Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Hum Vaccin Immunother ; 19(1): 2203632, 2023 12 31.
Article in English | MEDLINE | ID: mdl-37113012

ABSTRACT

Optimum formulation of Biological-E's protein subunit CORBEVAX™ vaccine was selected in phase-1 and -2 studies and found to be safe and immunogenic in healthy adult population. This is a phase-3 prospective, single-blinded, randomized, active controlled study conducted at 18 sites across India in 18-80 year-old subjects. This study has two groups; (i) immunogenicity-group, participants randomized either to CORBEVAX™ (n = 319) or COVISHIELD™ arms (n = 320). (ii) Safety-group containing single CORBEVAX™ arm (n = 1500) and randomization is not applicable. Healthy adults without a history of COVID-19 vaccination or SARS-CoV-2 infection were enrolled into immunogenicity arm and subjects seronegative to SARS-CoV-2 infection were enrolled into the safety arm. The safety profile of CORBEVAX™ vaccine was comparable to the comparator vaccine COVISHIELD™. Majority of reported AEs were mild in nature in both arms. The CORBEVAX™ to COVISHIELD™ GMT-ratios at day-42 time-point were 1·15 and 1·56 and the lower limit of the 95% confidence interval for the GMT-ratios was determined as 1·02 and 1·27 against Ancestral and Delta strains of SARS-COV-2 respectively. Both COVISHIELD™ and CORBEVAX™ vaccines showed comparable seroconversion post-vaccination against anti-RBD-IgG response. The subjects in CORBEVAX™ cohort also exhibited higher interferon-gamma secreting PBMC's post-stimulation with SARS-COV-2 RBD-peptides than subjects in COVISHIELD™ cohort.


Subject(s)
COVID-19 , Vaccines , Adult , Humans , Adolescent , Young Adult , Middle Aged , Aged , Aged, 80 and over , ChAdOx1 nCoV-19 , COVID-19 Vaccines/adverse effects , Leukocytes, Mononuclear , Prospective Studies , Single-Blind Method , COVID-19/prevention & control , SARS-CoV-2 , Immunogenicity, Vaccine , Antibodies, Viral , Antibodies, Neutralizing , Double-Blind Method
2.
Cancers (Basel) ; 15(7)2023 Mar 28.
Article in English | MEDLINE | ID: mdl-37046664

ABSTRACT

PURPOSE: HPV(-) OCSCC resists radiation treatment. The CDKN2A gene, encoding p16INK4A, is commonly disrupted in OCSCC. p16 inhibits CDK4/CDK6, leading to cell cycle arrest, but the biological sequelae of CDK4/6 inhibition in OCSCC remains understudied. This study examines whether inhibition of CDK4/6 enhances radiation response in OCSCC. METHODS: MTT assays were performed in OCSCC cell lines HN5 and CAL27 following treatment with palbociclib. Clonogenic survival and synergy were analyzed after radiation (RT-2 or 4Gy), palbociclib (P) (0.5 µM or 1 µM), or concurrent combination treatment (P+RT). DNA damage/repair and senescence were examined. CDK4/6 were targeted via siRNA to corroborate P+RT effects. Three-dimensional immortalized spheroids and organoids derived from patient tumors (conditionally reprogrammed OCSCC CR-06 and CR-18) were established to further examine and validate responses to P+RT. RESULTS: P+RT demonstrated reduced viability and synergy, increased ß-gal expression (~95%), and ~two-fold higher γH2AX. Rad51 and Ku80 were reduced after P+RT, indicating impairment of both HR and NHEJ. siCDK4/6 increased senescence with radiation. Spheroids showed reduced proliferation and size with P+RT. CR-06 and CR-18 further demonstrated three-fold reduced proliferation and organoids size with P+RT. CONCLUSION: Targeting CDK4/6 can lead to improved efficacy when combined with radiation in OCSCC by inducing senescence and inhibiting DNA damage repair.

3.
Sci Rep ; 11(1): 8844, 2021 04 23.
Article in English | MEDLINE | ID: mdl-33893330

ABSTRACT

A mouse model of human Familial Adenomatous Polyposis responds favorably to pharmacological inhibition of 5'-methylthioadenosine phosphorylase (MTAP). Methylthio-DADMe-Immucillin-A (MTDIA) is an orally available, transition state analogue inhibitor of MTAP. 5'-Methylthioadenosine (MTA), the substrate for MTAP, is formed in polyamine synthesis and is recycled by MTAP to S-adenosyl-L-methionine (SAM) via salvage pathways. MTDIA treatment causes accumulation of MTA, which inhibits growth of human head and neck (FaDu) and lung (H359, A549) cancers in immunocompromised mouse models. We investigated the efficacy of oral MTDIA as an anti-cancer therapeutic for intestinal adenomas in immunocompetent APCMin/+ mice, a murine model of human Familial Adenomatous Polyposis. Tumors in APCMin/+ mice were decreased in size by MTDIA treatment, resulting in markedly improved anemia and doubling of mouse lifespan. Metabolomic analysis of treated mice showed no changes in polyamine, methionine, SAM or ATP levels when compared with control mice but indicated an increase in MTA, the MTAP substrate. Generation of an MTDIA-resistant cell line in culture showed a four-fold amplification of the methionine adenosyl transferase (MAT2A) locus and expression of this enzyme. MAT2A is downstream of MTAP action and catalyzes synthesis of the SAM necessary for methylation reactions. Immunohistochemical analysis of treated mouse intestinal tissue demonstrated a decrease in symmetric dimethylarginine, a PRMT5-catalyzed modification. The anti-cancer effects of MTDIA indicate that increased cellular MTA inhibits PRMT5-mediated methylations resulting in attenuated tumor growth. Oral dosing of MTDIA as monotherapy has potential for delaying the onset and progression of colorectal cancers in Familial Adenomatous Polyposis (FAP) as well as residual duodenal tumors in FAP patients following colectomy. MTDIA causes a physiologic inactivation of MTAP and may also have efficacy in combination with inhibitors of MAT2A or PRMT5, known synthetic-lethal interactions in MTAP-/- cancer cell lines.


Subject(s)
Genes, APC , Longevity/genetics , Purine-Nucleoside Phosphorylase/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Adenine/therapeutic use , Adenine/toxicity , Adenomatous Polyposis Coli/drug therapy , Adenomatous Polyposis Coli/enzymology , Adenomatous Polyposis Coli/genetics , Animals , Disease Models, Animal , Metabolomics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein-Arginine N-Methyltransferases/metabolism , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/genetics , Pyrrolidines/pharmacology , Pyrrolidines/therapeutic use , Pyrrolidines/toxicity , Survival Analysis
4.
EBioMedicine ; 17: 30-44, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28233730

ABSTRACT

Programmed Cell Death-1 (PD-1) is an inhibitory immune receptor, which plays critical roles in T cell co-inhibition and exhaustion upon binding to its ligands PD-L1 and PD-L2. We report the crystal structure of the human PD-1 ectodomain and the mapping of the PD-1 binding interface. Mutagenesis studies confirmed the crystallographic interface, and resulted in mutant PD-1 receptors with altered affinity and ligand-specificity. In particular, a high-affinity mutant PD-1 (HA PD-1) exhibited 45 and 30-fold increase in binding to PD-L1 and PD-L2, respectively, due to slower dissociation rates. This mutant (A132L) was used to engineer a soluble chimeric Ig fusion protein for cell-based and in vivo studies. HA PD-1 Ig showed enhanced binding to human dendritic cells, and increased T cell proliferation and cytokine production in a mixed lymphocyte reaction (MLR) assay. Moreover, in an experimental model of murine Lewis lung carcinoma, HA PD-1 Ig treatment synergized with radiation therapy to decrease local and metastatic tumor burden, as well as in the establishment of immunological memory responses. Our studies highlight the value of structural considerations in guiding the design of a high-affinity chimeric PD-1 Ig fusion protein with robust immune modulatory properties, and underscore the power of combination therapies to selectively manipulate the PD-1 pathway for tumor immunotherapy.


Subject(s)
Carcinoma, Lewis Lung/therapy , Immunotherapy/methods , Programmed Cell Death 1 Receptor/chemistry , Animals , Cell Line, Tumor , Cells, Cultured , Cytokines/metabolism , Dendritic Cells/metabolism , Female , HEK293 Cells , Humans , Immunoglobulins/genetics , Immunoglobulins/immunology , Immunologic Memory , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/immunology , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Protein Domains , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology
5.
Cell ; 165(3): 643-55, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27104980

ABSTRACT

Oncogenic activation of RAS genes via point mutations occurs in 20%-30% of human cancers. The development of effective RAS inhibitors has been challenging, necessitating new approaches to inhibit this oncogenic protein. Functional studies have shown that the switch region of RAS interacts with a large number of effector proteins containing a common RAS-binding domain (RBD). Because RBD-mediated interactions are essential for RAS signaling, blocking RBD association with small molecules constitutes an attractive therapeutic approach. Here, we present evidence that rigosertib, a styryl-benzyl sulfone, acts as a RAS-mimetic and interacts with the RBDs of RAF kinases, resulting in their inability to bind to RAS, disruption of RAF activation, and inhibition of the RAS-RAF-MEK pathway. We also find that ribosertib binds to the RBDs of Ral-GDS and PI3Ks. These results suggest that targeting of RBDs across multiple signaling pathways by rigosertib may represent an effective strategy for inactivation of RAS signaling.


Subject(s)
Glycine/analogs & derivatives , RNA-Binding Proteins/chemistry , Signal Transduction/drug effects , Sulfones/pharmacology , Amino Acid Sequence , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/drug effects , Crystallography, X-Ray , Dimerization , Glycine/administration & dosage , Glycine/chemistry , Glycine/pharmacology , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Models, Molecular , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Pancreatic Neoplasms/drug therapy , Phosphorylation , Protein Serine-Threonine Kinases/chemistry , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins B-raf/chemistry , Proto-Oncogene Proteins B-raf/metabolism , RNA-Binding Proteins/metabolism , Sequence Alignment , Sulfones/administration & dosage , Sulfones/chemistry , ras Proteins/metabolism , Polo-Like Kinase 1
6.
J Immunol ; 196(4): 1964-76, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26755821

ABSTRACT

Immune responses against cancer cells are often hindered by immunosuppressive mechanisms that are developed in the tumor microenvironment. Induction of a hyporesponsive state in tumor Ag-specific T cells is one of the major events responsible for the inability of the adaptive immune system to mount an efficient antitumor response and frequently contributes to lessen the efficacy of immunotherapeutic approaches. Treatment of localized tumors by focused ultrasound (FUS) is a minimally invasive therapy that uses a range of input energy for in situ tumor ablation through the generation of thermal and cavitation effect. Using a murine B16 melanoma tumor model, we show that a variant of FUS that delivers a reduced level of energy at the focal point and generates mild mechanical and thermal stress in target cells has the ability to increase immunogenic presentation of tumor Ags, which results in reversal of tumor-induced T cell tolerance. Furthermore, we show that the combination of nonablative low-energy FUS with an ablative hypofractionated radiation therapy results in synergistic control of primary tumors and leads to a dramatic reduction in spontaneous pulmonary metastases while prolonging recurrence-free survival only in immunocompetent mice.


Subject(s)
Melanoma, Experimental/immunology , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Tumor Escape/immunology , Ultrasonic Therapy/methods , Animals , Combined Modality Therapy , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Melanoma, Experimental/pathology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis/pathology , Neoplasm Metastasis/therapy , Radiotherapy , Real-Time Polymerase Chain Reaction , Skin Neoplasms/pathology , Skin Neoplasms/therapy
7.
Int J Radiat Oncol Biol Phys ; 88(5): 1180-7, 2014 Apr 01.
Article in English | MEDLINE | ID: mdl-24529717

ABSTRACT

PURPOSE: To compare rigosertib versus cisplatin as an effective radiosensitizing agent for cervical malignancies. METHODS AND MATERIALS: Rigosertib and cisplatin were tested in cervical cancer cell lines, HeLa and C33A. A 24-hour incubation with rigosertib and cisplatin, before irradiation (2-8 Gy), was used for clonogenic survival assays. Cell cycle analysis (propidium iodide staining) and DNA damage (γ-H2AX expression) were evaluated by fluorescence-activated cell sorter cytometry. Rigosertib was also tested in vivo in tumor growth experiments on cervical cancer xenografts. RESULTS: Rigosertib was demonstrated to induce a G2/M block in cancer cells. Survival curve comparison revealed a dose modification factor, as index of radiosensitization effect, of 1.1-1.3 for cisplatin and 1.4-2.2 for rigosertib. With 6-Gy irradiation, an increase in DNA damage of 15%-25% was achieved in both HeLa and C33A cells with cisplatin pretreatment, and a 71-108% increase with rigosertib pretreatment. In vivo tumor growth studies demonstrated higher performance of rigosertib when compared with cisplatin, with 53% longer tumor growth delay. CONCLUSIONS: Rigosertib was more effective than cisplatin when combined with radiation and caused minimal toxicity. These data support the need for clinical trials with rigosertib in combination therapy for patients with cervical carcinoma.


Subject(s)
Chemoradiotherapy/methods , Glycine/analogs & derivatives , Radiation-Sensitizing Agents/therapeutic use , Sulfones/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Animals , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Separation , Cell Survival , Cisplatin/therapeutic use , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair , Dose-Response Relationship, Radiation , Female , Flow Cytometry , Glycine/therapeutic use , HeLa Cells , Humans , Inhibitory Concentration 50 , Mice , Mice, Nude , Mitosis/drug effects , Mitosis/radiation effects , Neoplasm Transplantation , Time Factors
8.
J Biol Chem ; 286(6): 4902-11, 2011 Feb 11.
Article in English | MEDLINE | ID: mdl-21135097

ABSTRACT

The S-adenosylmethionine (AdoMet) salvage enzyme 5'-methylthioadenosine phosphorylase (MTAP) has been implicated as both a cancer target and a tumor suppressor. We tested these hypotheses in mouse xenografts of human lung cancers. AdoMet recycling from 5'-methylthioadenosine (MTA) was blocked by inhibition of MTAP with methylthio-DADMe-Immucillin-A (MTDIA), an orally available, nontoxic, picomolar transition state analogue. Blood, urine, and tumor levels of MTA increased in response to MTDIA treatment. MTDIA treatment inhibited A549 (human non-small cell lung carcinoma) and H358 (human bronchioloalveolar non-small cell lung carcinoma cells) xenograft tumor growth in immunodeficient Rag2(-/-)γC(-/-) and NCr-nu mice. Systemic MTA accumulation is implicated as the tumor-suppressive metabolite because MTDIA is effective for in vivo treatment of A549 MTAP(-/-) and H358 MTAP(+/+) tumors. Tumors from treated mice showed increased MTA and decreased polyamines but little alteration in AdoMet, methionine, or adenine levels. Gene expression profiles of A549 tumors from treated and untreated mice revealed only modest alterations with 62 up-regulated and 63 down-regulated mRNAs (≥ 3-fold). MTDIA antitumor activity in xenografts supports MTAP as a target for lung cancer therapy.


Subject(s)
Adenine/analogs & derivatives , Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/enzymology , Lung Neoplasms/drug therapy , Lung Neoplasms/enzymology , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Pyrrolidines/pharmacology , Tumor Suppressor Proteins/antagonists & inhibitors , Adenine/pharmacology , Adenosine/analogs & derivatives , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Deoxyadenosines/metabolism , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Neoplasm Transplantation , Purine-Nucleoside Phosphorylase/genetics , Purine-Nucleoside Phosphorylase/metabolism , S-Adenosylmethionine/genetics , S-Adenosylmethionine/metabolism , Thionucleosides/metabolism , Transplantation, Heterologous , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays/methods
9.
Nucleic Acids Symp Ser (Oxf) ; (52): 75-6, 2008.
Article in English | MEDLINE | ID: mdl-18776260

ABSTRACT

Transition state structures can be derived from kinetic isotope effects and computational chemistry. Molecular electrostatic potential maps of transition states serve as blueprints to guide synthesis of transition state analogue inhibitors of target enzymes. 5'- Methylthioadenosine phosphorylase (MTAP) functions in the polyamine pathway by recycling methylthioadenosine (MTA) and maintaining cellular S-adenosylmethionine (SAM). Its transition state structure was used to guide synthesis of MT-DADMe-ImmA, a picomolar inhibitor that shows anticancer effects against solid tumors. Biochemical and genomic analysis suggests that MTAP inhibition acts by altered DNA methylation and gene expression patterns. A related bacterial enzyme, 5'-methylthioadenosine nucleosidase (MTAN), functions in pathways of quorum sensing involving AI-1 and AI-2 molecules. Transition states have been solved for several bacterial MTANs and used to guide synthesis of powerful inhibitors with dissociation constants in the femtomolar to picomolar range. BuT-DADMe-ImmA blocks quorum sensing in Vibrio cholerae without changing bacterial growth rates. Transition state analogue inhibitors show promise as anticancer and antibacterial agents.


Subject(s)
Adenine/analogs & derivatives , Anti-Bacterial Agents/chemistry , Antineoplastic Agents/chemistry , N-Glycosyl Hydrolases/antagonists & inhibitors , Purine-Nucleoside Phosphorylase/antagonists & inhibitors , Quorum Sensing/drug effects , Adenine/chemistry , Adenine/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Head and Neck Neoplasms/enzymology , Humans , Male , Mice , Pyrrolidines/chemistry , Pyrrolidines/pharmacology , S-Adenosylmethionine/metabolism , Xenograft Model Antitumor Assays
10.
J Biol Chem ; 282(29): 21477-86, 2007 Jul 20.
Article in English | MEDLINE | ID: mdl-17548352

ABSTRACT

Methylthio-DADMe-immucillin-A (MT-DADMe-ImmA) is an 86-pm inhibitor of human 5'-methylthioadenosine phosphorylase (MTAP). The sole function of MTAP is to recycle 5'-methylthioadenosine (MTA) to S-adenosylmethionine. Treatment of cultured cells with MT-DADMe-ImmA and MTA inhibited MTAP, increased cellular MTA concentrations, decreased polyamines, and induced apoptosis in FaDu and Cal27, two head and neck squamous cell carcinoma cell lines. The same treatment did not induce apoptosis in normal human fibroblast cell lines (CRL2522 and GM02037) or in MCF7, a breast cancer cell line with an MTAP gene deletion. MT-DADMe-ImmA alone did not induce apoptosis in any cell line, implicating MTA as the active agent. Treatment of sensitive cells caused loss of mitochondrial inner membrane potential, G(2)/M arrest, activation of mitochondria-dependent caspases, and apoptosis. Changes in cellular polyamines and MTA levels occurred in both responsive and nonresponsive cells, suggesting cell-specific epigenetic effects. A survey of aberrant DNA methylation in genomic DNA using a microarray of 12,288 CpG island clones revealed decreased CpG island methylation in treated FaDu cells compared with untreated cells. FaDu tumors in a mouse xenograft model were treated with MT-DADMe-ImmA, resulting in tumor remission. The selective action of MT-DADMe-ImmA on head and neck squamous cell carcinoma cells suggests potential as an agent for treatment of cancers sensitive to reduced CpG island methylation.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Head and Neck Neoplasms/enzymology , Purine-Nucleoside Phosphorylase/chemistry , Animals , CpG Islands , Epigenesis, Genetic , Gene Deletion , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/metabolism , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Models, Chemical , Neoplasm Transplantation , Purine-Nucleoside Phosphorylase/pharmacology
11.
Cancer Res ; 64(4): 1377-85, 2004 Feb 15.
Article in English | MEDLINE | ID: mdl-14973061

ABSTRACT

Re-expression of a tumor suppressor in tumor cells that lack it is an effective way to study its functional activities. However, because tumor cells contain multiple mutations, tumor suppressor functions that are dependent on (an)other regulators are unlikely to be identified by its re-expression alone if the other regulators are also mutated. In this study, we show that re-expression of retinoblastoma (RB) together with the androgen receptor (AR) in RB- and AR-deficient prostate cancer DU-145 cells resulted in an apoptotic activity, acting through the mitochondria damage-initiated caspase activation pathway, which was not present when RB, or the AR, was re-expressed alone. The ability of RB + AR to induce mitochondria damage was dependent on the proapoptotic proteins Bax and Bak and could be blocked by the antiapoptotic protein Bcl-x(L). Coexpressed AR did not detectably change RB's regulation of E2F and cell cycle progression in culture. On the other hand, coexpressed RB could activate the transactivation activity of the AR in an androgen-depleted media. Although androgen induced greater AR transactivation activity in this condition, it did not induce apoptosis in the absence of coexpressed RB. Analysis of mutants of RB and the AR indicated that intact pocket function of RB and the transactivation activity of the AR were required for RB + AR-induced apoptosis. These results provide direct functional data for an AR-dependent apoptosis-inducing activity of RB and highlight the importance of cell type-specific regulators in obtaining a more complete understanding of RB.


Subject(s)
Apoptosis , Prostatic Neoplasms/pathology , Receptors, Androgen/physiology , Retinoblastoma Protein/physiology , Cell Line, Tumor , Humans , Male , Mitochondria/physiology , Transcriptional Activation , Tumor Suppressor Protein p53/physiology
12.
Int J Cancer ; 105(3): 377-83, 2003 Jun 20.
Article in English | MEDLINE | ID: mdl-12704672

ABSTRACT

A male breast tumor associated antigen (MBTAA) was purified and partially characterized from human male breast tumor. Three protein peaks were obtained by DEAE-cellulose column chromatography of a crude extract of human male breast tumor tissues. Circulating antibodies against one of these peaks, MF1, which contained MBTAA, were observed in male breast cancer patients but not in normal male or male patients with carcinoma of other organs (stomach, colon, lung). The MBTAA was partially purified from MF1 by subjecting the fraction to SDS-PAGE and eluting the protein from band 3 (MB-3) and by subjecting MF1 to size exclusion-high performance liquid chromatography (SE-HPLC). The MBTAA was characterized as a glycoprotein with MW of approximately 72 kDa. It showed no immunological relatedness with TAG-72, a tumor associated antigen expressed in breast epithelial cells. A 72 kDa protein, immunologically related to MBTAA, was detected and partially purified from female breast tumor. The female breast cancer patients did not have circulating antibodies against this 72 kDa protein or MBTAA. Presence of 72 kDa glycoprotein MBTAA in MF1 and specificity of the anti-MBTAA antibodies in the sera of male breast cancer patients were further confirmed by Western blot analysis. Absence of anti-MBTAA antibodies in healthy men and in patients with other cancers suggested that expression of MBTAA may be malignancy-associated and is highly overexpressed in male breast cancer.


Subject(s)
Antigens, Neoplasm/chemistry , Antigens, Neoplasm/isolation & purification , Breast Neoplasms, Male/metabolism , Antibodies, Monoclonal/metabolism , Blotting, Western , Breast Neoplasms, Male/immunology , Chromatography, DEAE-Cellulose , Chromatography, High Pressure Liquid , Electrophoresis, Polyacrylamide Gel , Enzyme-Linked Immunosorbent Assay , Glycoproteins/chemistry , Humans , Male
SELECTION OF CITATIONS
SEARCH DETAIL
...