Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 18 de 18
1.
Pathology ; 54(3): 279-285, 2022 Apr.
Article En | MEDLINE | ID: mdl-34635319

Therapeutically actionable ROS1 rearrangements have been described in 1-3% of non-small cell lung cancer (NSCLC). Screening for ROS1 rearrangements is recommended to be by immunohistochemistry (IHC), followed by confirmation with fluorescence in situ hybridisation (FISH) or sequencing. However, in practise ROS1 IHC presents difficulties due to conflicting scoring systems, multiple clones and expression in tumours that are wild-type for ROS1. We assessed ROS1 IHC in 285 consecutive cases of NSCLC with non-squamous histology over a nearly 2-year period. IHC was scored with ROS1 clone D4D6 (n=270), clone SP384 (n=275) or both clones (n=260). Results were correlated with ROS1 break-apart FISH (n=67), ALK status (n=194), and sequence data of EGFR (n=178) and other drivers, where possible. ROS1 expression was detected in 161/285 cases (56.5%), including 13/14 ROS1 FISH-positive cases. There was no ROS1 expression in one ROS1 FISH-positive case in which sequencing detected an ALK-EML4 fusion, but not a ROS1 fusion. The other 13 ROS1 FISH-positive cases showed moderate to strong staining with both IHC clones. However, one case with a TPM3-ROS1 fusion would have been scored as negative with SP384 and D4D6 clones by some previous criteria. ROS1 expression was also detected in 58/285 cases (20.4%) that had driver mutations in genes other than ROS1. A sensitivity of 100% for detecting a ROS1 rearrangement by FISH was achieved by omitting intensity from the IHC scoring criteria and expression in >0% cells with D4D6 or in ≥50% cells with SP384. Excluding cases with driver events in any MAPK pathway gene (e.g., in ALK, EGFR, KRAS, BRAF, ERBB2 and MET) substantially reduced the number of cases proceeding to ROS1 FISH. Only 15.9% of MAPK-negative NSCLC would proceed to FISH for an IHC threshold of >0% cells with D4D6, with a specificity of 42.4%. For a threshold of ≥50% cells with SP384, only 18.5% of MAPK-negative cases would proceed to FISH, with a specificity of 31.4%. Based on our data we suggest an algorithm for screening for ROS1 rearrangements in NSCLC in which ROS1 FISH is only performed in cases that have been demonstrated to lack activating mutations in any MAPK pathway gene by comprehensive sequencing and ALK IHC, and show staining at any intensity in ≥50% of cells with clone SP384, or >0% cells with D4D6.


Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/genetics , Early Detection of Cancer , Gene Rearrangement , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence/methods , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
2.
Lab Invest ; 101(1): 26-37, 2021 01.
Article En | MEDLINE | ID: mdl-32873880

Most NUTM1-rearranged neoplasms (NRNs) have fusions between NUTM1 and BRD (bromodomain-containing) family members and are termed NUT carcinomas (NCs) because they show some squamous differentiation. However, some NRNs are associated with fusions between NUTM1 and members of the MAD (MAX dimerization) gene family of MYC antagonists. Here we describe a small round cell malignancy from the gastro-esophageal junction with a previously unreported fusion between NUTM1 and the MAD family member MXI1. In contrast to NCs, the MXI1-NUTM1 tumor did not show squamous differentiation and did not express MYC, TP63 or SOX2, genes known to be targets of BRD-NUTM1 proteins and critical for NC oncogenesis. Transcriptome analysis showed paradoxical enrichment of MYC target genes in the MXI1-NUTM1 tumor despite the lack of MYC expression. When expressed in vitro MXI1-NUTM1 partially phenocopied MYC, enhancing cell proliferation and cooperating with oncogenic HRAS to produce anchorage-independent cell growth. These data provide evidence that MAD family members, which are normally repressors of MYC activity, can be converted into MYC-like mimics by fusion to NUTM1. The pathological features and novel oncogenic mechanism of the MXI1-NUTM1 tumor show that identification of NUTM1 fusion partners can be important for accurate diagnostic classification of some NRN subtypes, and potentially may guide therapeutic options.


Basic Helix-Loop-Helix Transcription Factors/genetics , Esophageal Neoplasms/genetics , Esophagogastric Junction/pathology , Neoplasm Proteins/genetics , Nuclear Proteins/genetics , Stomach Neoplasms/genetics , Tumor Suppressor Proteins/genetics , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Fatal Outcome , Female , Humans , Middle Aged , Oncogene Proteins, Fusion , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Transcriptome
3.
Int J Surg Pathol ; 28(5): 553-562, 2020 Aug.
Article En | MEDLINE | ID: mdl-31931637

The transcription factor GLI1 is a critical effector of the sonic hedgehog pathway. Gene fusions that activate GLI1 have recently been reported in several tumor types including gastroblastoma, plexiform fibromyxoma, a subset of pericytomas, and other soft tissue tumors. These tumors arise in a wide variety of anatomical origins and have variable malignant potentials, morphologies, and immunohistochemistry profiles. In this case report, we describe a malignant tumor from the jejunum with a MALAT1-GLI1 gene fusion that expressed a truncated constitutively active GLI1 protein and GLI1 targets that were detectable by immunohistochemistry. The tumor showed high-grade epithelioid and spindle cell morphology, strongly expressed CD56, and focally expressed other neuroendocrine markers and cytokeratins, but not S100 protein or SMA. The tumor recurred multiple times in liver, soft tissue, and lung over the course of 26 years, the longest reported follow-up for a GLI1 fusion-associated tumor. These metastatic tumors were also composed of epithelioid and spindle cells, but showed lower morphological grade than the primary tumor. The metastatic tumors resembled the recently reported "malignant epithelioid neoplasms with GLI1 rearrangements." The tumor also had a relatively high tumor mutation burden for a sarcoma. This case report expands the sites of origin for GLI1 rearranged neoplasms and shows that despite being associated with high-grade morphology, these malignancies can be associated with very long-term survival.


Jejunum/pathology , RNA, Long Noncoding/genetics , Soft Tissue Neoplasms/genetics , Soft Tissue Neoplasms/pathology , Zinc Finger Protein GLI1/genetics , Humans , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Oncogene Proteins, Fusion/genetics , Soft Tissue Neoplasms/mortality
4.
J Clin Invest ; 129(5): 1940-1945, 2019 05 01.
Article En | MEDLINE | ID: mdl-30835257

BRAF and CRAF are critical components of the MAPK signaling pathway which is activated in many cancer types. In approximately 1% of melanomas, BRAF or CRAF are activated through structural arrangements. We describe here a metastatic melanoma with a GOLGA4-RAF1 fusion and pathogenic variants in CTNNB1 and CDKN2A. Anti-CTLA4/anti-PD1 combination immunotherapy failed to control tumor progression. In the absence of other actionable variants the patient was administered MEK inhibitor therapy on the basis of its potential action against RAF1 fusions. This resulted in a profound and clinically significant response. We demonstrated that GOLGA4-RAF1 expression was associated with ERK activation, elevated expression of the RAS/RAF downstream co-effector ETV5, and a high Ki67 index. These findings provide a rationale for the dramatic response to targeted therapy. This study shows that thorough molecular characterization of treatment-resistant cancers can identify therapeutic targets and personalize management, leading to improved patient outcomes.


Autoantigens/genetics , MAP Kinase Kinase 1/antagonists & inhibitors , Melanoma/genetics , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-raf/genetics , Skin Neoplasms/genetics , Aged , Alleles , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Fluorodeoxyglucose F18/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Male , Neoplasm Metastasis , Oncogene Proteins, Fusion/metabolism , Positron-Emission Tomography , beta Catenin/metabolism
5.
Immunotherapy ; 9(3): 249-259, 2017 03.
Article En | MEDLINE | ID: mdl-28183192

AIM: Pilot clinical trial of NY-ESO-1 (ESO) protein in ISCOMATRIX™ adjuvant pulsed onto peripheral blood dendritic cells (PBDC), to ascertain feasibility, evaluate toxicity and assess induction of ESO-specific immune responses. PATIENTS & METHODS: Eligible participants had resected cancers expressing ESO or LAGE-1 and were at high risk of relapse. PBDC were produced using CliniMACS®plus, with initial depletion of CD1c+ B cells followed by positive selection of CD1c+ PBDC. Patients received three intradermal vaccinations of ESO/IMX-pulsed PBDC at 4-week intervals. RESULTS: The process was feasible and safe. No vaccine-induced immune responses were detected. Assays of immunomodulatory cells did not correlate with outcomes. One patient had a long lasting complete remission. CONCLUSION: This method was feasible and safe but was minimally immunogenic.


Blood Cells/physiology , Cancer Vaccines/immunology , Carcinoma, Basal Cell/therapy , Dendritic Cells/physiology , Immunotherapy/methods , Skin Neoplasms/therapy , T-Lymphocytes/immunology , Aged , Antigen Presentation , Antigens, CD1/metabolism , Antigens, Neoplasm/metabolism , Blood Cells/transplantation , Carcinoma, Basal Cell/immunology , Carcinoma, Basal Cell/pathology , Cells, Cultured , Cholesterol/metabolism , Dendritic Cells/transplantation , Drug Combinations , Female , Follow-Up Studies , Glycoproteins/metabolism , Humans , Immunity, Humoral , Lymphocyte Activation , Male , Membrane Proteins/metabolism , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Phospholipids/metabolism , Pilot Projects , Saponins/metabolism , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Treatment Outcome
6.
Int J Cancer ; 136(6): E590-601, 2015 Mar 15.
Article En | MEDLINE | ID: mdl-25081390

Vaccination strategies based on repeated injections of NY-ESO-1 protein formulated in ISCOMATRIX particles (NY-ESO-1 ISCOMATRIX) have shown to elicit combined NY-ESO-1 specific antibody and T cell responses. However, it remains unclear whether heterologous prime-boost strategies based on the combination with NY-ESO-1 ISCOMATRIX with different NY-ESO-1 boosting reagents could be used to increase NY-ESO-1 CD8(+) or CD4(+) T cell responses. To address this question, we carried out a randomized clinical trial in 39 high-risk, resected melanoma patients vaccinated with NY-ESO-1 ISCOMATRIX, and then boosted with repeated injections of either recombinant fowlpox virus encoding full length NY-ESO-1 (rF-NY-ESO-1) (Arm A) or NY-ESO-1 ISCOMATRIX alone (Arm B). We have comprehensively analyzed NY-ESO-1 specific T cells and B cells response in all patients before and after vaccination for a total of seven time points per patient. NY-ESO-1 ISCOMATRIX alone elicited a strong NY-ESO-1 specific CD4(+) T cell and antibody response, which was maintained by both regiments at similar levels. However, CD8(+) T cell responses were significantly boosted in 3 out of 18 patients in Arm A after the first rF-NY-ESO-1 injection and such responses were maintained until the end of the trial, while no patients in Arm B showed similar CD8(+) T cell responses. In addition, our results clearly identified immunodominant regions in the NY-ESO-1 protein: NY-ESO-179-102 and NY-ESO-1115-138 for CD4+ T cells and NY-ESO-185-108 for CD8+ T cells in a large proportion of vaccinated patients. These regions of NY-ESO-1 protein should be considered in future clinical trials as immunodominant epitopes.


Adjuvants, Immunologic/pharmacology , Antigens, Neoplasm/immunology , Cholesterol/pharmacology , Melanoma/therapy , Membrane Proteins/immunology , Phospholipids/pharmacology , Saponins/pharmacology , Antibody Formation , Antigens, Neoplasm/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Drug Combinations , Fowlpox virus/genetics , Humans , Melanoma/immunology , Membrane Proteins/genetics , Vaccination , Vaccines, Synthetic/immunology
7.
Cancer Immunol Immunother ; 60(11): 1625-37, 2011 Nov.
Article En | MEDLINE | ID: mdl-21698545

BACKGROUND: NY-ESO-1 protein formulated in ISCOMATRIX™ results in CD4+, CD8+ T cell and antibody-mediated immunity. We evaluated persistence of immunity, relapse-free survival and tumour antigen expression upon relapse in patients vaccinated in an earlier trial. METHODS: Immunity was measured in 28 patients with resected NY-ESO-1-expressing tumours (melanoma 25, breast 3) 252-1,155 days (median = 681) after vaccination. In the earlier vaccination, trial patients received NY-ESO-1 with ISCOMATRIX™ adjuvant at three protein doses 10 µg, 30 µg or 100 µg (n = 14); 100 µg NY-ESO-1 protein (n = 8) or placebo (n = 6), together with 1 µg of intradermal (ID) NY-ESO-1 protein twice for DTH skin testing. Immune responses assessed in the current study included antibody titres, circulating NY-ESO-1-specific T cells and DTH reactivity 2 days after DTH skin testing with NY-ESO-1 protein (1 µg) or peptides (10 µg). Relapse-free survival was determined for 42 melanoma patients. On relapse NY-ESO-1 and HLA, class I was assessed by immunohistochemistry in 17. RESULTS: Persisting anti-NY-ESO-1 immunity was detected in 10/14 recipients who had previously received vaccine with ISCOMATRIX™ adjuvant. In contrast, immunity only persisted in 3/14 who received 100 µg un-adjuvanted NY-ESO-1 protein (3/8) or 2 µg DTH protein (0/6) P = 0.02. Hence, persisting NY-ESO-1 immunity was associated with prior adjuvant. Tumour NY-ESO-1 or HLA class I was downregulated in participants who relapsed suggesting immunoediting had occurred. CONCLUSION: Immunoediting suggests that a signal of anti-tumour activity was observed in high-risk resected melanoma patients vaccinated with NY-ESO-1/ISCOMATRIX™. This was associated with measurable persisting immunity in the majority of vaccinated subjects tested. A prospective randomised trial has been undertaken to confirm these results.


Antigens, Neoplasm/administration & dosage , Breast Neoplasms/therapy , Cancer Vaccines/administration & dosage , Cholesterol/administration & dosage , Melanoma/therapy , Membrane Proteins/administration & dosage , Phospholipids/administration & dosage , Saponins/administration & dosage , Adjuvants, Immunologic/administration & dosage , Adult , Aged , Amino Acid Sequence , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/immunology , Breast Neoplasms/immunology , Cancer Vaccines/immunology , Cholesterol/immunology , Disease-Free Survival , Down-Regulation , Drug Combinations , Drug Hypersensitivity/etiology , Drug Hypersensitivity/immunology , Female , Humans , Immunohistochemistry , Male , Melanoma/immunology , Membrane Proteins/biosynthesis , Membrane Proteins/immunology , Middle Aged , Molecular Sequence Data , Phospholipids/immunology , Prospective Studies , Saponins/immunology , Skin/immunology
8.
Eur J Cancer ; 47(3): 460-9, 2011 Feb.
Article En | MEDLINE | ID: mdl-21115342

To determine the effect of Cancer-Testis Antigen (CTAg) expression on the natural history of primary cutaneous melanoma we compared its impact on prognosis with that of known prognostic factors and its relationship with other clinicopathologic characteristics. The immunohistochemical expression of three CTAgs (MAGE-A1, MAGE-A4 and NY-ESO-1) in 348 cases of stage I and stage II primary cutaneous melanoma was analysed and correlated with clinicopathologic characteristics, relapse free survival (RFS) and overall survival (OS). A Cox proportional hazards regression model was used to analyse factors which independently predicted RFS. All three CTAgs were significantly co-expressed with each other (p < 0.001). The median RFS for patients with CTAg-negative tumours and CTAg-positive tumours was 72 months and 45 months, respectively, (P = 0.008). Univariate analysis demonstrated that the impact of CTAg expression on RFS was comparable in magnitude to that of Breslow thickness, ulceration and tumour mitotic rate. Multivariate Cox regression analysis indicated that CTAg expression was a powerful independent predictor of RFS (risk ratio (RR) = 1.715, 95% confidence interval (CI) = 0.430-0.902, P = 0.010). In contrast, CTAg expression was demonstrated to have no prognostic impact on overall survival. This study demonstrates that CTAg expression in primary cutaneous melanoma is a strong independent predictor of RFS and it is comparable to other known important prognostic factors. CTAg expression has no relationship with overall survival, suggesting anti-melanoma immunity directed towards CTAg expression may contribute to the natural history of the disease. In view of these results, further investigation of the function of CTAgs and their potential use in therapeutic targeting is warranted.


Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Melanoma/mortality , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Peptide Fragments/metabolism , Skin Neoplasms/mortality , Adult , Aged , Disease-Free Survival , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Melanoma/metabolism , Melanoma/pathology , Middle Aged , Mitosis , Prognosis , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Skin Ulcer/mortality , Skin Ulcer/pathology
9.
Clin Cancer Res ; 15(7): 2507-13, 2009 Apr 01.
Article En | MEDLINE | ID: mdl-19318477

PURPOSE: Ipilimumab is a monoclonal antibody that blocks the immune-inhibitory interaction between CTL antigen 4 (CTLA-4) and its ligands on T cells. Clinical trials in cancer patients with ipilimumab have shown promising antitumor activity, particularly in patients with advanced melanoma. Often, tumor regressions in these patients are correlated with immune-related side effects such as dermatitis, enterocolitis, and hypophysitis. Although these reactions are believed to be immune-mediated, the antigenic targets for the cellular or humoral immune response are not known. EXPERIMENTAL DESIGN: We enrolled patients with advanced melanoma in a phase II study with ipilimumab. One of these patients experienced a complete remission of his tumor. The specificity and functional properties of CD8-positive T cells in his peripheral blood, in regressing tumor tissue, and at the site of an immune-mediated skin rash were investigated. RESULTS: Regressing tumor tissue was infiltrated with CD8-positive T cells, a high proportion of which were specific for Melan-A. The skin rash was similarly infiltrated with Melan-A-specific CD8-positive T cells, and a dramatic (>30-fold) increase in Melan-A-specific CD8-positive T cells was apparent in peripheral blood. These cells had an effector phenotype and lysed Melan-A-expressing tumor cells. CONCLUSIONS: Our results show that Melan-A may be a major target for both the autoimmune and antitumor reactions in patients treated with anti-CTLA-4, and describe for the first time the antigen specificity of CD8-positive T cells that mediate tumor rejection in a patient undergoing treatment with an anti-CTLA-4 antibody. These findings may allow a better integration of ipilimumab into other forms of immunotherapy.


Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Melanoma/drug therapy , Neoplasm Proteins/immunology , Skin Neoplasms/drug therapy , T-Lymphocytes, Cytotoxic/immunology , Autoimmunity , Cytotoxicity, Immunologic , Double-Blind Method , Exanthema/chemically induced , Exanthema/immunology , Humans , Ipilimumab , Lymphocytes, Tumor-Infiltrating/immunology , MART-1 Antigen , Melanoma/diagnostic imaging , Melanoma/immunology , Skin Neoplasms/diagnostic imaging , Skin Neoplasms/immunology , Tomography, X-Ray Computed
10.
Cancer Immunol Immunother ; 58(10): 1635-46, 2009 Oct.
Article En | MEDLINE | ID: mdl-19221743

"Cancer stem cells" that resist conventional treatments may be a cause of therapeutic failure in melanoma. We report a subpopulation of clonogenic melanoma cells that are characterized by high prominin-1/CD133 expression in melanoma and melanoma cell lines. These cells have enhanced clonogenicity and self-renewal in vitro, and serve as a limited in vitro model for melanoma stem cells. In some cases clonogenic CD133(+) melanoma cells show increased expression of some cancer/testis (CT) antigens. The expression of NY-ESO-1 in an HLA-A2 expressing cell line allowed CD133(+) clonogenic melanoma cells to be targeted for killing in vitro by NY-ESO-1-specific CD8(+) T-lymphocytes. Our in vitro findings raise the hypothesis that if melanoma stem cells express CT antigens in vivo that immune targeting of these antigens may be a viable clinical strategy for the adjuvant treatment of melanoma.


Antigens, CD/metabolism , Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Glycoproteins/metabolism , Melanoma/immunology , Membrane Proteins/immunology , Peptides/metabolism , Skin Neoplasms/immunology , AC133 Antigen , CD8-Positive T-Lymphocytes/immunology , Colony-Forming Units Assay , Cytotoxicity, Immunologic , Fluorescent Antibody Technique , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , Humans , Immunoenzyme Techniques , Lymphatic Metastasis , Male , Melanoma/secondary , Melanoma/therapy , Peptide Fragments/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/pathology , Skin Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology
11.
Blood ; 113(6): 1257-67, 2009 Feb 05.
Article En | MEDLINE | ID: mdl-18987360

Professional antigen-presenting cells (APCs) are sentinel cells of the immune system that present antigen to T lymphocytes and mediate an appropriate immune response. It is therefore surprising that knowledge of the professional APCs in human lymph nodes is limited. Using 3-color immunohistochemistry, we have identified APCs in human lymph nodes, excluding plasmacytoid APCs, that fall into 2 nonoverlapping classes: (1) CD209+ APCs, coexpressing combinations of CD206, CD14, and CD68, that occupied the medullary cords, lined the capsule and trabeculae and were also scattered throughout the diffuse T-lymphocyte areas of the paracortex; and (2) APCs expressing combinations of CD1a, CD207, and CD208, that were always restricted to the paracortex. Surprisingly, this second class of APCs was almost entirely absent from many lymph nodes. Our data suggest that most CD208+ cells, often referred to as "interdigitating cells," derive from migratory APCs, and that the major APC subset consistently resident in the paracortex of human lymph nodes is the CD209+ subset. All APC subsets were demonstrated to be in close contact with the fibroreticular network. The identification of 2 distinct APC populations in the paracortex of human lymph nodes has important implications for understanding T-lymphocyte responses and optimizing vaccine design.


Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Lymph Nodes/immunology , Antigens, CD/metabolism , Axilla , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Lymphatic Diseases , Phenotype , Skin/cytology , Skin/immunology , Skin/metabolism , T-Lymphocytes/immunology
12.
Clin Cancer Res ; 14(11): 3291-8, 2008 Jun 01.
Article En | MEDLINE | ID: mdl-18519755

PURPOSE: Cancer cells recapitulate many behaviors of pluripotent embryonic cells such as unlimited proliferation, and the capacity to self-renew and to migrate. Embryo-cancer sequence A (ECSA), later named developmental pluripotency associated-2 (DPPA2), is an embryonic gene initially isolated from pluripotent human preimplantation embryos. We hypothesized that ECSA/DPPA2 would be quiescent in most normal tissues but expressed in cancers and may therefore be a useful target for immunotherapy. EXPERIMENTAL DESIGN: ECSA/DPPA2 expression was examined in a panel of normal and tumor tissue by reverse transcription PCR, quantitative real-time PCR, and immunohistochemistry. A panel of 110 non-small cell lung cancers (NSCLC) were further investigated for the presence of ECSA/DPPA2 transcripts and several cancer testis antigens (CTA). Sera from 104 patients were analyzed for spontaneous ECSA/DPPA2 antibody production by ELISA and Western blot. RESULTS: ECSA/DPPA2 transcripts were limited to normal testis, placenta, bone marrow, thymus, and kidney but expressed in a variety of tumors most notably in 30% of NSCLC. Enrichment for CTAs in ECSA/DPPA2-positive NSCLC was observed. Immunohistochemistry confirmed nuclear and cytoplasmic localization in subpopulations of cells with coexpression of the CTA MAGE-A3. Antibodies to recombinant ECSA/DPPA2 protein were detected in the sera of 4 of 104 patients with NSCLC but not in healthy controls. CONCLUSIONS: The restricted expression in normal tissues, expression in tumors with coexpression of CTAs, and spontaneous immunogenicity indicate that ECSA/DPPA2 is a promising target for antigen-specific immunotherapy in NSCLC.


Antigens, Neoplasm/biosynthesis , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Nuclear Proteins/biosynthesis , Blotting, Western , Cell Cycle Proteins , Enzyme-Linked Immunosorbent Assay , Gene Expression , Humans , Immunohistochemistry , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors
13.
Cancer Res ; 68(8): 3001-9, 2008 Apr 15.
Article En | MEDLINE | ID: mdl-18413770

FoxP3 is a member of the forkhead family of transcription factors critically involved in the development and function of CD25(+) regulatory T cells (Treg). Until recently, FoxP3 expression was thought to be restricted to the T-cell lineage. However, using immunohistochemistry and flow cytometric analysis of human melanoma tissue, we detected FoxP3 expression not only in the tumor infiltrating Treg but also in the melanoma cells themselves. FoxP3 is also widely expressed by established human melanoma cell lines (as determined by flow cytometry, PCR, and Western blot), as well as cell lines derived from other solid tumors. Normal B cells do not express FoxP3; however, expression could be induced after transformation with EBV in vitro and in vivo, suggesting that malignant transformation of healthy cells can induce FoxP3. In addition, a FOXP3 mRNA variant lacking exons 3 and 4 was identified in tumor cell lines but was absent from Treg. Interestingly, this alternative splicing event introduces a translation frame-shift that is predicted to encode a novel protein. Together, our results show that FoxP3, a key regulator of immune suppression, is not only expressed by Treg but also by melanoma cells, EBV-transformed B cells, and a wide variety of tumor cell lines.


B-Lymphocytes/immunology , Forkhead Transcription Factors/genetics , T-Lymphocytes, Regulatory/immunology , B-Lymphocytes/virology , Cell Line, Tumor , Flow Cytometry , Forkhead Transcription Factors/immunology , Glioma/genetics , Glioma/immunology , Herpesvirus 4, Human/immunology , Humans , Lymphocyte Activation/immunology , Male , Melanoma/genetics , Melanoma/immunology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology
14.
J Immunother ; 29(5): 499-511, 2006.
Article En | MEDLINE | ID: mdl-16971806

Flt3 ligand mobilizes dendritic cells (DCs) into blood, allowing generation in vivo of large numbers of DCs for immunotherapy. These immature DCs can be rapidly activated by soluble CD40 ligand (CD40L). We developed a novel overnight method using these cytokines to produce DCs for cancer immunotherapy. Flt3 ligand-mobilized DCs (FLDCs) were isolated, activated with CD40L, loaded with antigenic peptides from influenza matrix protein, hepatitis B core antigen, NY-ESO-1, MAGE-A4, and MAGE-A10, and injected into patients with resected melanoma. Three injections were given at 4-week intervals. Study end points included antigen-specific immune responses (skin reactions to peptides alone or peptide-pulsed FLDCs; circulating T-cell responses), safety, and toxicity. No patient had a measurable tumor. Six patients were entered. FLDCs were obtained, enriched, and cultured under Good Manufacturing Practice grade conditions. Overnight culture with soluble CD40L caused marked up-regulation of activation markers (CD83 and HLA-DR). These FLDCs were functional and able to stimulate antigen-specific T cells in vitro. No significant adverse events were attributable to FLDCs. Peptide-pulsed FLDCs caused strong local skin reactions up to 60 mm diameter with intense perivascular infiltration of T cells, exceeding those seen in our previous peptide-based protocols. Antigen-specific blood T-cell responses were induced, including responses to an antigen for which the patients were naive (hepatitis B core antigen) and MAGE-A10. MAGE-A10-specific T cells with a skewed T-cell receptor repertoire were detected in 1 patient in blood ex vivo and from tumor biopsies. Vaccination with FLDCs pulsed with peptides is safe and primes immune responses to cancer antigens.


CD40 Ligand/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/therapeutic use , Dendritic Cells/immunology , Melanoma/therapy , Membrane Proteins/immunology , Skin Neoplasms/therapy , Adult , Antigen Presentation , CD8 Antigens/immunology , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Cells, Cultured , Cytotoxicity, Immunologic , Female , Humans , Immunotherapy, Adoptive , Leukapheresis , Lymphocyte Activation , Male , Melanoma/immunology , Middle Aged , Peptides/immunology , Peptides/therapeutic use , Pilot Projects , Receptors, Antigen, T-Cell/metabolism , Skin Neoplasms/immunology
15.
Melanoma Res ; 16(4): 347-55, 2006 Aug.
Article En | MEDLINE | ID: mdl-16845331

Desmoplastic melanoma is a diagnostic and therapeutic challenge. Immunohistochemical analysis with antibodies to melanoma antigens can complement morphologic evaluation. Although staining for S100 protein is generally positive, staining for other melanoma differentiation antigens, particularly gp100, Melan-A/MART1 and tyrosinase, is often negative despite being commonly positive in other melanoma types. A high clinical index of suspicion and better diagnostic techniques are essential as atypical features and incorrect diagnosis can lead to poor clinical outcomes. Antigens associated with melanoma, such as the melanocyte differentiation and cancer testis antigen, may become important targets for immune therapies. We characterized the patterns of antigen expression of desmoplastic melanoma from 32 patients, including gp100, Melan-A/MART-1, tyrosinase, MAGE-A1, MAGE-A4 and NY-ESO-1. Consistent positive staining with S100 was observed. Differentiation antigens were expressed more frequently than cancer testis antigens regardless of the histological subtype of desmoplastic melanoma. When present, cancer testis antigen expression correlated to positive staining with differentiation antigens. The diagnostic yield of desmoplastic melanoma did not increase with the addition of cancer testis antigen typing. Low levels of expression of cancer testis antigen may indicate that they are suboptimal targets for vaccine development in desmoplastic melanoma.


Antigens, Differentiation/metabolism , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Cell Differentiation , Humans , MART-1 Antigen , Male , Melanoma/diagnosis , Melanoma-Specific Antigens , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Monophenol Monooxygenase/metabolism , Neoplasm Proteins/metabolism , Skin Neoplasms/diagnosis , gp100 Melanoma Antigen
16.
J Immunol ; 176(10): 5908-17, 2006 May 15.
Article En | MEDLINE | ID: mdl-16670298

Immunodominance has been well-demonstrated in many antiviral and antibacterial systems, but much less so in the setting of immune responses against cancer. Tumor Ag-specific CD8+ T cells keep cancer cells in check via immunosurveillance and shape tumor development through immunoediting. Because most tumor Ags are self Ags, the breadth and depth of antitumor immune responses have not been well-appreciated. To design and develop antitumor vaccines, it is important to understand the immunodominance hierarchy and its underlying mechanisms, and to identify the most immunodominant tumor Ag-specific T cells. We have comprehensively analyzed spontaneous cellular immune responses of one individual and show that multiple tumor Ags are targeted by the patient's immune system, especially the "cancer-testis" tumor Ag NY-ESO-1. The pattern of anti-NY-ESO-1 T cell responses in this patient closely resembles the classical broad yet hierarchical antiviral immunity and was confirmed in a second subject.


Antigens, Neoplasm/physiology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunodominant Epitopes/immunology , Membrane Proteins/physiology , Animals , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , CHO Cells , Cell Line, Transformed , Cell Line, Tumor , Cricetinae , Cricetulus , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , Humans , Immunity, Innate , Immunodominant Epitopes/metabolism , Male , Middle Aged
17.
Clin Cancer Res ; 12(3 Pt 1): 764-71, 2006 Feb 01.
Article En | MEDLINE | ID: mdl-16467087

PURPOSE: Melanoma cells express antigens that can induce T-cell and antibody responses. Obtaining a detailed understanding of antigen expression in primary and metastatic melanoma is essential if these molecules are to be useful targets for immunotherapy of melanoma. EXPERIMENTAL DESIGN: Malignant melanomas (n = 586) from 426 patients were typed for antigen expression. Multiple samples were available from 86 individuals, enabling analysis of antigen expression patterns over time. Paraffin-embedded samples were tested by immunohistochemistry for the presence of the differentiation antigens: gp100, Melan-A, tyrosinase, and the "cancer/testis" antigens MAGE-A1, MAGE-A4, and NY-ESO-1. RESULTS: Samples were primary tumors (n = 251), lymph node metastases (n = 174), s.c. metastases (n = 71), and distant metastases (n = 90). The differentiation antigens were strongly expressed in 93% to 95% of tumors regardless of stage. In contrast, the frequency of cancer/testis antigen expression in primary tumors for MAGE-A1, MAGE-A4, and NY-ESO-1 was lower (20%, 9%, and 45%, respectively). MAGE-A1 and MAGE-A4 were acquired with advancing disease (to 51% and 44% in distant metastases, respectively) but not NY-ESO-1, which remained positive in 45%. MAGE-A1 expression was twice as prevalent in ulcerated primaries as in nonulcerated primaries (30% versus 15%; P = 0.006) and in thicker as opposed to thin melanomas (26% versus 10%; P = 0.1). CONCLUSIONS: This large series describes patterns of antigen expression in melanoma and their evolution over time. This will help inform decisions about selection of patients and target antigens for melanoma immunotherapy clinical trials.


Antigens, Neoplasm/biosynthesis , Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic , Melanoma/genetics , Antibodies, Monoclonal , Antigens, Differentiation/analysis , Antigens, Differentiation/genetics , Antigens, Differentiation/immunology , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/immunology , Disease Progression , Humans , Immunohistochemistry , Male , Melanoma/immunology , Melanoma/pathology , Neoplasm Staging
18.
Clin Cancer Res ; 10(24): 8396-404, 2004 Dec 15.
Article En | MEDLINE | ID: mdl-15623618

PURPOSE: NY-ESO-1 and LAGE-1 are homologous cancer-testis antigens, which are expressed in many different cancers. It is essential to type tumors accurately to assess patient suitability for clinical trials which target these. This study evaluates typing strategies used to distinguish these two homologous but distinct antigens and to characterize and quantitate expression of each in clinical samples. EXPERIMENTAL DESIGN: We typed 120 malignant melanomas for the expression of NY-ESO-1 and LAGE-1 with immunohistochemistry, reverse transcription-PCR (RT-PCR), and quantitative real-time (qRT-PCR), which was also used to explore the relationship between NY-ESO-1 and LAGE expression. RESULTS: The two monoclonal antibodies ES121 and E978 had very similar immunohistochemistry reactivities. Both were specific for NY-ESO-1 because neither bound to homologous LAGE-1 peptides despite 84% overall amino acid homology. Of 120 melanomas tested by immunohistochemistry, NY-ESO-1 was expressed in >50% of cells in 23 melanomas (19%), between 11 and 50% cells in 15 (12.5%), <11% cells in 16 (13.5%), and negative in 66 (55%). Although specific for both antigens, the PCR methods did not provide this information about microheterogeneity. Polymorphisms in the LAGE-1 gene resulted in false negative LAGE-1 typing by qRT-PCR by inhibiting binding of oligonucleotide primers, thereby showing the exquisite specificity of qRT-PCR as a typing method. CONCLUSIONS: For NY-ESO-1 typing, immunohistochemistry compared favorably with the RT-PCR, with the added advantage of being able to characterize heterogeneity of antigen expression. Because neither mAb bound LAGE and because there was no coordinate expression LAGE and NY-ESO-1, separate typing for each is required.


Antigens, Neoplasm/metabolism , Melanoma/metabolism , Membrane Proteins/metabolism , Skin Neoplasms/metabolism , Antibodies, Monoclonal , Antigens, Neoplasm/genetics , Antigens, Surface , Gene Expression Regulation, Neoplastic , Humans , Immunoenzyme Techniques , Male , Melanoma/genetics , Melanoma/secondary , Membrane Proteins/genetics , Polymorphism, Genetic , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/genetics , Skin Neoplasms/secondary , Testis/immunology , Testis/metabolism , Tissue Distribution
...