneity.Significance: The present study provides insights into the ITH of the T-cell repertoire in localized lung adenocarcinomas and its potential biological and clinical impact. The results suggest that T-cell repertoire ITH may be tightly associated to genomic ITH and disease relapse. Cancer Discov; 7(10); 1088-97. ©2017 AACR.This article is highlighted in the In This Issue feature, p. 1047.


Subject(s)
Adenocarcinoma/surgery , Carcinoma, Non-Small-Cell Lung/surgery , Exome Sequencing/methods , Lung Neoplasms/surgery , Neoplasm Recurrence, Local/genetics , Receptors, Antigen, T-Cell/genetics , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Disease-Free Survival , Gene Expression Regulation, Neoplastic , Genetic Heterogeneity , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Mutation , Neoplasm Recurrence, Local/immunology
6.
Cancer Res ; 77(14): 3894-3907, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28566332

ABSTRACT

Oncolytic viruses selectively lyse tumor cells, disrupt immunosuppression within the tumor, and reactivate antitumor immunity, but they have yet to live up to their therapeutic potential. Immune checkpoint modulation has been efficacious in a variety of cancer with an immunogenic microenvironment, but is associated with toxicity due to nonspecific T-cell activation. Therefore, combining these two strategies would likely result in both effective and specific cancer therapy. To test the hypothesis, we first constructed oncolytic adenovirus Delta-24-RGDOX expressing the immune costimulator OX40 ligand (OX40L). Like its predecessor Delta-24-RGD, Delta-24-RGDOX induced immunogenic cell death and recruit lymphocytes to the tumor site. Compared with Delta-24-RGD, Delta-24-RGDOX exhibited superior tumor-specific activation of lymphocytes and proliferation of CD8+ T cells specific to tumor-associated antigens, resulting in cancer-specific immunity. Delta-24-RGDOX mediated more potent antiglioma activity in immunocompetent C57BL/6 but not immunodeficient athymic mice, leading to specific immune memory against the tumor. To further overcome the immune suppression mediated by programmed death-ligand 1 (PD-L1) expression on cancer cells accompanied with virotherapy, intratumoral injection of Delta-24-RGDOX and an anti-PD-L1 antibody showed synergistic inhibition of gliomas and significantly increased survival in mice. Our data demonstrate that combining an oncolytic virus with tumor-targeting immune checkpoint modulators elicits potent in situ autologous cancer vaccination, resulting in an efficacious, tumor-specific, and long-lasting therapeutic effect. Cancer Res; 77(14); 3894-907. ©2017 AACR.


Subject(s)
Cancer Vaccines/pharmacology , Neoplasms/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses/physiology , A549 Cells , Animals , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Glioma/immunology , Glioma/therapy , Glioma/virology , HEK293 Cells , Humans , Immunomodulation , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lung Neoplasms/virology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Melanoma, Experimental/virology , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasms/immunology , Neoplasms/virology , OX40 Ligand/biosynthesis , OX40 Ligand/genetics , OX40 Ligand/immunology , Oncolytic Viruses/genetics , Oncolytic Viruses/immunology
7.
Nat Med ; 23(5): 551-555, 2017 May.
Article in English | MEDLINE | ID: mdl-28346412

ABSTRACT

To date, anti-CTLA-4 (ipilimumab) or anti-PD-1 (nivolumab) monotherapy has not been demonstrated to be of substantial clinical benefit in patients with prostate cancer. To identify additional immune-inhibitory pathways in the prostate-tumor microenvironment, we evaluated untreated and ipilimumab-treated tumors from patients in a presurgical clinical trial. Levels of the PD-L1 and VISTA inhibitory molecules increased on independent subsets of macrophages in treated tumors. Our data suggest that VISTA represents another compensatory inhibitory pathway in prostate tumors after ipilimumab therapy.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , B7 Antigens/metabolism , B7-H1 Antigen/metabolism , Macrophages/metabolism , Prostatic Neoplasms/drug therapy , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adult , Aged , Animals , B7 Antigens/immunology , B7-H1 Antigen/immunology , Cell Line, Tumor , Disease Models, Animal , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Vitro Techniques , Ipilimumab , Macrophages/immunology , Male , Membrane Proteins/immunology , Membrane Proteins/metabolism , Mice , Middle Aged , Neoadjuvant Therapy , Prostatectomy , Prostatic Neoplasms/immunology , Prostatic Neoplasms/metabolism , T-Lymphocytes , Tissue Array Analysis
8.
Cancer Immunol Res ; 5(2): 100-105, 2017 02.
Article in English | MEDLINE | ID: mdl-28062513

ABSTRACT

We report here on a patient with metastatic melanoma who had extensive brain metastases. After being treated with the sequential combination of whole brain radiation therapy followed by the PD-1-inhibitory antibody, pembrolizumab, the patient had a durable complete response. Retrospective laboratory studies of T cells revealed that, after treatment with anti-PD-1 commenced, effector CD8+ T cells in the blood expanded and the ratio of CD8+:Treg T cells increased. A CD8+ T-cell clone present in the initial brain metastases was expanded in the blood after anti-PD-1 treatment, which suggested an antitumor role for this clone. Immunohistochemical analysis confirmed the presence of CD8+ T cells and low PD-L1 expression in the brain metastases before immunotherapy initiation. This sequence of therapy may provide an option for melanoma patients with unresponsive brain metastases. Cancer Immunol Res; 5(2); 100-5. ©2017 AACR.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Brain Neoplasms/secondary , Brain Neoplasms/therapy , Clonal Evolution , Cranial Irradiation , Melanoma/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Biomarkers , Brain/diagnostic imaging , Brain/pathology , Brain Neoplasms/diagnosis , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Clonal Evolution/genetics , Combined Modality Therapy , Cranial Irradiation/methods , Female , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Lymphocyte Count , Lymphocytes, Tumor-Infiltrating/immunology , Magnetic Resonance Imaging , Melanoma/genetics , Melanoma/metabolism , Middle Aged , Treatment Outcome
9.
Cancer Discov ; 6(8): 827-37, 2016 08.
Article in English | MEDLINE | ID: mdl-27301722

ABSTRACT

UNLABELLED: Immune checkpoint blockade represents a major breakthrough in cancer therapy; however, responses are not universal. Genomic and immune features in pretreatment tumor biopsies have been reported to correlate with response in patients with melanoma and other cancers, but robust biomarkers have not been identified. We studied a cohort of patients with metastatic melanoma initially treated with cytotoxic T-lymphocyte-associated antigen-4 (CTLA4) blockade (n = 53) followed by programmed death-1 (PD-1) blockade at progression (n = 46), and analyzed immune signatures in longitudinal tissue samples collected at multiple time points during therapy. In this study, we demonstrate that adaptive immune signatures in tumor biopsy samples obtained early during the course of treatment are highly predictive of response to immune checkpoint blockade and also demonstrate differential effects on the tumor microenvironment induced by CTLA4 and PD-1 blockade. Importantly, potential mechanisms of therapeutic resistance to immune checkpoint blockade were also identified. SIGNIFICANCE: These studies demonstrate that adaptive immune signatures in early on-treatment tumor biopsies are predictive of response to checkpoint blockade and yield insight into mechanisms of therapeutic resistance. These concepts have far-reaching implications in this age of precision medicine and should be explored in immune checkpoint blockade treatment across cancer types. Cancer Discov; 6(8); 827-37. ©2016 AACR.See related commentary by Teng et al., p. 818This article is highlighted in the In This Issue feature, p. 803.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers , Drug Resistance, Neoplasm , Immunomodulation/drug effects , Neoplasms/immunology , Neoplasms/metabolism , B7-H1 Antigen/antagonists & inhibitors , Biopsy , CTLA-4 Antigen/antagonists & inhibitors , Cluster Analysis , Gene Expression Profiling , Humans , Immunohistochemistry , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/drug therapy , Melanoma/immunology , Melanoma/metabolism , Neoplasms/diagnosis , Neoplasms/drug therapy , Prognosis , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
10.
Oncoimmunology ; 5(3): e1136044, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27141370

ABSTRACT

We have made major advances in the treatment of melanoma through the use of targeted therapy and immune checkpoint blockade; however, clinicians are posed with therapeutic dilemmas regarding timing and sequence of therapy. There is a growing appreciation of the impact of antitumor immune responses to these therapies, and we performed studies to test the hypothesis that clinical patterns and immune infiltrates differ at progression on these treatments. We observed rapid clinical progression kinetics in patients on targeted therapy compared to immune checkpoint blockade. To gain insight into possible immune mechanisms behind these differences, we performed deep immune profiling in tumors of patients on therapy. We demonstrated low CD8+ T-cell infiltrate on targeted therapy and high CD8+ T-cell infiltrate on immune checkpoint blockade at clinical progression. These data have important implications, and suggest that antitumor immune responses should be assessed when considering therapeutic options for patients with melanoma.

11.
PLoS One ; 9(5): e97407, 2014.
Article in English | MEDLINE | ID: mdl-24827739

ABSTRACT

BACKGROUND: Emerging evidence suggests anti-cancer immunity is involved in the therapeutic effect induced by oncolytic viruses. Here we investigate the effect of Delta-24-RGD oncolytic adenovirus on innate and adaptive anti-glioma immunity. DESIGN: Mouse GL261-glioma model was set up in immunocompetent C57BL/6 mouse for Delta-24-RGD treatment. The changes of the immune cell populations were analyzed by immunohistochemistry and flow cytometry. The anti-glioma immunity was evaluated with functional study of the splenocytes isolated from the mice. The efficacy of the virotherapy was assessed with animal survival analysis. The direct effect of the virus on the tumor-associated antigen presentation to CD8+ T cells was analyzed with an in vitro ovalbumin (OVA) modeling system. RESULTS: Delta-24-RGD induced cytotoxic effect in mouse glioma cells. Viral treatment in GL261-glioma bearing mice caused infiltration of innate and adaptive immune cells, instigating a Th1 immunity at the tumor site which resulted in specific anti-glioma immunity, shrunken tumor and prolonged animal survival. Importantly, viral infection and IFNγ increased the presentation of OVA antigen in OVA-expressing cells to CD8+ T-cell hybridoma B3Z cells, which is blocked by brefeldin A and proteasome inhibitors, indicating the activity is through the biosynthesis and proteasome pathway. CONCLUSIONS: Our results demonstrate that Delta-24-RGD induces anti-glioma immunity and offers the first evidence that viral infection directly enhances presentation of tumor-associated antigens to immune cells.


Subject(s)
Adaptive Immunity/immunology , Adenoviridae/immunology , Glioma/immunology , Immunity, Innate/immunology , Oncolytic Viruses/immunology , Adenoviridae Infections/immunology , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Disease Models, Animal , Hybridomas/immunology , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Oncolytic Virotherapy/methods
12.
Lancet Oncol ; 15(1): 69-77, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24332512

ABSTRACT

BACKGROUND: Endogenous or iatrogenic antitumour immune responses can improve the course of follicular lymphoma, but might be diminished by immune checkpoints in the tumour microenvironment. These checkpoints might include effects of programmed cell death 1 (PD1), a co-inhibitory receptor that impairs T-cell function and is highly expressed on intratumoral T cells. We did this phase 2 trial to investigate the activity of pidilizumab, a humanised anti-PD1 monoclonal antibody, with rituximab in patients with relapsed follicular lymphoma. METHODS: We did this open-label, non-randomised trial at the University of Texas MD Anderson Cancer Center (Houston, TX, USA). Adult (≥18 years) patients with rituximab-sensitive follicular lymphoma relapsing after one to four previous therapies were eligible. Pidilizumab was administered at 3 mg/kg intravenously every 4 weeks for four infusions, plus eight optional infusions every 4 weeks for patients with stable disease or better. Starting 17 days after the first infusion of pidilizumab, rituximab was given at 375 mg/m(2) intravenously weekly for 4 weeks. The primary endpoint was the proportion of patients who achieved an objective response (complete response plus partial response according to Revised Response Criteria for Malignant Lymphoma). Analysis was by intention to treat. This trial is registered with ClinicalTrials.gov, number NCT00904722. FINDINGS: We enrolled 32 patients between Jan 13, 2010, and Jan 20, 2012. Median follow-up was 15.4 months (IQR 10.1-21.0). The combination of pidilizumab and rituximab was well tolerated, with no autoimmune or treatment-related adverse events of grade 3 or 4. The most common adverse events of grade 1 were anaemia (14 patients) and fatigue (13 patients), and the most common adverse event of grade 2 was respiratory infection (five patients). Of the 29 patients evaluable for activity, 19 (66%) achieved an objective response: complete responses were noted in 15 (52%) patients and partial responses in four (14%). INTERPRETATION: The combination of pidilizumab plus rituximab is well tolerated and active in patients with relapsed follicular lymphoma. Our results suggest that immune checkpoint blockade is worthy of further study in follicular lymphoma. FUNDING: National Institutes of Health, Leukemia and Lymphoma Society, Cure Tech, and University of Texas MD Anderson Cancer Center.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lymphoma, Follicular/drug therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Adult , Aged , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Murine-Derived/administration & dosage , Female , Humans , Lymphoma, Follicular/mortality , Male , Middle Aged , Recurrence , Rituximab
13.
Biomark Cancer ; 5: 71-80, 2013.
Article in English | MEDLINE | ID: mdl-24324350

ABSTRACT

In order to develop a new tool for diagnosis of breast cancer based on autoantibodies against a panel of biomarkers, a clinical trial including blood samples from 507 subjects was conducted. All subjects showed a breast abnormality on exam or breast imaging and final biopsy pathology of either breast cancer patients or healthy controls. Using an enzyme-linked immunosorbent assay, the samples were tested for autoantibodies against a predetermined number of biomarkers in various models that were used to determine a diagnosis, which was compared to the clinical status. Our new assay achieved a sensitivity of 95.2% [CI = 92.8-96.8%] at a fixed specificity of 49.5%. Receiver-operator characteristic curve analysis showed an area under the curve of 80.1% [CI = 72.6-87.6%]. These results suggest that a blood test which is based on models comprising several autoantibodies to specific biomarkers may be a new and novel tool for improving the diagnostic evaluation of breast cancer.

14.
J Immunother ; 36(4): 276-86, 2013 May.
Article in English | MEDLINE | ID: mdl-23603862

ABSTRACT

Peptide vaccination against tumor-associated antigens remains one of the most common methods of immunization in cancer vaccine clinical trials. Although peptide vaccination has been reported to increase circulating antigen-specific T-cells, they have had limited clinical efficacy and there is a necessity to increase their capacity to generate strong antitumor responses. We sought to improve the clinical efficacy of peptide-based vaccines in cancer immunotherapy of metastatic melanoma using a LHRH agonist (leuprolide) as adjuvant. Seventy HLA-A*0201 stage IIb-IV melanoma patients were vaccinated with class I HLA-A*0201-restricted gp100209-2M peptide and stratified for HLA-DP4 restriction. HLA-DP4 patients were also vaccinated with class II HLA-DP4-restricted MAGE-3243-258 peptide. Patients from both groups were randomized to receive 2 doses of leuprolide or not. Here we report the increase in PBMC TREC levels at week 24 after peptide vaccination, which was independent of the leuprolide treatment. This change was mirrored by a small increase in the TREC-enriched CD8CD45RAROCD27CD103, but not the TREC-enriched CD4CD45RAROCD31 T-cell population. Serum concentration of 2 important factors for thymopoiesis was measured: insulin growth factor 1 (IGF-1) levels were not changed, whereas a moderate increase in IL-7 levels was noted in the sera of all patients 6 weeks after vaccination. Increased expression of CD127 (IL-7 receptor-α) at week 24, compared with baseline, was only seen in the CD8CD45RAROCD27CD103 T-cell population. Our results suggest that leuprolide has no effect on thymic output when used as peptide vaccine adjuvant, but IFA-based peptide vaccination may unexpectedly affect the thymus by increasing thymic output of new T cells.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Leuprolide/therapeutic use , Melanoma/immunology , Melanoma/therapy , Vaccines, Subunit/immunology , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Antineoplastic Agents, Hormonal/therapeutic use , Cancer Vaccines/administration & dosage , Female , Humans , Interleukin-7/blood , Lymphocyte Count , Male , Melanoma/pathology , Middle Aged , Neoplasm Proteins/chemistry , Neoplasm Proteins/immunology , Neoplasm Staging , Receptors, Antigen, T-Cell/metabolism , Treatment Outcome , Vaccines, Subunit/administration & dosage , Young Adult , gp100 Melanoma Antigen/chemistry , gp100 Melanoma Antigen/immunology
15.
J Investig Med ; 61(3): 564-8, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23360846

ABSTRACT

BACKGROUND: Several biomarkers are becoming available for the early detection of acute kidney injury (AKI), but few have been directly compared. OBJECTIVE: To compare urinary kidney injury molecule-1 (KIM-1), neutrophil gelatinase-associated lipocalin (NGAL), and N-acetyl glucosaminidase (NAG) against serum creatinine and renal histological score in the initiation, maintenance, and recovery phases of cisplatin (CP)-induced AKI. METHODS: Sprague-Dawley rats (300-350 g) were injected once through their tail veins with CP (CP group) at 5.5 mg/kg or with same volume of normal saline vehicle (Control group). Rats were euthanized at 2, 4, 6, 12, and 24 hours, and on days 2, 3, 6, and 10 (n = 12 in the CP group and n = 6 in the Control group at each time point), and urine, blood, and kidney samples were analyzed. RESULTS: A significant increase in serum creatinine was noted by day 3 in the CP group versus Control group [1.46 (0.12) vs 0.28 (0.03) mg/dL; mean (SE); P < 0.05]. The renal histology scores for brush border loss and tubular necrosis were significantly higher at 12 and 24 hours, respectively, in the CP group. Urinary kidney injury molecule-1 levels were significantly higher at 24 hours in the CP group than in the Control group [48.26 (13.13) vs 8.21 (3.31) pg/mg creatinine; P < 0.05] and remained elevated through day 10. Both urine NAG and NGAL levels were significantly higher by day 2 in the CP than in the Control group [NAG, 8.19 (0.82) vs 3.48 (0.40) pg/mg creatinine, P G 0.05; NGAL, 2911.80 (368.10) vs 1412.60 (250.20) pg/mg creatinine, P < 0.05]. Urinary NAG remained elevated for 6 days and NGAL for 3 days. CONCLUSIONS: Our study suggests a temporal hierarchy in the ability of certain urinary protein-based biomarkers to detect AKI after a well-defined tubular injury. Comparative analyses of urinary biomarkers are warranted in clinical settings such as patients receiving CP to discern the time course and pattern of expression.


Subject(s)
Acute Kidney Injury/blood , Acute Kidney Injury/urine , Acute-Phase Proteins/urine , Cell Adhesion Molecules/urine , Creatinine/blood , Hexosaminidases/urine , Kidney Tubules/pathology , Lipocalins/urine , Proto-Oncogene Proteins/urine , Animals , Biomarkers/blood , Biomarkers/urine , Cisplatin/toxicity , Disease Models, Animal , Kidney Tubules/drug effects , Kidney Tubules/metabolism , Lipocalin-2 , Rats , Rats, Sprague-Dawley , Time Factors
16.
J Immunol ; 189(11): 5476-84, 2012 Dec 01.
Article in English | MEDLINE | ID: mdl-23105141

ABSTRACT

PR1 is a HLA-A2-restricted peptide that has been targeted successfully in myeloid leukemia with immunotherapy. PR1 is derived from the neutrophil granule proteases proteinase 3 (P3) and neutrophil elastase (NE), which are both found in the tumor microenvironment. We recently showed that P3 and NE are taken up and cross-presented by normal and leukemia-derived APCs, and that NE is taken up by breast cancer cells. We now extend our findings to show that P3 and NE are taken up and cross-presented by human solid tumors. We further show that PR1 cross-presentation renders human breast cancer and melanoma cells susceptible to killing by PR1-specific CTLs (PR1-CTL) and the anti-PR1/HLA-A2 Ab 8F4. We also show PR1-CTL in peripheral blood from patients with breast cancer and melanoma. Together, our data identify cross-presentation as a novel mechanism through which cells that lack endogenous expression of an Ag become susceptible to therapies that target cross-presented Ags and suggest PR1 as a broadly expressed tumor Ag.


Subject(s)
Antigens, Neoplasm/immunology , Breast Neoplasms/therapy , Immunotherapy , Leukocyte Elastase/immunology , Melanoma/therapy , Myeloblastin/immunology , Skin Neoplasms/therapy , Antibodies/pharmacology , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Antigens, Neoplasm/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cross-Priming , Female , HLA-A2 Antigen/immunology , Humans , Leukocyte Elastase/chemistry , Melanoma/immunology , Melanoma/pathology , Molecular Targeted Therapy , Myeloblastin/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , Tumor Cells, Cultured
17.
Clin Cancer Res ; 18(17): 4733-42, 2012 Sep 01.
Article in English | MEDLINE | ID: mdl-22761473

ABSTRACT

PURPOSE: T-cell receptor (TCR) variable Vα and Vß gene diversity is a surrogate biomarker for the therapeutic potential of adoptive immunotherapy and cellular immunity. Therefore, creating a straightforward, rapid, sensitive, and reliable method to view the global changes of both TCRVα and Vß transcripts in heterogeneous populations of T cells is appealing. EXPERIMENTAL DESIGN: We designed a "direct TCR expression assay" (DTEA) using a panel of customized bar-coded probes that simultaneously detects and quantifies 45 Vα and 46 Vß transcripts in a nonenzymatic digital multiplexed assay from a small number of cells (10(4) cells) or as little as 100 ng of total RNA. RESULTS: We evaluated DTEA on total RNA samples of tumor-infiltrating lymphocytes and peripheral blood obtained from patients with melanoma after adoptive T-cell therapy. DTEA detected a similar spectrum of the dominant patterns of TCRVß gene usage as sequencing cloned TCRVß CDR3 regions. However, DTEA was rapid, achieved a level of sensitivity to identify rare T-cell populations, and simultaneously tracked the full array of Vα and Vß transcripts. CONCLUSIONS: DTEA can rapidly and sensitively track changes in TCRVα and Vß gene usages in T-cell pools following immune interventions, such as adoptive T-cell transfer, and may also be used to assess impact of vaccination or reconstitution of T-cell compartment after hematopoietic stem cell transplantation.


Subject(s)
Immunotherapy, Adoptive , Melanoma/blood , Receptors, Antigen, T-Cell, alpha-beta/genetics , Biomarkers, Pharmacological/blood , Humans , Lymphocytes, Tumor-Infiltrating/metabolism , Melanoma/immunology , Melanoma/therapy , RNA/blood , RNA/genetics , Receptors, Antigen, T-Cell, alpha-beta/blood
18.
Cancer ; 117(1): 39-47, 2011 Jan 01.
Article in English | MEDLINE | ID: mdl-20740500

ABSTRACT

BACKGROUND: Through this study, the authors sought to investigate the biologic and immunologic effects of preoperative trastuzumab in patients with ductal carcinoma in situ (DCIS) of the breast. METHODS: Patients with DCIS were enrolled in this open-label phase 2 trial and tested for HER2. Trastuzumab was given by intravenous infusion (8 mg/kg). The patients then had surgery 14 to 28 days after treatment. Tissue and peripheral blood samples were obtained before therapy and at the time of surgery to examine residual disease and immunologic response. RESULTS: Median age of the 69 enrolled patients was 53 years, mean mammographic size of the DCIS lesions was 5.2 ± 1.2 cm, and 24 patients (35%) were found to have HER2 overexpression/amplification (12 received trastuzumab and 12 untreated patients provided tissue for blinded, controlled biomarker analyses). No overt histologic evidence of response was noted. No significant change in mean pretherapy staining for Ki-67 (44.3 ± 3.4%) and cleaved caspase-3 (2.6 ± 0.8%) was noted when surgical specimens from drug-treated patient samples were compared with those not treated. Trastuzumab significantly augmented antibody-dependent cell mediated cytotoxicity (ADCC) in 100% of patients; this was demonstrated to be mediated through CD56+ degranulating natural killer cells (P < .01). One patient developed a significant anti-HER2 humoral CD4 T-cell response. CONCLUSIONS: Single-dose monotherapy with trastuzumab for patients with HER2-positive DCIS does not result in significant, clinically overt, histologic, antiproliferative, or apoptotic changes, but does result in the ability to mount ADCC mediated through natural killer cells and may also induce T-cell dependent humoral immunity. Further studies of trastuzumab for DCIS appear warranted. Cancer 2011. © 2010 American Cancer Society.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Carcinoma in Situ/drug therapy , Carcinoma, Ductal, Breast/drug therapy , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal, Humanized , Antibody-Dependent Cell Cytotoxicity , Breast Neoplasms/immunology , Carcinoma in Situ/immunology , Carcinoma in Situ/surgery , Carcinoma, Ductal, Breast/immunology , Female , Humans , Middle Aged , Preoperative Period , Receptor, ErbB-2/metabolism , Trastuzumab , Treatment Outcome
19.
J Immune Based Ther Vaccines ; 6: 1, 2008 Mar 06.
Article in English | MEDLINE | ID: mdl-18321390

ABSTRACT

BACKGROUND: Human monocyte-derived DC (mDC) loaded with peptides, protein, tumor cell lysates, or tumor cell RNA, are being tested as vaccines against multiple human malignancies and viral infection with great promise. One of the factors that has limited more widespread use of these vaccines is the need to generate mDC in large scale. Current methods for the large-scale cultivation of mDC in static culture vessels are labor- and time- intensive, and also require many culture vessels. Here, we describe a new method for the large-scale generation of human mDC from human PBMC from leukopheresis or buffy coat products using roller bottles, never attempted before for mDC generation. We have tested this technology using 850 cm2 roller bottles compared to conventional T-175 flat-bottom static culture flasks. METHODS: DC were generated from adherent human PBMC from buffy coats or leukopherisis products using GM-CSF and IL-4 in T-175 static flasks or 850 cm2 roller bottles. The cells were matured over two days, harvested and analyzed for cell yield and mature DC phenotype by flow cytometry, and then functionally analyzed for their ability to activate allogeneic T-cell or recall antigen peptide-specific T-cell responses. RESULTS: Monocytes were found to adhere inside roller bottles to the same extent as in static culture flasks. The phenotype and function of the mDC harvested after maturation from both type of culture systems were similar. The yield of mDC from input PBMC in the roller bottle system was similar as in the static flask system. However, each 850 cm2 roller bottle could be seeded with 4-5 times more input PBMC and could yield 4-5 times as many mDC per culture vessel than the static flasks as a result. CONCLUSION: Our results indicate that the roller bottle technology can generate similar numbers of mDC from adherent PBMC as traditional static flask methods, but with having to use fewer culture vessels. Thus, this may be a more practical method to generate mDC in large-scale cutting down on the amount of laboratory manipulations, and can save both time and labor costs.

SELECTION OF CITATIONS
SEARCH DETAIL
...