Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 15 de 15
1.
Cancer Lett ; 587: 216713, 2024 Apr 10.
Article En | MEDLINE | ID: mdl-38364961

Human leukocyte antigen (HLA) class I defects are associated with cancer progression. However, their prognostic significance is controversial and may be modulated by immune checkpoints. Here, we investigated whether the checkpoint B7-H3 modulates the relationship between HLA class I and pancreatic ductal adenocarcinoma (PDAC) prognosis. PDAC tumors were analyzed for the expression of B7-H3, HLA class I, HLA class II molecules, and for the presence of tumor-infiltrating immune cells. We observed defective HLA class I and HLA class II expressions in 75% and 59% of PDAC samples, respectively. HLA class I and B7-H3 expression were positively related at mRNA and protein level, potentially because of shared regulation by RELA, a sub-unit of NF-kB. High B7-H3 expression and low CD8+ T cell density were indicators of poor survival, while HLA class I was not. Defective HLA class I expression was associated with unfavorable survival only in patients with low B7-H3 expression. Favorable survival was observed only when HLA class I expression was high and B7-H3 expression low. Our results provide the rationale for targeting B7-H3 in patients with PDAC tumors displaying high HLA class I levels.


Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , B7 Antigens/genetics , B7 Antigens/metabolism , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Carcinoma, Pancreatic Ductal/pathology , Disease Progression , Histocompatibility Antigens Class I , Lymphocytes, Tumor-Infiltrating , Pancreatic Neoplasms/metabolism , Prognosis
2.
Nat Med ; 29(11): 2814-2824, 2023 Nov.
Article En | MEDLINE | ID: mdl-37857711

Tebotelimab, a bispecific PD-1×LAG-3 DART molecule that blocks both PD-1 and LAG-3, was investigated for clinical safety and activity in a phase 1 dose-escalation and cohort-expansion clinical trial in patients with solid tumors or hematologic malignancies and disease progression on previous treatment. Primary endpoints were safety and maximum tolerated dose of tebotelimab when administered as a single agent (n = 269) or in combination with the anti-HER2 antibody margetuximab (n = 84). Secondary endpoints included anti-tumor activity. In patients with advanced cancer treated with tebotelimab monotherapy, 68% (184/269) experienced treatment-related adverse events (TRAEs; 22% were grade ≥3). No maximum tolerated dose was defined; the recommended phase 2 dose (RP2D) was 600 mg once every 2 weeks. There were tumor decreases in 34% (59/172) of response-evaluable patients in the dose-escalation cohorts, with objective responses in multiple solid tumor types, including PD-1-refractory disease, and in LAG-3+ non-Hodgkin lymphomas, including CAR-T refractory disease. To enhance potential anti-tumor responses, we tested margetuximab plus tebotelimab. In patients with HER2+ tumors treated with tebotelimab plus margetuximab, 74% (62/84) had TRAEs (17% were grade ≥3). The RP2D was 600 mg once every 3 weeks. The confirmed objective response rate in these patients was 19% (14/72), including responses in patients typically not responsive to anti-HER2/anti-PD-1 combination therapy. ClinicalTrials.gov identifier: NCT03219268 .


Hematologic Neoplasms , Immunoconjugates , Neoplasms , Humans , Programmed Cell Death 1 Receptor/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Neoplasms/pathology , Hematologic Neoplasms/drug therapy
3.
Nat Med ; 29(4): 888-897, 2023 04.
Article En | MEDLINE | ID: mdl-37012549

B7 homolog 3 (B7-H3; CD276), a tumor-associated antigen and possible immune checkpoint, is highly expressed in prostate cancer (PCa) and is associated with early recurrence and metastasis. Enoblituzumab is a humanized, Fc-engineered, B7-H3-targeting antibody that mediates antibody-dependent cellular cytotoxicity. In this phase 2, biomarker-rich neoadjuvant trial, 32 biological males with operable intermediate to high-risk localized PCa were enrolled to evaluate the safety, anti-tumor activity and immunogenicity of enoblituzumab when given before prostatectomy. The coprimary outcomes were safety and undetectable prostate-specific antigen (PSA) level (PSA0) 1 year postprostatectomy, and the aim was to obtain an estimate of PSA0 with reasonable precision. The primary safety endpoint was met with no notable unexpected surgical or medical complications, or surgical delay. Overall, 12% of patients experienced grade 3 adverse events and no grade 4 events occurred. The coprimary endpoint of the PSA0 rate 1 year postprostatectomy was 66% (95% confidence interval 47-81%). The use of B7-H3-targeted immunotherapy in PCa is feasible and generally safe and preliminary data suggest potential clinical activity. The present study validates B7-H3 as a rational target for therapy development in PCa with larger studies planned. The ClinicalTrials.gov identifier is NCT02923180.


Antineoplastic Agents , Prostatic Neoplasms , Male , Humans , Prostate-Specific Antigen/therapeutic use , Neoadjuvant Therapy , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/surgery , Prostatic Neoplasms/pathology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/therapeutic use , B7 Antigens
4.
Mol Cancer Ther ; 21(7): 1047-1059, 2022 07 05.
Article En | MEDLINE | ID: mdl-35511740

ADAM metallopeptidase domain 9 (ADAM9) is a member of the ADAM family of multifunctional, multidomain type 1 transmembrane proteins. ADAM9 is overexpressed in many cancers, including non-small cell lung, pancreatic, gastric, breast, ovarian, and colorectal cancer, but exhibits limited expression in normal tissues. A target-unbiased discovery platform based on intact tumor and progenitor cell immunizations, followed by an IHC screen, led to the identification of anti-ADAM9 antibodies with selective tumor-versus-normal tissue binding. Subsequent analysis revealed anti-ADAM9 antibodies were efficiently internalized and processed by tumor cells making ADAM9 an attractive target for antibody-drug conjugate (ADC) development. Here, we describe the preclinical evaluation of IMGC936, a novel ADC targeted against ADAM9. IMGC936 is comprised of a high-affinity humanized antibody site-specifically conjugated to DM21-C, a next-generation linker-payload that combines a maytansinoid microtubule-disrupting payload with a stable tripeptide linker, at a drug antibody ratio of approximately 2.0. In addition, the YTE mutation (M252Y/S254T/T256E) was introduced into the CH2 domain of the antibody Fc to maximize in vivo plasma half-life and exposure. IMGC936 exhibited cytotoxicity toward ADAM9-positive human tumor cell lines, as well as bystander killing, potent antitumor activity in human cell line-derived xenograft and patient-derived xenograft tumor models, and an acceptable safety profile in cynomolgus monkeys with favorable pharmacokinetic properties. Our preclinical data provide a strong scientific rationale for the further development of IMGC936 as a therapeutic candidate for the treatment of ADAM9-positive cancers. A first-in-human study of IMGC936 in patients with advanced solid tumors has been initiated (NCT04622774).


Immunoconjugates , ADAM Proteins , Cell Line, Tumor , Heterografts , Humans , Immunoconjugates/chemistry , Membrane Proteins/genetics , Xenograft Model Antitumor Assays
5.
IEEE Trans Vis Comput Graph ; 28(6): 2457-2469, 2022 06.
Article En | MEDLINE | ID: mdl-33090955

The analysis of bipartite networks is critical in a variety of application domains, such as exploring entity co-occurrences in intelligence analysis and investigating gene expression in bio-informatics. One important task is missing link prediction, which infers the existence of unseen links based on currently observed ones. In this article, we propose a visual analysis system, MissBiN, to involve analysts in the loop for making sense of link prediction results. MissBiN equips a novel method for link prediction in a bipartite network by leveraging the information of bi-cliques in the network. It also provides an interactive visualization for understanding the algorithm outputs. The design of MissBiN is based on three high-level analysis questions (what, why, and how) regarding missing links, which are distilled from the literature and expert interviews. We conducted quantitative experiments to assess the performance of the proposed link prediction algorithm, and interviewed two experts from different domains to demonstrate the effectiveness of MissBiN as a whole. We also provide a comprehensive usage scenario to illustrate the usefulness of the tool in an application of intelligence analysis.


Algorithms , Computer Graphics , Computational Biology
6.
J Data Sci Stat Vis ; 2(5)2022 Sep 07.
Article En | MEDLINE | ID: mdl-38318468

Identifying unique characteristics in a network through comparison with another network is an essential network analysis task. For example, with networks of protein interactions obtained from normal and cancer tissues, we can discover unique types of interactions in cancer tissues. This analysis task could be greatly assisted by contrastive learning, which is an emerging analysis approach to discover salient patterns in one dataset relative to another. However, existing contrastive learning methods cannot be directly applied to networks as they are designed only for high-dimensional data analysis. To address this problem, we introduce a new analysis approach called contrastive network representation learning (cNRL). By integrating two machine learning schemes, network representation learning and contrastive learning, cNRL enables embedding of network nodes into a low-dimensional representation that reveals the uniqueness of one network compared to another. Within this approach, we also design a method, named i-cNRL, which offers interpretability in the learned results, allowing for understanding which specific patterns are only found in one network. We demonstrate the effectiveness of i-cNRL for network comparison with multiple network models and real-world datasets. Furthermore, we compare i-cNRL and other potential cNRL algorithm designs through quantitative and qualitative evaluations.

7.
Cell Rep Med ; 1(9): 100163, 2020 12 22.
Article En | MEDLINE | ID: mdl-33377134

Combination immunotherapy with antibodies directed against PD-1 and CTLA-4 shows improved clinical benefit across cancer indications compared to single agents, albeit with increased toxicity. Leveraging the observation that PD-1 and CTLA-4 are co-expressed by tumor-infiltrating lymphocytes, an investigational PD-1 x CTLA-4 bispecific DART molecule, MGD019, is engineered to maximize checkpoint blockade in the tumor microenvironment via enhanced CTLA-4 blockade in a PD-1-binding-dependent manner. In vitro, MGD019 mediates the combinatorial blockade of PD-1 and CTLA-4, confirming dual inhibition via a single molecule. MGD019 is well tolerated in non-human primates, with evidence of both PD-1 and CTLA-4 blockade, including increases in Ki67+CD8 and ICOS+CD4 T cells, respectively. In the ongoing MGD019 first-in-human study enrolling patients with advanced solid tumors (NCT03761017), an analysis undertaken following the dose escalation phase revealed acceptable safety, pharmacodynamic evidence of combinatorial blockade, and objective responses in multiple tumor types typically unresponsive to checkpoint inhibitor therapy.


Antibodies/therapeutic use , CTLA-4 Antigen/immunology , Immunotherapy , Programmed Cell Death 1 Receptor/antagonists & inhibitors , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/drug effects , Humans , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/drug effects , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
8.
Mol Cancer Ther ; 19(11): 2235-2244, 2020 11.
Article En | MEDLINE | ID: mdl-32967924

B7-H3, also referred to as CD276, is a member of the B7 family of immune regulatory proteins. B7-H3 is overexpressed on many solid cancers, including prostate cancer, renal cell carcinoma, melanoma, squamous cell carcinoma of the head and neck, non-small cell lung cancer, and breast cancer. Overexpression of B7-H3 is associated with disease severity, risk of recurrence and reduced survival. In this article, we report the preclinical development of MGC018, an antibody-drug conjugate targeted against B7-H3. MGC018 is comprised of the cleavable linker-duocarmycin payload, valine-citrulline-seco duocarmycin hydroxybenzamide azaindole (vc-seco-DUBA), conjugated to an anti-B7-H3 humanized IgG1/kappa mAb through reduced interchain disulfides, with an average drug-to-antibody ratio of approximately 2.7. MGC018 exhibited cytotoxicity toward B7-H3-positive human tumor cell lines, and exhibited bystander killing of target-negative tumor cells when cocultured with B7-H3-positive tumor cells. MGC018 displayed potent antitumor activity in preclinical tumor models of breast, ovarian, and lung cancer, as well as melanoma. In addition, antitumor activity was observed toward patient-derived xenograft models of breast, prostate, and head and neck cancer displaying heterogeneous expression of B7-H3. Importantly, MGC018 exhibited a favorable pharmacokinetic and safety profile in cynomolgus monkeys following repeat-dose administration. The antitumor activity observed preclinically with MGC018, together with the positive safety profile, provides evidence of a potentially favorable therapeutic index and supports the continued development of MGC018 for the treatment of solid cancers. GRAPHICAL ABSTRACT: http://mct.aacrjournals.org/content/molcanther/19/11/2235/F1.large.jpg.


B7 Antigens/antagonists & inhibitors , Drug Evaluation, Preclinical , Immune Checkpoint Inhibitors/pharmacology , Immunoconjugates/pharmacology , Neoplasms/drug therapy , Animals , B7 Antigens/genetics , B7 Antigens/metabolism , Bystander Effect , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Dose-Response Relationship, Drug , Drug Monitoring , Gene Knockdown Techniques , Humans , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/isolation & purification , Immunoconjugates/chemistry , Immunoconjugates/isolation & purification , Mice , Neoplasms/metabolism , Neoplasms/pathology , Treatment Outcome , Xenograft Model Antitumor Assays
9.
Mol Cancer Ther ; 17(8): 1761-1772, 2018 08.
Article En | MEDLINE | ID: mdl-29866746

We have developed MGD007 (anti-glycoprotein A33 x anti-CD3), a DART protein designed to redirect T cells to target gpA33 expressing colon cancer. The gpA33 target was selected on the basis of an antibody-based screen to identify cancer antigens universally expressed in both primary and metastatic colorectal cancer specimens, including putative cancer stem cell populations. MGD007 displays the anticipated-bispecific binding properties and mediates potent lysis of gpA33-positive cancer cell lines, including models of colorectal cancer stem cells, through recruitment of T cells. Xenograft studies showed tumor growth inhibition at doses as low as 4 µg/kg. Both CD8 and CD4 T cells mediated lysis of gpA33-expressing tumor cells, with activity accompanied by increases in granzyme and perforin. Notably, suppressive T-cell populations could also be leveraged to mediate lysis of gpA33-expressing tumor cells. Concomitant with CTL activity, both T-cell activation and expansion are observed in a gpA33-dependent manner. No cytokine activation was observed with human PBMC alone, consistent with the absence of gpA33 expression on peripheral blood cell populations. Following prolonged exposure to MGD007 and gpA33 positive tumor cells, T cells express PD-1 and LAG-3 and acquire a memory phenotype but retain ability to support potent cell killing. In cynomolgus monkeys, 4 weekly doses of 100 µg/kg were well tolerated, with prolonged PK consistent with that of an Fc-containing molecule. Taken together, MGD007 displays potent activity against colorectal cancer cells consistent with a mechanism of action endowed in its design and support further investigation of MGD007 as a potential novel therapeutic treatment for colorectal cancer. Mol Cancer Ther; 17(8); 1761-72. ©2018 AACR.


Colorectal Neoplasms/drug therapy , Immunotherapy/methods , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Female , Haplorhini , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Metastasis
10.
IEEE Trans Vis Comput Graph ; 24(1): 195-204, 2018 01.
Article En | MEDLINE | ID: mdl-28866549

Discovering and analyzing biclusters, i.e., two sets of related entities with close relationships, is a critical task in many real-world applications, such as exploring entity co-occurrences in intelligence analysis, and studying gene expression in bio-informatics. While the output of biclustering techniques can offer some initial low-level insights, visual approaches are required on top of that due to the algorithmic output complexity. This paper proposes a visualization technique, called BiDots, that allows analysts to interactively explore biclusters over multiple domains. BiDots overcomes several limitations of existing bicluster visualizations by encoding biclusters in a more compact and cluster-driven manner. A set of handy interactions is incorporated to support flexible analysis of biclustering results. More importantly, BiDots addresses the cases of weighted biclusters, which has been underexploited in the literature. The design of BiDots is grounded by a set of analytical tasks derived from previous work. We demonstrate its usefulness and effectiveness for exploring computed biclusters with an investigative document analysis task, in which suspicious people and activities are identified from a text corpus.

11.
Sci Transl Med ; 7(289): 289ra82, 2015 May 27.
Article En | MEDLINE | ID: mdl-26019218

Current therapies for acute myeloid leukemia (AML) are largely ineffective, and AML patients may benefit from targeted immunotherapy approaches. MGD006 is a bispecific CD3xCD123 dual-affinity re-targeting (DART) molecule that binds T lymphocytes and cells expressing CD123, an antigen up-regulated in several hematological malignancies including AML. MGD006 mediates blast killing in AML samples, together with concomitant activation and expansion of residual T cells. MGD006 is designed to be rapidly cleared, and therefore requires continuous delivery. In a mouse model of continuous administration, MGD006 eliminated engrafted KG-1a cells (an AML-M0 line) in human PBMC (peripheral blood mononuclear cell)-reconstituted NSG/ß2m(-/-) mice at doses as low as 0.5 µg/kg per day for ~7 days. MGD006 binds to human and cynomolgus monkey antigens with similar affinities and redirects T cells from either species to kill CD123-expressing target cells. MGD006 was well tolerated in monkeys continuously infused with 0.1 µg/kg per day escalated weekly to up to 1 µg/kg per day during a 4-week period. Depletion of circulating CD123-positive cells was observed as early as 72 hours after treatment initiation and persisted throughout the infusion period. Cytokine release, observed after the first infusion, was reduced after subsequent administrations, even when the dose was escalated. T cells from animals with prolonged in vivo exposure exhibited unperturbed target cell lysis ex vivo, indicating no exhaustion. A transient decrease in red cell mass was observed, with no neutropenia or thrombocytopenia. These studies support clinical testing of MGD006 in hematological malignancies, including AML.


CD3 Complex/metabolism , Interleukin-3 Receptor alpha Subunit/metabolism , Leukemia, Myeloid, Acute/immunology , T-Lymphocytes/immunology , Animals , Antigens, CD , Bone Marrow/pathology , Cell Death , Cell Proliferation , Cytokines/metabolism , Dose-Response Relationship, Immunologic , Female , Hematopoiesis , Humans , Leukemia, Myeloid, Acute/pathology , Lymphocyte Activation/immunology , Lymphocyte Count , Macaca fascicularis , Male , Mice , Protein Binding , Protein Engineering
12.
PLoS One ; 8(12): e79456, 2013.
Article En | MEDLINE | ID: mdl-24324581

There is increasing evidence that many solid tumors are hierarchically organized with the bulk tumor cells having limited replication potential, but are sustained by a stem-like cell that perpetuates the tumor. These cancer stem cells have been hypothesized to originate from transformation of adult tissue stem cells, or through re-acquisition of stem-like properties by progenitor cells. Adenosquamous carcinoma (ASC) is an aggressive type of lung cancer that contains a mixture of cells with squamous (cytokeratin 5+) and adenocarcinoma (cytokeratin 7+) phenotypes. The origin of these mixtures is unclear as squamous carcinomas are thought to arise from basal cells in the upper respiratory tract while adenocarcinomas are believed to form from stem cells in the bronchial alveolar junction. We have isolated and characterized cancer stem-like populations from ASC through application of selective defined culture medium initially used to grow human lung stem cells. Homogeneous cells selected from ASC tumor specimens were stably expanded in vitro. Primary xenografts and metastatic lesions derived from these cells in NSG mice fully recapitulate both the adenocarcinoma and squamous features of the patient tumor. Interestingly, while the CSLC all co-expressed cytokeratins 5 and 7, most xenograft cells expressed either one, or neither, with <10% remaining double positive. We also demonstrated the potential of the CSLC to differentiate to multi-lineage structures with branching lung morphology expressing bronchial, alveolar and neuroendocrine markers in vitro. Taken together the properties of these ASC-derived CSLC suggests that ASC may arise from a primitive lung stem cell distinct from the bronchial-alveolar or basal stem cells.


Biomarkers, Tumor/genetics , Carcinoma, Adenosquamous/pathology , Carcinoma, Squamous Cell/pathology , Keratin-5/genetics , Keratin-7/genetics , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , Adult , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Biomarkers, Tumor/metabolism , Bronchi/metabolism , Bronchi/pathology , Carcinoma, Adenosquamous/genetics , Carcinoma, Adenosquamous/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Differentiation , Cell Proliferation , Clone Cells , Gene Expression , Gene Expression Profiling , Humans , Keratin-5/metabolism , Keratin-7/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, SCID , Neoplastic Stem Cells/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Alveoli/pathology , Transplantation, Heterologous
13.
Clin Cancer Res ; 18(14): 3834-45, 2012 Jul 15.
Article En | MEDLINE | ID: mdl-22615450

PURPOSE: The goal of this research was to harness a monoclonal antibody (mAb) discovery platform to identify cell-surface antigens highly expressed on cancer and develop, through Fc optimization, potent mAb therapies toward these tumor-specific antigens. EXPERIMENTAL DESIGN: Fifty independent mAbs targeting the cell-surface immunoregulatory B7-H3 protein were obtained through independent intact cell-based immunizations using human tissue progenitor cells, cancer cell lines, or cell lines displaying cancer stem cell properties. Binding studies revealed this natively reactive B7-H3 mAb panel to bind a range of independent B7-H3 epitopes. Immunohistochemical analyses showed that a subset displayed strong reactivity to a broad range of human cancers while exhibiting limited binding to normal human tissues. A B7-H3 mAb displaying exquisite tumor/normal differential binding was selected for humanization and incorporation of an Fc domain modified to enhance effector-mediated antitumor function via increased affinity for the activating receptor CD16A and decreased binding to the inhibitory receptor CD32B. RESULTS: MGA271, the resulting engineered anti-B7-H3 mAb, mediates potent antibody-dependent cellular cytotoxicity against a broad range of tumor cell types. Furthermore, in human CD16A-bearing transgenic mice, MGA271 exhibited potent antitumor activity in B7-H3-expressing xenograft models of renal cell and bladder carcinoma. Toxicology studies carried out in cynomolgus monkeys revealed no significant test article-related safety findings. CONCLUSIONS: This data supports evaluation of MGA271 clinical utility in B7-H3-expressing cancer, while validating a combination of a nontarget biased approach of intact cell immunizations and immunohistochemistry to identify novel cancer antigens with Fc-based mAb engineering to enable potent antitumor activity.


Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/immunology , Antigens, Surface/immunology , B7 Antigens/immunology , Neoplasms , Animals , Cell Line, Tumor , Epitopes/immunology , Humans , Immunoglobulin Fc Fragments/immunology , Immunotherapy , Mice , Mice, Inbred BALB C , Mice, Transgenic , Neoplasms/drug therapy , Neoplasms/immunology
14.
Arch Pathol Lab Med ; 133(9): 1403-12, 2009 Sep.
Article En | MEDLINE | ID: mdl-19722746

CONTEXT: RAAG12 is a primate-restricted N-linked carbohydrate antigen present on multiple membrane-associated proteins. RAAG12 is recognized by the RAV12 monoclonal antibody. RAV12 binds to RAAG12-expressing gastrointestinal adenocarcinomas, modifies growth factor-mediated signaling, induces oncotic cell death in vitro, and has antitumor activity toward gastrointestinal tumor xenografts. OBJECTIVE: To determine the expression pattern of RAAG12 in normal and tumor tissue to identify indications for clinical study and potential safety issues. DESIGN: Immunohistochemistry of 36 normal human tissues and a broad range of tumor tissues to profile RAAG12 expression. RESULTS: More than 90% of colon, gastric, and pancreatic adenocarcinomas expressed RAAG12, and expression was uniform in most samples. Expression of RAAG12 at lower frequency and/or uniformity was observed in other cancers, including esophageal, ovarian, liver, breast, and prostate carcinomas and adenocarcinomas. Similar RAAG12 expression was observed between primary and metastatic colon adenocarcinomas. No staining was seen on cardiovascular, endocrine, neuromuscular, hematopoietic, or nervous system tissue from non-tumor-bearing individuals. RAAG12 was expressed on mucosal and glandular/ductal epithelium. The gastrointestinal tract mucosa and pancreatic/biliary ducts displayed the most uniform reactivity. RAAG12 exhibited differential subcellular localization in these normal, compared with tumor, tissues. Normal polarized epithelia primarily displayed apical membrane and cytoplasmic staining, whereas tumors exhibited whole membrane staining that increased with decreasing differentiation. CONCLUSIONS: High expression of RAAG12 on tumors of gastrointestinal origin suggests these cancers are appropriate targets for RAV12 therapy. Differential subcellular location of RAAG12 on normal epithelia may limit accessibility of RAV12 to the subset of normal tissues that exhibit antigen expression.


Adenocarcinoma/immunology , Antibodies, Monoclonal/immunology , Antigens, Tumor-Associated, Carbohydrate/immunology , Neoplasms/immunology , Animals , Antigens, Tumor-Associated, Carbohydrate/metabolism , Cell Line, Tumor , Female , Humans , Male , Mice , Mice, SCID , Xenograft Model Antitumor Assays
15.
Cancer Res ; 62(9): 2468-73, 2002 May 01.
Article En | MEDLINE | ID: mdl-11980634

Platelet-derived growth factor (PDGF) has been directly implicated in developmental and physiological processes, as well as in human cancer and other proliferative disorders. We have recently isolated and characterized a novel protease-activated member of the PDGF family, PDGF D. PDGF D has been shown to be proliferative for cells of mesenchymal origin, signaling through PDGF receptors. Comprehensive and systematic PDGF D transcript analysis revealed expression in many cell lines derived from ovarian, renal, and lung cancers, as well as from astrocytomas and medulloblastomas. beta PDGF receptor profiling further suggested autocrine signaling in several brain tumor cell lines. PDGF D transforming ability and tumor formation in SCID mice was further demonstrated. Exploiting a sensitive PDGF D sandwich ELISA using fully human monoclonal antibodies, PDGF D was detected at elevated levels in the sera of ovarian, renal, lung, and brain cancer patients. Immunohistochemical analysis confirmed PDGF D localization to ovarian and lung tumor tissues. Together, these data demonstrate that PDGF D plays a role in certain human cancers.


Neoplasms/metabolism , Platelet-Derived Growth Factor/physiology , 3T3 Cells , Animals , Cell Transformation, Neoplastic , Humans , Immunohistochemistry , Mice , Mice, SCID , Neoplasms/blood , Neoplasms/pathology , Phosphorylation , Platelet-Derived Growth Factor/biosynthesis , Platelet-Derived Growth Factor/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptor, Platelet-Derived Growth Factor beta/physiology , Signal Transduction/physiology , Tumor Cells, Cultured
...