Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 27(5): 1505-1515, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33219015

ABSTRACT

PURPOSE: The use of high-dose per fraction radiotherapy delivered as stereotactic body radiotherapy is a standard of care for prostate cancer. It is hypothesized that high-dose radiotherapy may enhance or suppress tumor-reactive immunity. The objective of this study was to assess both antitumor and immunosuppressive effects induced by high-dose radiotherapy in prostate cancer coclinical models, and ultimately, to test whether a combination of radiotherapy with targeted immunotherapy can enhance antitumor immunity. EXPERIMENTAL DESIGN: We studied the effects of high-dose per fraction radiotherapy with and without anti-Gr-1 using syngeneic murine allograft prostate cancer models. The dynamic change of immune populations, including tumor-infiltrating lymphocytes (TIL), T regulatory cells (Treg), and myeloid-derived suppressive cells (MDSC), was evaluated using flow cytometry and IHC. RESULTS: Coclinical prostate cancer models demonstrated that high-dose per fraction radiotherapy induced a rapid increase of tumor-infiltrating MDSCs and a subsequent rise of CD8 TILs and circulating CD8 T effector memory cells. These radiation-induced CD8 TILs were more functionally potent than those from nonirradiated controls. While systemic depletion of MDSCs by anti-Gr-1 effectively prevented MDSC tumor infiltration, it did not enhance radiotherapy-induced antitumor immunity due to a compensatory expansion of Treg-mediated immune suppression. CONCLUSIONS: In allograft prostate cancer models, high-dose radiotherapy induced an early rise of MDSCs, followed by a transient increase of functionally active CD8 TILs. However, systemic depletion of MDSC did not augment the antitumor efficacy of high-dose radiotherapy due to a compensatory Treg response, indicating blocking both MDSCs and Tregs might be necessary to enhance radiotherapy-induced antitumor immunity.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Gamma Rays/therapeutic use , Immunosuppressive Agents/therapeutic use , Lymphocytes, Tumor-Infiltrating/immunology , Myeloid-Derived Suppressor Cells/immunology , Prostatic Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , Cell Proliferation , Humans , Male , Mice , Mice, Inbred C57BL , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Clin Cancer Res ; 25(1): 188-200, 2019 01 01.
Article in English | MEDLINE | ID: mdl-30301826

ABSTRACT

PURPOSE: The inability to intraoperatively distinguish primary tumor, as well as lymphatic spread, increases the probability of positive surgical margins, tumor recurrence, and surgical toxicity. The goal of this study was to develop a tumor-specific optical probe for real-time fluorescence-guided surgery. EXPERIMENTAL DESIGN: A humanized antibody fragment against PSCA (A11 minibody, A11 Mb) was conjugated with a near-infrared fluorophore, IRDye800CW. The integrity and binding of the probe to PSCA were confirmed by gel electrophoresis, size-exclusion chromatography, and flow cytometry, respectively. The ability of the probe to detect tumor-infiltrated lymph nodes and metastatic lesions was evaluated in 2 xenograft models, as well as in transgenic mice expressing human PSCA (hPSCA). An invasive intramuscular model was utilized to evaluate the efficacy of the A11 Mb-IRDye800CW-guided surgery. RESULTS: A11 Mb was successfully conjugated with IRDye800CW and retained specific binding to PSCA. In vivo imaging showed maximal signal-to-background ratios at 48 hours. The A11 Mb-IRDye800CW specifically detected PSCA-positive primary tumors, tumor-infiltrated lymph nodes, and distant metastases with high contrast. Fluorescence guidance facilitated more complete tumor resection, reduced tumor recurrence, and improved overall survival, compared with conventional white light surgery. The probe successfully identified primary orthotopic tumors and metastatic lesions in hPSCA transgenic mice. CONCLUSIONS: Real-time fluorescence image-guided surgery with A11 Mb-IRDye800CW enabled detection of lymph node metastases and positive surgical margins, facilitated more complete tumor removal, and improved survival, compared with white light surgery. These results may be translatable into clinical practice to improve surgical and patient outcomes.


Subject(s)
Antigens, Surface/genetics , Glutamate Carboxypeptidase II/genetics , Indoles/pharmacology , Prostatic Neoplasms/diagnostic imaging , Surgery, Computer-Assisted , Animals , Antigens, Surface/isolation & purification , Cell Line, Tumor , Disease Models, Animal , Fluorescence , Gene Expression Regulation, Neoplastic/genetics , Glutamate Carboxypeptidase II/isolation & purification , Heterografts , Humans , Infrared Rays , Male , Margins of Excision , Mice , Optical Imaging , Prostate/surgery , Prostatectomy/methods , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/surgery , Spectroscopy, Near-Infrared
3.
Prostate ; 73(13): 1378-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23728788

ABSTRACT

BACKGROUND: Prostate cancer stem cells (PCSC) offer theoretical explanations to many clinical and biological behaviors of the disease in human. In contrast to approaches of using side populations and cell-surface markers to isolate and characterize the putative PCSC, we hypothesize that androgen deprivation leads to functional enrichment of putative PCSC. METHODS AND RESULTS: Human prostate cancer lines LNCaP, LAPC4 and LAPC9 were depleted of androgen in cell cultures and in castrated SCID mice. The resultant androgen deprivation-resistant or castration-resistant populations, in particular in LNCaP and its derivative cell lines, displayed increased expression of pluripotency transactivators and significantly higher tumorigenicity. Individual tumor cell clones were isolated from castration-resistant bulk cultures of LNCaP (CR-LNCaP) and tested for tumorigenicity in male SCID mice under limiting dilution conditions. As few as 200 cells were able to form spheres in vitro, and generate tumors with similar growth kinetics as 10(6) LNCaP or 10(4) CR-LNCaP cells in vivo. These putative PCSC were CD44(+) /CD24(-) and lack the expression of prostate lineage proteins. When transplanted into the prostate of an intact male SCID mouse, these putative PCSC seemed to show limited differentiation into Ck5(+) , Ck8(+) , Ck5(+) /Ck8(+) , and AR(+) cells. On the other hand, stable transduction of LNCaP with retrovirus encoding Sox2 led to androgen-deprivation resistant growth and down-regulation of major prostate lineage gene products in vitro. CONCLUSION: Concurrence of overexpression of pluripotency transactivators and resistance to androgen deprivation supported the role of putative PCSC in the emergence of prostate cancer resistant to androgen deprivation.


Subject(s)
Androgens/metabolism , Neoplastic Stem Cells/pathology , Prostate/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Up-Regulation , Animals , Castration , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, SCID , Neoplastic Stem Cells/metabolism , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics
4.
Nat Med ; 16(12): 1414-20, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21057494

ABSTRACT

The transition from androgen-dependent to castration-resistant prostate cancer (CRPC) is a lethal event of uncertain molecular etiology. Comparing gene expression in isogenic androgen-dependent and CRPC xenografts, we found a reproducible increase in N-cadherin expression, which was also elevated in primary and metastatic tumors of individuals with CRPC. Ectopic expression of N-cadherin in nonmetastatic, androgen-dependent prostate cancer models caused castration resistance, invasion and metastasis. Monoclonal antibodies against the ectodomain of N-cadherin reduced proliferation, adhesion and invasion of prostate cancer cells in vitro. In vivo, these antibodies slowed the growth of multiple established CRPC xenografts, blocked local invasion and metastasis and, at higher doses, led to complete regression. N-cadherin-specific antibodies markedly delayed the time to emergence of castration resistance, markedly affected tumor histology and angiogenesis, and reduced both AKT serine-threonine kinase activity and serum interleukin-8 (IL-8) secretion. These data indicate that N-cadherin is a major cause of both prostate cancer metastasis and castration resistance. Therapeutic targeting of this factor with monoclonal antibodies may have considerable clinical benefit.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cadherins/immunology , Gene Expression Regulation, Neoplastic/drug effects , Immunotherapy/methods , Neoplasm Metastasis/prevention & control , Prostatic Neoplasms/immunology , Prostatic Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Blotting, Western , Cadherins/genetics , Cell Line, Tumor , Disease Progression , Enzyme-Linked Immunosorbent Assay , Gene Knockdown Techniques , Humans , Immunohistochemistry , Interleukin-8/metabolism , Male , Mice , Mice, SCID
5.
Cancer Res ; 65(20): 9495-500, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16230414

ABSTRACT

Prostate stem cell antigen (PSCA), a 123-amino acid cell surface glycoprotein, is highly expressed in both local and metastatic prostate cancers as well as in a large proportion of bladder and pancreatic cancers. PSCA overexpression correlates with a high risk of recurrence after primary therapy for prostate cancer. We have reported previously that anti-PSCA monoclonal antibody (mAb) 1G8 inhibits tumor growth, prevents metastasis, and prolongs the survival of mice inoculated with human prostate cancer cell lines and xenografts. The current study was undertaken to elucidate the mechanism of action of anti-PSCA antibody therapy. In particular, we asked whether antitumor activity resulted from recruitment of an immune response or a direct effect on the tumor cell itself. In vitro assays show that both intact 1G8 and F(ab')2 fragments of 1G8 induce prostate cancer cell death. The anti-PSCA antibody-induced cell death is caspase independent and requires antigen cross-linking. These results were confirmed in in vivo models in which both 1G8 and F(ab')2 fragments were able to inhibit prostate tumor formation and growth equally. These results suggest that the anti-PSCA mAb 1G8 acts by a direct, Fc-independent mechanism to inhibit prostate tumor growth both in vitro and in vivo.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunoglobulin Fragments/pharmacology , Membrane Glycoproteins/immunology , Neoplasm Proteins/immunology , Prostatic Neoplasms/therapy , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm , Caspases/metabolism , Cell Death/drug effects , Cell Death/immunology , Cell Growth Processes/drug effects , Cell Growth Processes/immunology , Enzyme-Linked Immunosorbent Assay , GPI-Linked Proteins , Immunoglobulin Fragments/immunology , Male , Mice , Mice, SCID , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Xenograft Model Antitumor Assays
6.
Clin Cancer Res ; 11(7): 2591-6, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15814638

ABSTRACT

PURPOSE: Prostate stem cell antigen (PSCA) is expressed by a majority of prostate cancers and is a promising therapeutic target. PSCA protein and mRNA expression was examined in prostate cancer bone, lymph node, and visceral metastases to assess the potential of PSCA as an immunotherapeutic target in advanced prostate cancer. EXPERIMENTAL DESIGN: Immunohistochemical analysis of PSCA protein expression and quantitative mRNA expression analysis of PSCA was done on clinical specimens of prostate cancer bone, lymph node, and visceral metastases. PSCA protein and mRNA expression levels were quantified and compared between available matched pairs of bone and lymph node or visceral metastases. RESULTS: Bone metastases stained with higher intensity of PSCA compared with lymph node or liver metastases in seven of eight (87.5%) matched pairs (P = 0.035). PSCA mRNA expression was equal or greater than that of LAPC-9, a PSCA expressing xenograft, in 12 of 24 (50%) cases of prostate cancer metastases and was significantly correlated with PSCA protein expression (sigma = 0.84, P = 0.0019). Overall, PSCA protein expression was detected in 41 of 47 (87.2%), four of six (66.7%), and two of three (66.7%) cases of bone, lymph node, and liver metastases, respectively. Mean PSCA staining intensity was significantly higher in prostate cancer bone metastases compared with lymph node metastases (2.0 +/- 0.02 versus 0.83 +/- 0.31, P = 0.014). CONCLUSIONS: Prostate cancer metastases express PSCA. However, greater PSCA staining intensity and level of PSCA mRNA expression was associated with bone metastases compared with lymph node metastases. This study suggests that PSCA is a promising tumor marker and potential therapeutic target for patients with metastatic prostate cancer.


Subject(s)
Bone Neoplasms/secondary , Liver Neoplasms/secondary , Lymph Nodes/pathology , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Prostatic Neoplasms/pathology , Antigens, Neoplasm , Bone Neoplasms/genetics , Bone Neoplasms/metabolism , GPI-Linked Proteins , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Lymph Nodes/chemistry , Lymph Nodes/metabolism , Lymphatic Metastasis , Male , Membrane Glycoproteins/analysis , Neoplasm Proteins/analysis , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
7.
Mol Cancer Res ; 1(2): 113-21, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12496358

ABSTRACT

Prostate stem cell antigen (PSCA, named for its strong sequence homology to the thymocyte marker stem cell antigen 2) is a cell surface antigen expressed in normal prostate and associated with human and murine prostate cancer. To begin to investigate a possible link between PSCA expression in normal prostate and prostate carcinogenesis, we characterized the phenotype and proliferative behavior of normal PSCA-expressing prostate epithelial cells (PrEC) in tissue culture. PSCA was expressed in a subset of prostate epithelial cells that coexpress basal and secretory cytokeratins. PSCA-positive cells were the direct progeny of PSCA-negative cells and were characterized by a more differentiated morphology and a slower proliferative rate than PSCA-negative cells. Although PSCA-positive cells continued to express basal cell markers such as CD44, they lost expression of the basal cell marker p63. In contrast, expression of prostate specific antigen and androgen receptor transcripts was detectable in PSCA-positive PrEC. These findings suggest that PSCA is a unique marker of an intermediate subpopulation of PrEC in transition from a basal to a terminally differentiated secretory phenotype and may be a useful marker for the study of normal and malignant prostate development.


Subject(s)
Epithelial Cells/metabolism , Membrane Glycoproteins/metabolism , Membrane Proteins , Neoplasm Proteins/metabolism , Prostate/metabolism , Antigens, Neoplasm , Cell Division , Cell Separation , DNA-Binding Proteins , Flow Cytometry , GPI-Linked Proteins , Genes, Tumor Suppressor , Humans , Hyaluronan Receptors/biosynthesis , Male , Microscopy, Fluorescence , Phenotype , Phosphoproteins/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Trans-Activators/biosynthesis , Transcription Factors , Tumor Suppressor Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...