Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Cancer Immunol Res ; 11(12): 1589-1597, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37871333

ABSTRACT

Transgenic T-cell receptor (TCR) T cell-based adoptive cell therapies for solid tumors are associated with dramatic initial response rates, but there remain many instances of treatment failure and disease relapse. The association of infusion product cytokine profiles with clinical response has not been explored in the context of TCR T-cell therapy products. Single-cell antigen-dependent secretomic and proteomic analysis of preinfusion clinical TCR T-cell therapy products revealed that TNFα cytokine functionality of CD8+ T cells and phospho-STAT3 signaling in these cells were both associated with superior clinical responsiveness to therapy. By contrast, CD4+ T-helper 2 cell cytokine profiles were associated with inferior clinical responses. In parallel, preinfusion levels of IL15, Flt3-L, and CX3CL1 were all found to be associated with clinical response to therapy. These results have implications for the development of therapeutic biomarkers and identify potential targets for enrichment in the design of transgenic TCR T-cell therapies for solid tumors.


Subject(s)
Neoplasms , Tumor Necrosis Factor-alpha , Animals , Humans , Mice , Proteomics , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Cytokines , Animals, Genetically Modified , Cell- and Tissue-Based Therapy , Mice, Transgenic , STAT3 Transcription Factor
2.
Cancer Res Commun ; 3(8): 1628-1637, 2023 08.
Article in English | MEDLINE | ID: mdl-37621406

ABSTRACT

Purpose: Neoadjuvant combination immune checkpoint blockade and intralesional oncolytic virotherapy have the potential to activate antitumor responses in patients with breast cancer. Experimental Design: Eligibility for this pilot phase I trial included patients with localized HER2-negative breast cancer who received systemic nivolumab and ipilimumab and intratumor talimogene laherparepvec (T-VEC; NCT04185311). The primary objective was to evaluate the safety and adverse event profile of immunotherapy combined with T-VEC in patients with localized, HER2-negative breast cancer. Results: Six patients were enrolled, 4 having relapses after prior neoadjuvant chemotherapy and 2 who were previously untreated. Toxicities included 1 patient having grade 3 hypotension and type 1 diabetes mellitus, 3 patients with hypothyroidism, and all patients having constitutional symptoms known to be associated with the administration of T-VEC. One patient had a pathologic complete response, 3 patients had pathologic partial responses, 1 showed no significant response, and 1 had disease progression. Biopsies demonstrated increased immune cell infiltration in samples from patients who responded to therapy. Conclusions: This triple immunotherapy regimen provided responses in patients with advanced or relapsed HER2-negative breast cancer, at the expense of long-term toxicities. Significance: Systemic immune checkpoint blockade with a programmed death receptor 1 and a CTL antigen-4 blocking antibody, combined with intralesional oncolytic virotherapy, is a chemotherapy-free combination aimed at inducing an antitumor immune response locally and systemic immunity.


Subject(s)
Breast Neoplasms , Melanoma , Oncolytic Virotherapy , Female , Humans , Breast Neoplasms/therapy , Immune Checkpoint Inhibitors , Ipilimumab/therapeutic use , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Nivolumab/therapeutic use , Oncolytic Virotherapy/adverse effects , Pilot Projects
3.
Front Immunol ; 14: 1156746, 2023.
Article in English | MEDLINE | ID: mdl-37325672

ABSTRACT

Immune checkpoint inhibitors (ICIs) targeting programmed cell death-1 (PD-1) and its corresponding ligand PD-L1 are being increasingly used for a wide variety of cancers, including refractory sarcomas. Autoimmune hepatitis is a known side effect of ICIs, and is typically managed with broad, non-specific immunosuppression. Here, we report a case of severe autoimmune hepatitis occurring after anti-PD-1 therapy with nivolumab in a patient with osteosarcoma. Following prolonged unsuccessful treatment with intravenous immunoglobulin, steroids, everolimus, tacrolimus, mycophenolate, and anti-thymoglobulin, the patient was eventually treated with the anti-CD25 monoclonal antibody basiliximab. This resulted in prompt, sustained resolution of her hepatitis without significant side effects. Our case demonstrates that basiliximab may be an effective therapy for steroid-refractory severe ICI-associated hepatitis.


Subject(s)
Hepatitis, Autoimmune , Immune Checkpoint Inhibitors , Humans , Female , Immune Checkpoint Inhibitors/adverse effects , Basiliximab , Hepatitis, Autoimmune/diagnosis , Hepatitis, Autoimmune/drug therapy , Hepatitis, Autoimmune/etiology , Nivolumab/adverse effects , Antibodies, Monoclonal/adverse effects
4.
Brief Bioinform ; 24(4)2023 07 20.
Article in English | MEDLINE | ID: mdl-37291798

ABSTRACT

The ability to identify and track T-cell receptor (TCR) sequences from patient samples is becoming central to the field of cancer research and immunotherapy. Tracking genetically engineered T cells expressing TCRs that target specific tumor antigens is important to determine the persistence of these cells and quantify tumor responses. The available high-throughput method to profile TCR repertoires is generally referred to as TCR sequencing (TCR-Seq). However, the available TCR-Seq data are limited compared with RNA sequencing (RNA-Seq). In this paper, we have benchmarked the ability of RNA-Seq-based methods to profile TCR repertoires by examining 19 bulk RNA-Seq samples across 4 cancer cohorts including both T-cell-rich and T-cell-poor tissue types. We have performed a comprehensive evaluation of the existing RNA-Seq-based repertoire profiling methods using targeted TCR-Seq as the gold standard. We also highlighted scenarios under which the RNA-Seq approach is suitable and can provide comparable accuracy to the TCR-Seq approach. Our results show that RNA-Seq-based methods are able to effectively capture the clonotypes and estimate the diversity of TCR repertoires, as well as provide relative frequencies of clonotypes in T-cell-rich tissues and low-diversity repertoires. However, RNA-Seq-based TCR profiling methods have limited power in T-cell-poor tissues, especially in highly diverse repertoires of T-cell-poor tissues. The results of our benchmarking provide an additional appealing argument to incorporate RNA-Seq into the immune repertoire screening of cancer patients as it offers broader knowledge into the transcriptomic changes that exceed the limited information provided by TCR-Seq.


Subject(s)
Benchmarking , Neoplasms , Humans , Receptors, Antigen, T-Cell/genetics , T-Lymphocytes , Neoplasms/genetics , Sequence Analysis, RNA
5.
Cancer Immunol Res ; 10(12): 1433-1440, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36259217

ABSTRACT

A major complication of chimeric antigen receptor (CAR) T-cell therapy is immune effector cell-associated neurotoxicity syndrome (ICANS), which presents as aphasia, confusion, weakness, somnolence, seizures, and coma. This is similar to the neurologic manifestations of hypophosphatemia, which can result from sudden increases in metabolic demand for phosphorylated intermediates (e.g., refeeding syndrome and sepsis). Given these similarities, we investigated whether CAR T-cell effector metabolic activity is associated with increased extracellular phosphate consumption and a possible association between hypophosphatemia and ICANS. In vitro 4-1BB and CD28 CD19-targeted CAR T-cell effector activity was found to be associated with increased consumption of media phosphorus, which was temporally associated with increased single-cell effector secretomic activity and increased phosphorus-dependent metabolic demand of the CAR T cells. A clinical cohort of 77 patients treated with CD19-targeted CAR T-cell therapy demonstrated a significant anticorrelation between serum phosphorus and ICANS incidence and severity, with earlier onset of hypophosphatemia after CAR T-cell infusion more likely to result in neurotoxicity. These results imply phosphorous level monitoring could alert to the development of ICANS in clinical scenarios. See related Spotlight by Tobin et al., p. 1422.


Subject(s)
Hypophosphatemia , Neurotoxicity Syndromes , Humans , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell , Antigens, CD19 , Neurotoxicity Syndromes/etiology , Hypophosphatemia/chemically induced , Phosphorus
6.
Cell Rep Med ; 2(10): 100419, 2021 10 19.
Article in English | MEDLINE | ID: mdl-34755133

ABSTRACT

Immune checkpoint blockade using PD-1 inhibition is an effective approach for treating a wide variety of cancer subtypes. While lower gastrointestinal (GI) side effects are more common, upper gastrointestinal adverse events are rarely reported. Here, we present a case of nivolumab-associated autoimmune gastritis. To elucidate the immunology underlying this condition, we leverage multiplexed ion beam imaging by time-of-flight (MIBI-TOF) to identify the presence and proportion of infiltrating immune cells from a single section of biopsy specimen. Using MIBI-TOF, we analyze formalin-fixed, paraffin-embedded human gastric tissue with 28 labels simultaneously. Our analyses reveal a gastritis characterized by severe mucosal injury, interferon gamma (IFN-γ)-producing gastric epithelial cells, and mixed inflammation that includes CD8 and CD4 T cell infiltrates with reduced expression of granzyme B and FOXP3, respectively. Here, we provide a comprehensive multiplexed histopathological mapping of gastric tissue, which identifies IFN-γ-producing epithelial cells as possible contributors to the nivolumab-associated gastritis.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Gastritis/chemically induced , Immune Checkpoint Inhibitors/adverse effects , Interferon-gamma/immunology , Nivolumab/adverse effects , Antineoplastic Agents, Immunological/administration & dosage , Biopsy , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Gastric Mucosa/drug effects , Gastric Mucosa/immunology , Gastric Mucosa/pathology , Gastritis/genetics , Gastritis/immunology , Gastritis/pathology , Gene Expression , Granzymes/genetics , Granzymes/immunology , Humans , Immune Checkpoint Inhibitors/administration & dosage , Interferon-gamma/genetics , Middle Aged , Nivolumab/administration & dosage , Stomach/drug effects , Stomach/immunology , Stomach/pathology , Uterine Neoplasms/drug therapy , Uterine Neoplasms/genetics , Uterine Neoplasms/immunology , Uterine Neoplasms/pathology
7.
Cancer Discov ; 10(11): 1645-1653, 2020 11.
Article in English | MEDLINE | ID: mdl-32699033

ABSTRACT

Transgenic T-cell receptor (TCR) adoptive cell therapies recognizing tumor antigens are associated with robust initial response rates, but frequent disease relapse. This usually occurs in the setting of poor long-term persistence of cells expressing the transgenic TCR, generated using murine stem cell virus (MSCV) γ-retroviral vectors. Analysis of clinical transgenic adoptive cell therapy products in vivo revealed that despite strong persistence of the transgenic TCR DNA sequence over time, its expression was profoundly decreased over time at the RNA and protein levels. Patients with the greatest degrees of expression suppression displayed significant increases in DNA methylation over time within the MSCV promoter region, as well as progressive increases in DNA methylation within the entire MSCV vector over time. These increases in vector methylation occurred independently of its integration site within the host genomes. These results have significant implications for the design of future viral vector gene-engineered adoptive cell transfer therapies. SIGNIFICANCE: Cellular immunotherapies' reliance on retroviral vectors encoding foreign genetic material can be vulnerable to progressive acquisition of DNA methylation and subsequent epigenetic suppression of the transgenic product in TCR adoptive cell therapy. This must be considered in the design of future generations of cellular immunotherapies for cancer.This article is highlighted in the In This Issue feature, p. 1611.


Subject(s)
Epigenesis, Genetic/genetics , Genetic Vectors/genetics , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/metabolism , Transduction, Genetic/methods , Humans
9.
Nat Cancer ; 1(1): 46-58, 2020.
Article in English | MEDLINE | ID: mdl-34368780

ABSTRACT

Lack of tumor infiltration by immune cells is the main mechanism of primary resistance to programmed cell death protein 1 (PD-1) blockade therapies for cancer. It has been postulated that cancer cell-intrinsic mechanisms may actively exclude T cells from tumors, suggesting that the finding of actionable molecules that could be inhibited to increase T cell infiltration may synergize with checkpoint inhibitor immunotherapy. Here, we show that p21-activated kinase 4 (PAK4) is enriched in non-responding tumor biopsies with low T cell and dendritic cell infiltration. In mouse models, genetic deletion of PAK4 increased T cell infiltration and reversed resistance to PD-1 blockade in a CD8 T cell-dependent manner. Furthermore, combination of anti-PD-1 with the PAK4 inhibitor KPT-9274 improved anti-tumor response compared with anti-PD-1 alone. Therefore, high PAK4 expression is correlated with low T cell and dendritic cell infiltration and a lack of response to PD-1 blockade, which could be reversed with PAK4 inhibition.


Subject(s)
Immune Checkpoint Inhibitors , Immunotherapy , Neoplasms , Programmed Cell Death 1 Receptor , p21-Activated Kinases , Animals , CD8-Positive T-Lymphocytes , Mice , Neoplasms/drug therapy , p21-Activated Kinases/genetics
10.
Clin Cancer Res ; 25(7): 2096-2108, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30573690

ABSTRACT

PURPOSE: Transgenic adoptive cell therapy (ACT) targeting the tumor antigen NY-ESO-1 can be effective for the treatment of sarcoma and melanoma. Preclinical models have shown that this therapy can be improved with the addition of dendritic cell (DC) vaccination and immune checkpoint blockade. We studied the safety, feasibility, and antitumor efficacy of transgenic ACT with DC vaccination, with and without CTLA-4 blockade with ipilimumab. PATIENTS AND METHODS: Freshly prepared autologous NY-ESO-1-specific T-cell receptor (TCR) transgenic lymphocytes were adoptively transferred together with NY-ESO-1 peptide-pulsed DC vaccination in HLA-A2.1-positive subjects alone (ESO, NCT02070406) or with ipilimumab (INY, NCT01697527) in patients with advanced sarcoma or melanoma. RESULTS: Six patients were enrolled in the ESO cohort, and four were enrolled in the INY cohort. Four out of six patients treated per ESO (66%), and two out of four patients treated per INY (50%) displayed evidence of tumor regression. Peripheral blood reconstitution with NY-ESO-1-specific T cells peaked within 2 weeks of ACT, indicating rapid in vivo expansion. Tracking of transgenic T cells to the tumor sites was demonstrated in on-treatment biopsies via TCR sequencing. Multiparametric mass cytometry of transgenic cells demonstrated shifting of transgenic cells from memory phenotypes to more terminally differentiated effector phenotypes over time. CONCLUSIONS: ACT of fresh NY-ESO-1 transgenic T cells prepared via a short ex vivo protocol and given with DC vaccination, with or without ipilimumab, is feasible and results in transient antitumor activity, with no apparent clinical benefit of the addition of ipilimumab. Improvements are needed to maintain tumor responses.


Subject(s)
Adoptive Transfer , Antineoplastic Agents, Immunological/pharmacology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Ipilimumab/pharmacology , Neoplasms/immunology , Neoplasms/therapy , Adoptive Transfer/methods , Adult , Animals , CTLA-4 Antigen/antagonists & inhibitors , Cell Line, Tumor , Combined Modality Therapy , Dendritic Cells/metabolism , Female , Gene Knock-In Techniques , Humans , Immunotherapy , Lymphocytes/immunology , Lymphocytes/metabolism , Male , Mice , Middle Aged , Molecular Targeted Therapy , Neoplasms/pathology , Phenotype , Pilot Projects , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Young Adult
11.
Clin Cancer Res ; 24(21): 5347-5356, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29898988

ABSTRACT

Purpose: We have shown that the aged microenvironment increases melanoma metastasis, and decreases response to targeted therapy, and here we queried response to anti-PD1.Experimental Design: We analyzed the relationship between age, response to anti-PD1, and prior therapy in 538 patients. We used mouse models of melanoma, to analyze the intratumoral immune microenvironment in young versus aged mice and confirmed our findings in human melanoma biopsies.Results: Patients over the age of 60 responded more efficiently to anti-PD-1, and likelihood of response to anti-PD-1 increased with age, even when we controlled for prior MAPKi therapy. Placing genetically identical tumors in aged mice (52 weeks) significantly increased their response to anti-PD1 as compared with the same tumors in young mice (8 weeks). These data suggest that this increased response in aged patients occurs even in the absence of a more complex mutational landscape. Next, we found that young mice had a significantly higher population of regulatory T cells (Tregs), skewing the CD8+:Treg ratio. FOXP3 staining of human melanoma biopsies revealed similar increases in Tregs in young patients. Depletion of Tregs using anti-CD25 increased the response to anti-PD1 in young mice.Conclusions: While there are obvious limitations to our study, including our inability to conduct a meta-analysis due to a lack of available data, and our inability to control for mutational burden, there is a remarkable consistency in these data from over 500 patients across 8 different institutes worldwide. These results stress the importance of considering age as a factor for immunotherapy response. Clin Cancer Res; 24(21); 5347-56. ©2018 AACR See related commentary by Pawelec, p. 5193.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Immunomodulation/drug effects , Programmed Cell Death 1 Receptor/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Age Factors , Animals , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Disease Models, Animal , Female , Humans , Melanoma/drug therapy , Melanoma/immunology , Melanoma/metabolism , Melanoma/pathology , Mice , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
12.
Lancet Oncol ; 19(3): 310-322, 2018 03.
Article in English | MEDLINE | ID: mdl-29449192

ABSTRACT

BACKGROUND: Obesity has been linked to increased mortality in several cancer types; however, the relation between obesity and survival outcomes in metastatic melanoma is unknown. The aim of this study was to examine the association between body-mass index (BMI) and progression-free survival or overall survival in patients with metastatic melanoma who received targeted therapy, immunotherapy, or chemotherapy. METHODS: This retrospective study analysed independent cohorts of patients with metastatic melanoma assigned to treatment with targeted therapy, immunotherapy, or chemotherapy in randomised clinical trials and one retrospective study of patients treated with immunotherapy. Patients were classified according to BMI, following the WHO definitions, as underweight, normal, overweight, or obese. Patients without BMI and underweight patients were excluded. The primary outcomes were the associations between BMI and progression-free survival or overall survival, stratified by treatment type and sex. We did multivariable analyses in the independent cohorts, and combined adjusted hazard ratios in a mixed-effects meta-analysis to provide a precise estimate of the association between BMI and survival outcomes; heterogeneity was assessed with meta-regression analyses. Analyses were done on the predefined intention-to-treat population in the randomised controlled trials and on all patients included in the retrospective study. FINDINGS: The six cohorts consisted of a total of 2046 patients with metastatic melanoma treated with targeted therapy, immunotherapy, or chemotherapy between Aug 8, 2006, and Jan 15, 2016. 1918 patients were included in the analysis. Two cohorts containing patients from randomised controlled trials treated with targeted therapy (dabrafenib plus trametinib [n=599] and vemurafenib plus cobimetinib [n=240]), two cohorts containing patients treated with immunotherapy (one randomised controlled trial of ipilimumab plus dacarbazine [n=207] and a retrospective cohort treated with pembrolizumab, nivolumab, or atezolizumab [n=331]), and two cohorts containing patients treated with chemotherapy (two randomised controlled trials of dacarbazine [n=320 and n=221]) were classified according to BMI as normal (694 [36%] patients), overweight (711 [37%]), or obese (513 [27%]). In the pooled analysis, obesity, compared with normal BMI, was associated with improved survival in patients with metastatic melanoma (average adjusted hazard ratio [HR] 0·77 [95% CI 0·66-0·90] for progression-free survival and 0·74 [0·58-0·95] for overall survival). The survival benefit associated with obesity was restricted to patients treated with targeted therapy (HR 0·72 [0·57-0·91] for progression-free survival and 0·60 [0·45-0·79] for overall survival) and immunotherapy (HR 0·75 [0·56-1·00] and 0·64 [0·47-0·86]). No associations were observed with chemotherapy (HR 0·87 [0·65-1·17, pinteraction=0·61] for progression-free survival and 1·03 [0·80-1·34, pinteraction=0·01] for overall survival). The association of BMI with overall survival for patients treated with targeted and immune therapies differed by sex, with inverse associations in men (HR 0·53 [0·40-0·70]), but no associations observed in women (HR 0·85 [0·61-1·18, pinteraction=0·03]). INTERPRETATION: Our results suggest that in patients with metastatic melanoma, obesity is associated with improved progression-free survival and overall survival compared with those outcomes in patients with normal BMI, and that this association is mainly seen in male patients treated with targeted or immune therapy. These results have implications for the design of future clinical trials for patients with metastatic melanoma and the magnitude of the benefit found supports further investigation of the underlying mechanism of these associations. FUNDING: ASCO/CCF Young Investigator Award, ASCO/CCF Career Development Award, MD Anderson Cancer Center (MDACC) Melanoma Moonshot Program, MDACC Melanoma SPORE, and the Dr Miriam and Sheldon G Adelson Medical Research Foundation.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Body Mass Index , Melanoma/drug therapy , Molecular Targeted Therapy , Obesity/epidemiology , Skin Neoplasms/drug therapy , Adolescent , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Female , Humans , Male , Melanoma/immunology , Melanoma/mortality , Melanoma/secondary , Middle Aged , Molecular Targeted Therapy/adverse effects , Molecular Targeted Therapy/mortality , Obesity/diagnosis , Obesity/mortality , Progression-Free Survival , Protective Factors , Randomized Controlled Trials as Topic , Retrospective Studies , Risk Assessment , Risk Factors , Skin Neoplasms/immunology , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Time Factors , Treatment Outcome , Young Adult
13.
J Immunother ; 41(5): 248-259, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29470191

ABSTRACT

Adoptive cell therapy (ACT) consisting of genetically engineered T cells expressing tumor antigen-specific T-cell receptors displays robust initial antitumor activity, followed by loss of T-cell activity/persistence and frequent disease relapse. We characterized baseline and longitudinal T-cell phenotype variations resulting from different manufacturing and administration protocols in patients who received ACT. Patients with melanoma who enrolled in the F5-MART-1 clinical trial (NCT00910650) received infusions of MART-1 T-cell receptors transgenic T cells with MART-1 peptide-pulsed dendritic cell vaccination. Patients were divided into cohorts based on several manufacturing changes in the generation and administration of the transgenic T cells: decreasing ex vivo stimulation/expansion time, increased cell dose, and receiving fresh instead of cryopreserved cells. T-cell phenotypes were analyzed by flow cytometry at baseline and longitudinally in peripheral blood. Transgenic T cells with shorter ex vivo culture/expansion periods displayed significantly increased expression of markers associated with less differentiated naive/memory populations, as well as significantly decreased expression of the inhibitory receptor programmed death 1 (PD1). Patients receiving fresh infusions of transgenic cells demonstrated expansion of central memory T cells and delayed acquisition of PD1 expression compared with patients who received cryopreserved products. Freshly infused transgenic T cells showed persistence and expansion of naive and memory T-cell populations and delayed acquisition of PD1 expression, which correlated with this cohort's superior persistence of transgenic cells and response to dendritic cell vaccines. These results may be useful in designing future ACT protocols.


Subject(s)
Cancer Vaccines/immunology , Immunotherapy, Adoptive/methods , Melanoma/therapy , Skin Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Adult , Aged , Cells, Cultured , Cryopreservation , Female , Humans , Immunologic Memory , Immunophenotyping , Lymphocyte Activation , MART-1 Antigen/metabolism , Male , Melanoma/immunology , Middle Aged , Phenotype , Skin Neoplasms/immunology
14.
Cancer J ; 24(1): 47-53, 2018.
Article in English | MEDLINE | ID: mdl-29360728

ABSTRACT

Cancer immunotherapy utilizing blockade of the PD-1/PD-L1 checkpoint has revolutionized the treatment of a wide variety of malignancies, leading to durable therapeutic responses not typically seen with traditional cytotoxic anticancer agents. However, these therapies are ineffective in a significant percentage of patients, and some initial responders eventually develop resistance to these therapies with relapsed disease. The mechanisms leading to both primary and acquired resistance to PD-1/PD-L1 inhibition are varied and can be both multifactorial and overlapping in an individual patient. As the mechanisms of resistance to PD-1/PD-L1 blockade continue to be further characterized, new strategies are being developed to prevent or reverse resistance to therapy, leading to improved patient outcomes.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , B7-H1 Antigen/antagonists & inhibitors , Drug Resistance, Neoplasm/drug effects , Neoplasms/therapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , Humans , Immunotherapy/methods , Neoplasms/immunology
15.
Cancer Immunol Res ; 5(2): 118-126, 2017 02.
Article in English | MEDLINE | ID: mdl-28039162

ABSTRACT

Tumors expressing programmed death ligand 1 (PD-L1) interact with the corresponding negative-signal generating immune receptor on the surface of CD8 T cells, PD-1, thereby suppressing antitumor activity. Therapeutics blocking this interaction have shown promise in various cancers by restoring functional antitumor T-cell activity. We explored the degree of PD-L1, PD-1, and CD8 expression in a retrospective analysis of 29 clinical synovial sarcoma samples. Quantitative immunohistochemistry and multiplex immunofluorescence were used to determine relative quantification of CD8+ and PD-1+ T cells and PD-L1 expression within the intratumor area and the interface between the tumor and the surrounding nontumor tissue (i.e., invasive margin), and colocalization of these factors, respectively. PD-L1, PD-1, and CD8 cell densities in the tumor-invasive margins were significantly higher in the metastatic tumors than the primary tumors (P < 0.01), and PD-L1, PD-1, and CD8 cell densities were all significantly positively correlated with one other (P < 0.0001). PD-1 cell density in the tumor-invasive margin was significantly associated with worse progression-free survival. Multiplex immunofluorescence demonstrated coexpression of PD-1 and CD8 on lymphocytes within the invasive margin, as well as relative proximity between PD-1+ CD8 cells and PD-L1+ tumor cells. Our results provide a preclinical rationale for screening of patients with synovial sarcoma for the colocalization of CD8, PD-1, and PD-L1, which may be a marker for response to PD-1 blockade therapy. Cancer Immunol Res; 5(2); 118-26. ©2016 AACR.


Subject(s)
B7-H1 Antigen/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Programmed Cell Death 1 Receptor/genetics , Sarcoma, Synovial/genetics , Sarcoma, Synovial/immunology , B7-H1 Antigen/metabolism , Gene Expression , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Neoplasm Metastasis , Neoplasm Staging , Programmed Cell Death 1 Receptor/metabolism , Retrospective Studies , Sarcoma, Synovial/diagnosis , Sarcoma, Synovial/mortality
16.
Pediatr Res ; 79(3): 371-7, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26595537

ABSTRACT

Progress has slowed substantially in improving survival rates for pediatric sarcomas, particularly in refractory and metastatic disease. Significant progress has been made in the field of tumor vaccines for such malignancies, which target established tumor antigens. While tumor vaccines have demonstrated safety and improved survival rates, they are inadequate in mediating the regression of established tumor masses and metastases. Programmed cell death ligand 1 (PDL1) is a cell-surface protein induced in a number of adult malignancies. By acting on the corresponding T-cell receptor PD1, PDL1 is able to suppress cytotoxic T-cell-mediated tumor responses. Recent therapeutics blocking this interaction have shown promise in various adult cancers by restoring a functional T-cell response and by directing this response toward an activated, rather than regulatory, T-cell phenotype. We shall discuss the current state of tumor vaccines targeting pediatric sarcomas, review PD1-PDL1 interactions and current therapies targeting these interactions in adult malignancies, and discuss recent studies in which tumor vaccines, combined with PDL1 blockades, produced superior tumor regression compared with the vaccine alone. These studies provide a compelling case for investigation of PDL1 expression and its inhibition in pediatric sarcomas, while continuing to utilize tumor vaccines in tandem to achieve superior clinical outcomes.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Cancer Vaccines/chemistry , Immunotherapy/methods , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Sarcoma/therapy , Skin Neoplasms/therapy , Adult , B7-H1 Antigen/physiology , CD8-Positive T-Lymphocytes/cytology , Cancer Vaccines/immunology , Child , Clinical Trials as Topic , Gene Expression Regulation, Neoplastic , Humans , Inflammation , Neoplasm Metastasis , Phenotype , Programmed Cell Death 1 Receptor/physiology , Receptors, Antigen, T-Cell/immunology , Sarcoma/immunology , Skin Neoplasms/immunology , T-Lymphocytes/cytology
17.
Indian J Pediatr ; 80(7): 570-5, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23263974

ABSTRACT

OBJECTIVE: To describe the clinical features, treatment and prognosis of acquired thrombotic thrombocytopenic purpura (TTP) in children based on a single institution experience. METHODS: This study is a retrospective review of all 12 children with TTP seen at New York Medical College- Westchester Medical Center during a period of 15 y from 1993 to 2008. RESULTS: There were 7 females and 5 males with acquired TTP, with a median age of 13 (range, 6-17); and no cases of congenital TTP. The classic pentad of TTP (microangiopathic hemolytic anemia, thrombocytopenia, neurologic symptoms, renal dysfunction and fever) was seen in only three patients. Nine had renal involvement; eight had neurologic symptoms; and four had fever. All 12 patients had thrombocytopenia, anemia, and elevated LDH. Nine had idiopathic TTP. Three patients had one of the following underlying disorders: systemic lupus erythematosus, mixed connective tissue disorder, and aplastic anemia (post-bone marrow transplant on cyclosporine). ADAMTS13 level was decreased in 7 of 8 patients studied. Eight of 10 patients achieved remission with plasmapheresis alone. Two needed additional treatment before achieving remission. Two had one or more relapses, requiring immunosupressive treatment with vincrisine, prednisone, or rituximab. The patient with aplastic anemia died of pulmonary hypertension 5 y after bone marrow transplantation. All other 11 patients are alive and free of TTP for a median follow-up of 12 mo (range, 3-72 mo). CONCLUSIONS: Acquired pediatric TTP responds well to plasmapheresis. However, many patients do require additional treatment because of refractoriness to plasmapheresis or relapse. The clinical features, response to treatment, and relapse rate of pediatric TTP appear similar to those of adult TTP.


Subject(s)
Purpura, Thrombotic Thrombocytopenic/diagnosis , Purpura, Thrombotic Thrombocytopenic/therapy , Adolescent , Child , Diagnosis, Differential , Female , Humans , Male , New York/epidemiology , Prognosis , Purpura, Thrombotic Thrombocytopenic/epidemiology , Retrospective Studies , Treatment Outcome
18.
J Cell Biochem ; 113(6): 1998-2008, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22253131

ABSTRACT

Arachidonate 5-lipoxygenase (ALOX5) expression and activity has been implicated in tumor pathogenesis, yet its role in papillary thyroid carcinoma (PTC) has not been characterized. ALOX5 protein and mRNA were upregulated in PTC compared to matched, normal thyroid tissue, and ALOX5 expression correlated with invasive tumor histopathology. Evidence suggests that PTC invasion is mediated through the induction of matrix metalloproteinases (MMPs) that can degrade and remodel the extracellular matrix (ECM). A correlation between MMP-9 and ALOX5 protein expression was established by immunohistochemical analysis of PTC and normal thyroid tissues using a tissue array. Transfection of ALOX5 into a PTC cell line (BCPAP) increased MMP-9 secretion and cell invasion across an ECM barrier. The ALOX5 product, 5(S)-hydroxyeicosatetraenoic acid also increased MMP-9 protein expression by BCPAP in a dose-dependent manner. Inhibitors of MMP-9 and ALOX5 reversed ALOX5-enhanced invasion. Here we describe a new role for ALOX5 as a mediator of invasion via MMP-9 induction; this ALOX5/MMP9 pathway represents a new avenue in the search for functional biomarkers and/or potential therapeutic targets for aggressive PTC.


Subject(s)
Arachidonate 5-Lipoxygenase/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Matrix Metalloproteinase 9/metabolism , Neoplasm Invasiveness , Thyroid Neoplasms/metabolism , Adult , Aged , Arachidonate 5-Lipoxygenase/genetics , Biomarkers, Tumor , Carcinoma , Carcinoma, Papillary , Cell Cycle , Cell Line, Tumor , Cell Movement , Cell Proliferation , Extracellular Matrix/metabolism , Female , Humans , Hydroxyeicosatetraenoic Acids/pharmacology , Lipoxygenase Inhibitors/pharmacology , Male , Matrix Metalloproteinase Inhibitors , Middle Aged , RNA, Messenger/genetics , RNA, Messenger/metabolism , Thyroid Cancer, Papillary , Thyroid Neoplasms/pathology , Young Adult
20.
Pediatr Hematol Oncol ; 28(2): 167-72, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20469972

ABSTRACT

Our patient first developed thrombotic thrombocytopenic purpura (TTP) at age 10 years with an initial platelet count of 10,000/microL. She achieved remission with plasmapheresis (PE), but suffered 2 relapses in the next 2 years, each approximately 1 year from PE, with ADAMTS13 levels of <5%. Early in her third remission, with vincristine (weekly x 4 doses) and prednisone (for 2 weeks) her ADAMTS13 increased to 99% in 24 weeks, but decreased to <4% in the next 38 weeks. After 4 weekly doses of rituximab (375 mg/m(2)), her ADAMTS13 level reached 101% in 9 weeks and has remained consistently above 97% on bimonthly monitoring for more than a year. She remains in continuous clinical and hematologic remission with an ADAMTS13 level of 108% at 60 weeks from rituximab therapy and 124 weeks from her second relapse. This case report suggests that monitoring ADAMTS13 level at regular intervals in recurrent TTP may help us identify patients at risk for further relapse; and such a relapse may be prevented, or at least delayed with timely rituximab therapy, thus reducing morbidity from relapsed TTP and its treatment.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasm Recurrence, Local/prevention & control , Purpura, Thrombotic Thrombocytopenic/drug therapy , ADAM Proteins/metabolism , ADAMTS13 Protein , Child , Chronic Disease , Female , Humans , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Purpura, Thrombotic Thrombocytopenic/metabolism , Purpura, Thrombotic Thrombocytopenic/pathology , Remission Induction , Rituximab , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...