Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 23
1.
Thyroid ; 32(2): 153-163, 2022 02.
Article En | MEDLINE | ID: mdl-34641722

Background: Combination therapy with lenvatinib plus programmed death-1 (PD-1) immune checkpoint blockades (ICBs) is under investigation in many solid tumors, including thyroid cancer. Lenvatinib is known to reduce angiogenesis and may overturn the immunosuppressive effects of vascular endothelial growth factor in the tumor microenvironment. Previous studies investigating the effects of VEGF receptor inhibition on the immune response were performed in rapidly growing tumor models where immune equilibrium is not established before treatment. We hypothesize that physiologically relevant preclinical models are necessary to define mechanisms of resistance to immune-targeted combination therapies. Methods: We utilized the TPO-CreER/BrafV600E/wt/Trp53Δex2-10/Δex2-10 inducible transgenic model of advanced thyroid cancer to investigate lenvatinib treatment in the context of an anti-PD-1 ICB. Following tumor establishment, 3.5 months postinduction, mice were treated with high- (10 mg/kg) or low-dose (2 mg/kg) lenvatinib, anti-PD-1, or combination of lenvatinib with anti-PD-1. Tumor volume and lung metastases were assessed in each group. Immune infiltrate was characterized by flow cytometry and immunohistochemistry, and TCRß sequencing was performed to further investigate the T cell response. Results: Both low- and high-dose lenvatinib reduced tumor volume, while anti-PD-1 had no effect, alone or in combination. Although both low- and high-dose lenvatinib reduced vascular density, low-dose lenvatinib was superior in controlling tumor size. Lung metastases and survival were not improved with therapy despite the effects of lenvatinib on primary tumor size. Low-dose lenvatinib treatment led to a subtle reduction in the dominant Ly6G+CD11b+ myeloid cell population and was associated with increased CD4+ T cell infiltrate and enrichment in 4-1BB+ and granzyme B+ CD4+ T cells and FoxP3+ regulatory T cells. Polyclonal T cell expansion was evident in the majority of mice, suggesting that a tumor-specific T cell response was generated. Conclusions: The effects of lenvatinib on the immune response were most pronounced in mice treated with low-dose lenvatinib, suggesting that dose should be considered in clinical application. While the immune-modulating potential of lenvatinib is encouraging, alterations in the immune milieu and T cell activation status were insufficient to sustain durable tumor regression, even with added anti-PD-1. Additional studies are necessary to develop more effective combination approaches in low-mutation burden tumors, such as thyroid cancer.


Drug Resistance , Immune Checkpoint Inhibitors , Phenylurea Compounds/administration & dosage , Quinolines/administration & dosage , Thyroid Neoplasms/pathology , Vascular Endothelial Growth Factor A/administration & dosage , Animals , Antineoplastic Agents/therapeutic use , Drug Therapy, Combination , Humans , Mice , Models, Animal , Thyroid Neoplasms/chemically induced
2.
Nat Rev Endocrinol ; 16(11): 629-641, 2020 11.
Article En | MEDLINE | ID: mdl-32839578

In the past decade, the field of cancer immunotherapy has been revolutionized by immune checkpoint blockade (ICB) technologies. Success across a broad spectrum of cancers has led to a paradigm shift in therapy for patients with advanced cancer. Early data are now accumulating in progressive thyroid cancers treated with single-agent ICB therapies and combination approaches that incorporate ICB technologies. This Review discusses our current knowledge of the immune response in thyroid cancers, the latest and ongoing immune-based approaches, and the future of immunotherapies in thyroid cancer. Physiologically relevant preclinical mouse models and human correlative research studies will inform development of the next stage of immune-based therapies for patients with advanced thyroid cancer.


Immunotherapy/methods , Thyroid Neoplasms/therapy , Animals , Combined Modality Therapy , Humans , Immune Checkpoint Inhibitors , Immunotherapy/trends , Mice , Thyroid Neoplasms/genetics , Thyroid Neoplasms/immunology
3.
Thyroid ; 30(9): 1263-1279, 2020 09.
Article En | MEDLINE | ID: mdl-32242507

Background: Despite advances in targeted kinase inhibitor development for patients with medullary thyroid cancer (MTC), most patients develop resistance and would benefit from alternative approaches. Immune-based therapies are now considered for patients with progressive MTC. This study is the first comprehensive assessment of the immune milieu, immune-suppressive molecules, and potential tumor antigens in patients with MTC. Methods: Primary and/or regionally metastatic tumor tissues from 46 patients with MTC were screened for immune infiltrates by using standard immunohistochemistry (IHC) and further analyzed by multispectral imaging for T cell and myeloid markers. RNASeq expression profiling was performed in parallel. RNASeq, targeted sequencing, and IHC techniques identified cancer-associated mutations and MTC-enriched proteins. Results: Organized immune infiltration was observed in 49% and 90% of primary and metastatic tumors, respectively. CD8+ cells were the dominant T cell subtype in most samples, while CD163+ macrophages were most frequent among myeloid infiltrates. PD-1+ T cells were evident in 24% of patients. Myeloid subsets were largely major histocompatibility complex II (MHCII-), suggesting a dysfunctional phenotype. Expression profiling confirmed enrichment in T cell, macrophage, and inflammatory profiles in a subset of samples. PD-L1 was expressed at low levels in a small subset of patients, while the immune regulatory molecules CD155 and CD47 were broadly expressed. Calcitonin, GRP, HIST1H4E, NOMO3, and NPIPA2 were highly and specifically expressed in MTC. Mutations in tumor suppressors, PTEN and p53, and mismatch repair genes, MSH2 and MSH6, may be relevant to disease progression and antigenicity. Conclusions: This study suggests that MTC is a more immunologically active tumor that has been previously reported. Patients with advanced MTC should be screened for targetable antigens and immune checkpoints to determine their eligibility for current clinical trials. Additional studies are necessary to fully characterize the antigenic potential of MTC and may encourage the development of adoptive T cells therapies for this rare tumor.


Antigens, Neoplasm/metabolism , Carcinoma, Neuroendocrine/immunology , RNA-Seq , Thyroid Neoplasms/immunology , Calcitonin/metabolism , Carcinoma, Neuroendocrine/metabolism , Clinical Trials as Topic , DNA Mutational Analysis , Disease Progression , Gene Expression Profiling , Humans , Immune System , Immunohistochemistry , Leukocytes/metabolism , Macrophages/metabolism , MutS Homolog 2 Protein/metabolism , Mutation , Receptors, Virus/metabolism , Thyroid Neoplasms/metabolism , Thyroiditis, Autoimmune/metabolism , United States
4.
J Clin Endocrinol Metab ; 105(1)2020 01 01.
Article En | MEDLINE | ID: mdl-31513709

CONTEXT: Although the development of immune checkpoint inhibitors has transformed treatment strategies of several human malignancies, research models to study immunotherapy in adrenocortical carcinoma (ACC) are lacking. OBJECTIVE: To explore the effect of anti-PD1 immunotherapy on the alteration of the immune milieu in ACC in a newly generated preclinical model and correlate with the response of the matched patient. DESIGN, SETTING, AND INTERVENTION: To characterize the CU-ACC2-M2B patient-derived xenograft in a humanized mouse model, evaluate the effect of a PD-1 inhibitor therapy, and compare it with the CU-ACC2 patient with metastatic disease. RESULTS: Characterization of the CU-ACC2-humanized cord blood-BALB/c-Rag2nullIl2rγnullSirpaNOD model confirmed ACC origin and match with the original human tumor. Treatment of the mice with pembrolizumab demonstrated significant tumor growth inhibition (60%) compared with controls, which correlated with increased tumor infiltrating lymphocyte activity, with an increase of human CD8+ T cells (P < 0.05), HLA-DR+ T cells (P < 0.05) as well as Granzyme B+ CD8+ T cells (<0.001). In parallel, treatment of the CU-ACC2 patient, who had progressive disease, demonstrated a partial response with 79% to 100% reduction in the size of target lesions, and no new sites of metastasis. Pretreatment analysis of the patient's metastatic liver lesion demonstrated abundant intratumoral CD8+ T cells by immunohistochemistry. CONCLUSIONS: Our study reports the first humanized ACC patient-derived xenograft mouse model, which may be useful to define mechanisms and biomarkers of response and resistance to immune-based therapies, to ultimately provide more personalized care for patients with ACC.


Adrenal Cortex Neoplasms/drug therapy , Adrenocortical Carcinoma/drug therapy , Antibodies, Monoclonal, Humanized/pharmacology , Disease Models, Animal , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/immunology , Adrenal Cortex Neoplasms/immunology , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/immunology , Adrenocortical Carcinoma/pathology , Animals , Antineoplastic Agents, Immunological/pharmacology , Apoptosis , Cell Proliferation , Female , Humans , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Mice, Nude , Programmed Cell Death 1 Receptor/immunology , Tumor Cells, Cultured , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
5.
Clin Cancer Res ; 24(13): 3059-3068, 2018 07 01.
Article En | MEDLINE | ID: mdl-29615459

Purpose: To define the genetic landscape of advanced differentiated and anaplastic thyroid cancer (ATC) and identify genetic alterations of potential diagnostic, prognostic, and therapeutic significance.Experimental Design: The genetic profiles of 583 advanced differentiated and 196 ATCs generated with targeted next-generation sequencing cancer-associated gene panels MSK-IMPACT and FoundationOne were analyzed.Results: ATC had more genetic alterations per tumor, and pediatric papillary thyroid cancer had fewer genetic alterations per tumor when compared with other thyroid cancer types. DNA mismatch repair deficit and activity of APOBEC cytidine deaminases were identified as mechanisms associated with high mutational burden in a subset of differentiated thyroid cancers and ATCs. Copy number losses and mutations of CDKN2A and CDKN2B, amplification of CCNE1, amplification of receptor tyrosine kinase genes KDR, KIT, and PDGFRA, amplification of immune evasion genes CD274, PDCD1LG2, and JAK2, and activating point mutations in small GTPase RAC1 were associated with ATC. An association of KDR, KIT, and PDGFRA amplification with the sensitivity of thyroid cancer cells to lenvatinib was shown in vitro Three genetically distinct types of ATCs are proposed.Conclusions: This large-scale analysis describes genetic alterations in a cohort of thyroid cancers enriched in advanced cases. Many novel genetic events previously not seen in thyroid cancer were found. Genetic alterations associated with anaplastic transformation were identified. An updated schematic of thyroid cancer genetic evolution is proposed. Clin Cancer Res; 24(13); 3059-68. ©2018 AACR.


Biomarkers, Tumor , Genetic Variation , Thyroid Carcinoma, Anaplastic/diagnosis , Thyroid Carcinoma, Anaplastic/genetics , Algorithms , Computational Biology/methods , DNA Mismatch Repair , Gene Frequency , Genetic Testing , Humans , Mutation , Neoplasm Grading , Neoplasm Staging , Oncogenes , Pharmacogenomic Variants , Prognosis , Promoter Regions, Genetic , Thyroid Carcinoma, Anaplastic/drug therapy , Tumor Suppressor Proteins/genetics
8.
Lancet Diabetes Endocrinol ; 5(6): 469-481, 2017 06.
Article En | MEDLINE | ID: mdl-27773653

Inflammation has long been associated with the thyroid and with thyroid cancers, raising seminal questions about the role of the immune system in the pathogenesis of advanced thyroid cancers. With a growing understanding of dynamic tumour-immune cell interactions and the mechanisms by which tumour cells evade antitumour immunity, the field of cancer immunotherapy has been revolutionised. In this Review, we provide evidence to support the presence of an antitumour immune response in advanced thyroid cancers linked to cytotoxic T cells and NK cells. This antitumour response, however, is likely blunted by the presence of immunosuppressive pathways within the microenvironment, facilitated by tumour-associated macrophages or increased expression of negative regulators of cytotoxic T-cell function. Current and future efforts to incorporate immune-based therapies into existing tumour cell or endothelial-derived therapies-eg, with kinase inhibitors targeting tumour-associated macrophages or antibodies blocking negative regulators on T cells-could provide improved and durable responses for patients with disease that is otherwise refractory to treatment.


Immunotherapy/trends , Thyroid Neoplasms/therapy , Humans , Immunomodulation , Killer Cells, Natural/physiology , Macrophages/physiology , T-Lymphocytes/physiology , Thyroid Neoplasms/immunology
9.
J Clin Endocrinol Metab ; 101(7): 2863-73, 2016 07.
Article En | MEDLINE | ID: mdl-27045886

CONTEXT: Five to 10% of patients with differentiated thyroid cancers (DTC) develop invasive and/or distant metastatic disease that is marginally improved with standard therapies. Prognosis is poor for patients with anaplastic thyroid cancer, with a median survival of 3-5 months. We suggest that a paradigm shift is necessary in the treatment of advanced cases. OBJECTIVE: We hypothesized that a T-cell response is generated in advanced thyroid cancer and may be a viable therapeutic target. DESIGN: Primary DTCs were analyzed by quantitative RT-PCR (n = 92) for expression of CD3, CD8, forkhead box (Fox)-P3, programmed death (PD)-1, PD-1 ligand-1, and PD-1 ligand-2 and biopsied for cellular analysis by flow cytometry (n = 11). Advanced pT4 cases (n = 22) and metastases (n = 5) were analyzed by immunohistochemistry. SETTING: The study was conducted at the University of Colorado Hospital. PATIENTS: Thyroid cancer patients undergoing thyroidectomy or completion surgery for advanced disease between 2002 and 2013 participated in the study. INTERVENTION: There were no interventions. MAIN OUTCOME MEASURE: Immune markers were analyzed for association with disease severity. RESULTS: Immune markers were commonly expressed at the RNA level. PD-L1 was higher (P = .0443) in patients with nodal metastases. FoxP3(+) (P < .0001), PD-1(+)CD8(+) (P = .0058), and PD-1(+)CD4(+) (P = .0104) T cells were enriched in DTC biopsies. CD8(+) and FoxP3(+) T cells were detected by immunohistochemistry in all pT4 tumors and a subset of metastases. PD-1(+) lymphocytes were found in 50% of DTCs. PD-L1 was expressed by tumor and associated leukocytes in 13 of 22 cases, and expression was more diffuse in anaplastic thyroid cancer (P = .0373). BRAF(V600E) mutation was associated with higher frequencies of tumor-associated lymphocytes (P = .0095) but not PD-L1 expression. CONCLUSIONS: PD-1 checkpoint blockades may have therapeutic efficacy in patients with aggressive forms of thyroid cancer.


Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/genetics , Thyroid Carcinoma, Anaplastic/genetics , Thyroid Carcinoma, Anaplastic/immunology , Thyroid Neoplasms/genetics , Thyroid Neoplasms/immunology , Adult , Aged , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cell Cycle Checkpoints/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Invasiveness , Prognosis , Programmed Cell Death 1 Receptor/physiology , Signal Transduction/genetics , Thyroid Carcinoma, Anaplastic/diagnosis , Thyroid Carcinoma, Anaplastic/pathology , Thyroid Neoplasms/diagnosis , Thyroid Neoplasms/pathology
10.
Cancer Immunol Res ; 3(6): 620-30, 2015 Jun.
Article En | MEDLINE | ID: mdl-25701326

Regional metastatic differentiated thyroid cancer (mDTC) provides a unique model in which to study the tumor-immune interface. These lymph node metastases persist for years, generally without progression to distant metastases. Although the immune system likely impedes disease progression, it is unsuccessful in eliminating disease. Our previous studies revealed that programmed death-1 (PD-1)(+) T cells were enriched in tumor-involved lymph nodes (TILN). Tumor-associated leukocytes and tumor cells were collected from grossly involved lymph nodes from 12 patients to further characterize the phenotype and functional potential of mDTC-associated PD-1(+) T cells. PD-1(+)CD4(+) and PD-1(+)CD8(+) T cells were enriched in 8 of 12 TILN samples. PD-1(+) T cells coexpressed Tim-3 and CD69 and failed to downregulate CD27. CD8(+) T cells, but not CD4(+) T cells, from these samples were variably deficient in their ability to produce effector cytokines when compared with control TILNs that lacked resident PD-1(+) T cells. PD-1(+)CD8(+) T cells were capable of exocytosis but lacked intracellular perforin. Surprisingly, T-cell proliferative capacity was largely maintained in all samples. Thus, although PD-1 expression by mDTC-associated CD8(+) T cells was associated with dysfunction, exhaustion was not complete. Notably, molecular markers of exhaustion did not translate to dysfunction in all samples or in CD4(+) T cells. Regulatory T cells (Treg), PD-L1, and galectin-9 were commonly found in mDTC and likely contributed to the initiation of T-cell exhaustion and disease progression. Therapies that release the effects of PD-1 and Tim-3 and reduce the suppressive effects of Tregs may encourage tumor elimination in patients with mDTC.


CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Membrane Proteins/metabolism , Programmed Cell Death 1 Receptor/metabolism , Thyroid Neoplasms/immunology , Thyroid Neoplasms/metabolism , Adult , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Female , Hepatitis A Virus Cellular Receptor 2 , Humans , Immunophenotyping , Lymphocyte Count , Male , Middle Aged , Neoplasm Grading , Neoplasm Metastasis , Neoplasm Staging , Phenotype , Recurrence , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thyroid Neoplasms/pathology , Young Adult
11.
Thyroid ; 23(5): 529-42, 2013 May.
Article En | MEDLINE | ID: mdl-23317509

BACKGROUND: With our growing understanding of the immune system and mechanisms employed by tumors to evade destruction, the field of cancer immunotherapy has been revitalized. Concurrent inflammation has long been associated with follicular cell-derived thyroid cancer (FDTC). In the last decade, much research has focused on characterizing the tumor-associated immune response in patients with FDTC. SUMMARY: Mast cells, natural killer cells, macrophages, dendritic cells, B cells, and T cells have been identified within FDTC-associated immune infiltrate. Collectively, these findings suggest that the immune response to FDTC is compromised and may even promote tumor progression. A more thorough characterization of the tumor-associated immune response in FDTC may lead to the development of immune-based adjuvant therapies for patients with aggressive disease. CONCLUSIONS: Immune-based therapies could provide essential alternatives to patients that cannot be treated surgically, those with recurrent or persistent lymph node metastases, and those with anaplastic thyroid cancer.


Adenocarcinoma, Follicular/therapy , Immunomodulation , Thyroid Neoplasms/therapy , Adaptive Immunity , Adenocarcinoma, Follicular/immunology , Adenocarcinoma, Follicular/secondary , Animals , Humans , Immunity, Cellular , Lymphatic Metastasis/immunology , Thyroid Neoplasms/immunology
12.
Immunol Res ; 55(1-3): 22-33, 2013 Mar.
Article En | MEDLINE | ID: mdl-22941561

There will be over half a million cancer-related deaths in the United States in 2012, with lung cancer being the leader followed by prostate in men and breast in women. There is estimated to be more than one and a half million new cases of cancer in 2012, making the development of effective therapies a high priority. As tumor immunologists, we are interested in the development of immunotherapies because the immune response offers exquisite specificity and the potential to target tumor cells without harming normal cells. In this review, we highlight the current advances in the field of immunotherapy and the current work being completed by laboratories at University of Colorado School of Medicine in multiple malignancies, including breast cancer, lung cancer, melanoma, thyroid cancer, and glioblastoma. This work focuses on augmenting the anti-tumor response of CD8 T cells in the blood, lymph nodes, and tumors of patients, determining biomarkers for patients who are more likely to respond to immunotherapy, and identifying additional anti-tumor and immunosuppressive cells that influence the overall response to tumors. These collaborative efforts will identify mechanisms to improve immune function, which may elucidate therapeutic targets for clinical trials to improve patient health and survival.


CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Colorado , Humans , Immunotherapy , Neoplasms/therapy , Universities
13.
J Clin Endocrinol Metab ; 97(6): E934-43, 2012 Jun.
Article En | MEDLINE | ID: mdl-22466343

CONTEXT: Recurrent metastatic lymph node (LN) disease is common in patients with papillary thyroid cancer (PTC). Novel prognostic markers may be helpful in guiding a therapeutic approach. Our previous studies revealed that immune suppression is evident in PTC and associated with more severe disease. OBJECTIVE: To characterize the immune response to metastatic PTC, we assessed CD4(+) T cell polarization in LN. In addition, we investigated the role of programmed death-1 (PD-1) and T cell exhaustion. DESIGN: Uninvolved (UILN) and tumor-involved lymph nodes (TILN) were sampled ex vivo by fine-needle biopsy. T cell subsets were identified by flow cytometry. In parallel, archived TILN specimens were characterized by immunofluorescence. SETTING: The study was conducted at the University of Colorado Hospital. PATIENTS: Data were collected on 94 LN from 19 patients with PTC undergoing neck dissection. MAIN OUTCOME: T cell subset frequencies were compared in UILN and TILN and assessed for correlation with recurrent disease and extranodal invasion. RESULTS: Regulatory CD4(+) T cells (Treg) were enriched in TILN compared with UILN and further elevated in TILN from patients with recurrent disease. PD-1(+) T cells were present at high frequency in TILN and markedly enriched in TILN that showed evidence of extranodal invasion. In TILN, Treg frequency correlated with PD-1(+) T cell frequencies. Although PD-1(+) T cells produced interferon-γ, they failed to fully down-regulate CD27 and were not actively proliferating. CONCLUSIONS: Increased Treg and PD-1(+) T cell frequencies in LN may be indicative of aggressive recurrent PTC. Future prospective studies are necessary to determine the prognostic and therapeutic value of these findings in PTC.


Biomarkers, Tumor/metabolism , Carcinoma, Papillary/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes, Regulatory/metabolism , Thyroid Neoplasms/metabolism , Biopsy, Fine-Needle , Carcinoma, Papillary/mortality , Carcinoma, Papillary/secondary , Carcinoma, Papillary/surgery , Flow Cytometry , Humans , Immunophenotyping , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphatic Metastasis , Lymphocyte Activation/physiology , Neoplasm Invasiveness/pathology , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/mortality , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Prognosis , T-Lymphocytes, Regulatory/pathology , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology , Thyroid Neoplasms/surgery
14.
J Clin Endocrinol Metab ; 95(5): 2325-33, 2010 May.
Article En | MEDLINE | ID: mdl-20207826

CONTEXT: Ten to 30% of patients with papillary thyroid cancer (PTC) develop recurrent disease and may benefit from innovative adjuvant therapies. Immune-based therapies are under investigation to treat many types of cancer. The role of the immune system in PTC is poorly understood. OBJECTIVE: We investigated whether tumor-associated lymphocytes (TAL), in the absence of background thyroiditis (LT), contribute to disease severity. We hypothesized that the type of lymphocytes associated with PTC would correlate with parameters of disease. DESIGN: This retrospective study analyzed archived PTC samples for the presence of TAL and/or LT. A group of patients with TAL was evaluated for lymphocyte subsets by immunohistofluorescence. PATIENTS AND SETTING: One hundred PTC patients were analyzed for LT and TAL, and 10 PTC patients with TAL were assessed for lymphocyte subsets at University of Colorado Hospital. MAIN OUTCOME: We assessed correlations between disease and the presence of TAL, LT, and lymphocyte subset frequency. RESULTS: Patients with TAL exhibited higher disease stage and increased incidence of invasion and lymph node metastasis compared with patients without lymphocytes or with LT. CD4(+) T cell frequency correlated with tumor size (r = 0.742; P = 0.017). FoxP3(+) regulatory T cell (Treg) frequency correlated with lymph node metastases (r = 0.858; P = 0.002), and CD8 to Treg ratio correlated inversely with tumor size (r = -0.804; P = 0.007). CONCLUSIONS: TAL and high Treg frequency in primary thyroid tumors correlates with more aggressive disease. Future prospective studies may identify Treg frequency as a predictive factor in PTC, and the suppressive effects of Treg should be considered in the design of immune-based therapies.


Carcinoma, Papillary/immunology , Forkhead Transcription Factors/immunology , T-Lymphocytes, Regulatory/immunology , Thyroid Neoplasms/immunology , Adult , Aged , Aged, 80 and over , Antibody Specificity , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Carcinoma, Papillary/pathology , Female , Humans , Lymphatic Metastasis/immunology , Lymphatic Metastasis/pathology , Lymphocyte Subsets/immunology , Lymphocyte Subsets/pathology , Male , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Retrospective Studies , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology , Thyroid Gland/immunology , Thyroid Gland/pathology , Thyroid Neoplasms/pathology , Young Adult
15.
J Clin Endocrinol Metab ; 94(6): 2199-203, 2009 Jun.
Article En | MEDLINE | ID: mdl-19293266

CONTEXT: Focal adhesion kinase (FAK) and Src are overexpressed and activated in many cancers and have been associated with tumor progression. The role of the Src-FAK complex has not been characterized in papillary and anaplastic thyroid cancer (PTC and ATC). OBJECTIVE: The goal of this study was to determine the role of Src and FAK in the growth and invasion of PTC and ATC. DESIGN: PTC and ATC cells were treated with the oral Src inhibitor, AZD0530, to determine the consequences of Src inhibition using growth and invasion assays. FAK and phospho-FAK levels were analyzed in cell lines as well as in PTC tumor samples. RESULTS: AZD0530 treatment inhibited the growth and invasion in four of five thyroid cancer cell lines, and inhibition did not correlate with basal levels of phospho-Src. Instead, we show for the first time that FAK, a critical substrate and effector of Src, is phosphorylated at tyrosine residue 861 (pY861) in PTC and ATC cells, and high levels of phospho-FAK correlate with AZD0530 sensitivity. We further showed that pY861-FAK phosphorylation is Src-dependent. Sensitivity to AZD0530 was confirmed using a preclinical three-dimensional culture model. Phospho-ERK1/2 was not affected by AZD0530, indicating that Src signaling does not require MAPK. Finally, FAK and pY861-FAK were expressed in 10 of 10 and five of 10 PTC tumors, respectively. CONCLUSIONS: Inhibition of the Src-FAK complex represents a promising therapeutic strategy for patients with advanced thyroid cancer, and phospho-FAK represents a potential biomarker for response.


Benzodioxoles/therapeutic use , Carcinoma, Papillary/drug therapy , Carcinoma/drug therapy , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/antagonists & inhibitors , Quinazolines/therapeutic use , Thyroid Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Benzodioxoles/pharmacology , Carcinoma/metabolism , Carcinoma, Papillary/metabolism , Cell Culture Techniques , Cell Proliferation/drug effects , Drug Delivery Systems/methods , Drug Evaluation, Preclinical , Enzyme Activation/drug effects , Focal Adhesion Protein-Tyrosine Kinases/antagonists & inhibitors , Humans , Phosphorylation/drug effects , Protein Binding/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins pp60(c-src)/metabolism , Quinazolines/pharmacology , Thyroid Neoplasms/metabolism , Tumor Cells, Cultured
16.
J Immunol ; 182(4): 1892-900, 2009 Feb 15.
Article En | MEDLINE | ID: mdl-19201842

Successful application of gammadelta T cells in adoptive cell therapies depends upon our ability to maintain these cells in vivo. Using an adoptive transfer model to study lymphopenia-induced homeostatic expansion, we show that CD8(+) and NK1.1(+) gammadelta T cell subsets are differentially regulated. While CD8(+) gammadelta T cells have an early and sustained advantage following transfer into TCRbeta(-/-)/delta(-/-) mice, NK1.1(+) gammadelta T cells proliferate slowly and are maintained at low numbers. The advantage of the CD8(+) subset could not be explained by increased bcl-2 or cytokine receptor expression but did correlate with Vgamma4(+) and Vdelta5(+) expression. Despite the role of CD8 in MHC class I recognition by alphabeta T cells, beta(2)-microglobulin (beta(2)m)-associated MHC class I molecules were not required for CD8(+) gammadelta T cell homeostatic expansion. Surprisingly, all gammadelta T cells, including the CD8(+) subset, exhibited enhanced proliferation following adoptive transfer into Rag1(-/-)/beta(2)m(-/-) compared with Rag1(-/-) recipients. This effect was most notable for the NK1.1(+) subset, which expresses high levels of NKG2A/CD94 and Ly49. Although expression of these inhibitory receptors correlated with poor homeostatic expansion in the presence of beta(2)m, gammadelta T cell homeostatic proliferation in TCRbeta(-/-)/delta(-/-) mice was not altered in the presence of Ly49C/I- and NKG2-blocking Abs. While the mechanism by which beta(2)m negatively regulates gammadelta T cell homeostasis remains to be determined, this observation is unique to gammadelta T cells and confirms that multiple mechanisms are in place to maintain strict regulation of both the size and the composition of the gammadelta T cell pool.


Homeostasis/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , beta 2-Microglobulin/immunology , Adoptive Transfer , Animals , Female , Flow Cytometry , Gene Expression Regulation/immunology , Male , Mice , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Interleukin/biosynthesis , T-Lymphocyte Subsets/metabolism , beta 2-Microglobulin/metabolism
17.
J Immunol ; 179(8): 5576-83, 2007 Oct 15.
Article En | MEDLINE | ID: mdl-17911645

Murine gammadelta T cell subsets, defined by their Vgamma chain usage, have been shown in various disease models to have distinct functional roles. In this study, we examined the responses of the two main peripheral gammadelta T cell subsets, Vgamma1(+) and Vgamma4(+) cells, during collagen-induced arthritis (CIA), a mouse model that shares many hallmarks with human rheumatoid arthritis. We found that whereas both subsets increased in number, only the Vgamma4(+) cells became activated. Surprisingly, these Vgamma4(+) cells appeared to be Ag selected, based on preferential Vgamma4/Vdelta4 pairing and very limited TCR junctions. Furthermore, in both the draining lymph node and the joints, the vast majority of the Vgamma4/Vdelta4(+) cells produced IL-17, a cytokine that appears to be key in the development of CIA. In fact, the number of IL-17-producing Vgamma4(+) gammadelta T cells in the draining lymph nodes was found to be equivalent to the number of CD4(+)alphabeta(+) Th-17 cells. When mice were depleted of Vgamma4(+) cells, clinical disease scores were significantly reduced and the incidence of disease was lowered. A decrease in total IgG and IgG2a anti-collagen Abs was also seen. These results suggest that Vgamma4/Vdelta4(+) gammadelta T cells exacerbate CIA through their production of IL-17.


Arthritis, Experimental/immunology , Arthritis, Experimental/metabolism , Collagen Type II/immunology , Interleukin-17/biosynthesis , Receptors, Antigen, T-Cell, gamma-delta/biosynthesis , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Animals , Arthritis, Experimental/pathology , Cattle , Collagen Type II/administration & dosage , Epitopes, T-Lymphocyte/immunology , Interleukin-17/physiology , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Male , Mice , Mice, Inbred DBA , T-Lymphocyte Subsets/pathology
18.
J Immunol ; 179(5): 2961-8, 2007 Sep 01.
Article En | MEDLINE | ID: mdl-17709511

Mice sensitized and challenged with OVA were used to investigate the role of innate T cells in the development of allergic airway hyperresponsiveness (AHR). AHR, but not eosinophilic airway inflammation, was induced in T cell-deficient mice by small numbers of cotransferred gammadelta T cells and invariant NKT cells, whereas either cell type alone was not effective. Only Vgamma1+Vdelta5+ gammadelta T cells enhanced AHR. Surprisingly, OVA-specific alphabeta T cells were not required, revealing a pathway of AHR development mediated entirely by innate T cells. The data suggest that lymphocytic synergism, which is key to the Ag-specific adaptive immune response, is also intrinsic to T cell-dependent innate responses.


Killer Cells, Natural/immunology , Receptors, Antigen, T-Cell, gamma-delta/analysis , Respiratory Hypersensitivity/immunology , T-Lymphocytes/immunology , Animals , Antigens/immunology , Mice , Mice, Mutant Strains , Ovalbumin/immunology , Receptors, Antigen, T-Cell, alpha-beta/analysis , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, gamma-delta/genetics
19.
J Clin Immunol ; 27(2): 133-44, 2007 Mar.
Article En | MEDLINE | ID: mdl-17333410

Lymphocytes expressing gammadelta T cell receptors (TCR) constitute an entire system of functionally specialized subsets that have been implicated in the regulation of immune responses, including responses to pathogens and allergens, and in tissue repair. The gammadelta TCRs share structural features with adaptive receptors and peripheral selection of gammadelta T cells occurs. Nevertheless, their specificities may be primarily directed at self-determinants, and the responses of gammadelta T cells exhibit innate characteristics. Continuous cross talk between gammadelta T cells and myeloid cells is evident in histological studies and in in vitro co-culture experiments, suggesting that gammadelta T cells play a functional role as an integral component of the innate immune system.


Immunity, Innate , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Animals , Humans , Lymphocyte Activation/immunology
20.
Immunol Rev ; 215: 77-88, 2007 Feb.
Article En | MEDLINE | ID: mdl-17291280

The gammadelta T-cell receptors (TCRs) are limited in their diversity, suggesting that their natural ligands may be few in number. Ligands for gammadeltaTCRs that have thus far been determined are predominantly of host rather than foreign origin. Correlations have been noted between the Vgamma and/or Vdelta genes a gammadelta T cell expresses and its functional role. The reason for these correlations is not yet known, but several different mechanisms are conceivable. One possibility is that interactions between particular TCR-V domains and ligands determine function or functional development. However, a recent study showed that at least for one ligand, receptor specificity is determined by the complementarity-determining region 3 (CDR3) component of the TCR-delta chain, regardless of the Vgamma and/or Vdelta. To determine what is required in the TCR for other specificities and to test whether recognition of certain ligands is connected to cell function, more gammadeltaTCR ligands must be defined. The use of recombinant soluble versions of gammadeltaTCRs appears to be a promising approach to finding new ligands, and recent results using this method are reviewed.


Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocyte Subsets/immunology , Animals , Complementarity Determining Regions/immunology , Humans , Ligands
...