Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
J Clin Invest ; 128(11): 5137-5149, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30188869

ABSTRACT

Despite the success of immune checkpoint blockade against melanoma, many "cold" tumors like prostate cancer remain unresponsive. We found that hypoxic zones were prevalent across preclinical prostate cancer and resisted T cell infiltration even in the context of CTLA-4 and PD-1 blockade. We demonstrated that the hypoxia-activated prodrug TH-302 reduces or eliminates hypoxia in these tumors. Combination therapy with this hypoxia-prodrug and checkpoint blockade cooperated to cure more than 80% of tumors in the transgenic adenocarcinoma of the mouse prostate-derived (TRAMP-derived) TRAMP-C2 model. Immunofluorescence imaging showed that TH-302 drives an influx of T cells into hypoxic zones, which were expanded by checkpoint blockade. Further, combination therapy reduced myeloid-derived suppressor cell density by more than 50%, and durably reduced the capacity of the tumor to replenish the granulocytic subset. Spontaneous prostate tumors in TRAMP transgenic mice, which completely resist checkpoint blockade, showed minimal adenocarcinoma tumor burden at 36 weeks of age and no evidence of neuroendocrine tumors with combination therapy. Survival of Pb-Cre4, Ptenpc-/-Smad4pc-/- mice with aggressive prostate adenocarcinoma was also significantly extended by this combination of hypoxia-prodrug and checkpoint blockade. Hypoxia disruption and T cell checkpoint blockade may sensitize some of the most therapeutically resistant cancers to immunotherapy.


Subject(s)
Adenocarcinoma/therapy , Immunotherapy , Neoplasms, Experimental/therapy , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Prostatic Neoplasms/therapy , T-Lymphocytes/immunology , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Animals , Cell Hypoxia/genetics , Cell Hypoxia/immunology , Cell Line, Tumor , Male , Mice , Mice, Knockout , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasms, Experimental/genetics , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , T-Lymphocytes/pathology
2.
Clin Cancer Res ; 24(5): 1138-1151, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29301830

ABSTRACT

Purpose: Agonist antibodies targeting the T-cell costimulatory receptor 4-1BB (CD137) are among the most effective immunotherapeutic agents across preclinical cancer models. In the clinic, however, development of these agents has been hampered by dose-limiting liver toxicity. Lack of knowledge of the mechanisms underlying this toxicity has limited the potential to separate 4-1BB agonist-driven tumor immunity from hepatotoxicity.Experimental Design: The capacity of 4-1BB agonist antibodies to induce liver toxicity was investigated in immunocompetent mice, with or without coadministration of checkpoint blockade, via (i) measurement of serum transaminase levels, (ii) imaging of liver immune infiltrates, and (iii) qualitative and quantitative assessment of liver myeloid and T cells via flow cytometry. Knockout mice were used to clarify the contribution of specific cell subsets, cytokines, and chemokines.Results: We find that activation of 4-1BB on liver myeloid cells is essential to initiate hepatitis. Once activated, these cells produce interleukin-27 that is required for liver toxicity. CD8 T cells infiltrate the liver in response to this myeloid activation and mediate tissue damage, triggering transaminase elevation. FoxP3+ regulatory T cells limit liver damage, and their removal dramatically exacerbates 4-1BB agonist-induced hepatitis. Coadministration of CTLA-4 blockade ameliorates transaminase elevation, whereas PD-1 blockade exacerbates it. Loss of the chemokine receptor CCR2 blocks 4-1BB agonist hepatitis without diminishing tumor-specific immunity against B16 melanoma.Conclusions: 4-1BB agonist antibodies trigger hepatitis via activation and expansion of interleukin-27-producing liver Kupffer cells and monocytes. Coadministration of CTLA-4 and/or CCR2 blockade may minimize hepatitis, but yield equal or greater antitumor immunity. Clin Cancer Res; 24(5); 1138-51. ©2018 AACR.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemical and Drug Induced Liver Injury/immunology , Interleukins/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Animals , Antineoplastic Agents, Immunological/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cell Line, Tumor/transplantation , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/pathology , Drug Evaluation, Preclinical , Humans , Interleukins/immunology , Liver/cytology , Liver/drug effects , Liver/immunology , Liver/pathology , Male , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/drug effects , Myeloid Cells/immunology , Myeloid Cells/metabolism , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/immunology , Signal Transduction/drug effects , Signal Transduction/immunology , Skin Neoplasms/drug therapy , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
3.
Proc Natl Acad Sci U S A ; 112(38): E5290-9, 2015 Sep 22.
Article in English | MEDLINE | ID: mdl-26351680

ABSTRACT

Antibody modulation of T-cell coinhibitory (e.g., CTLA-4) or costimulatory (e.g., 4-1BB) receptors promotes clinical responses to a variety of cancers. Therapeutic cancer vaccination, in contrast, has produced limited clinical benefit and no curative therapies. The E6 and E7 oncoproteins of human papilloma virus (HPV) drive the majority of genital cancers, and many oropharyngeal tumors. We discovered 15-19 amino acid peptides from HPV-16 E6/E7 for which induction of T-cell immunity correlates with disease-free survival in patients treated for high-grade cervical neoplasia. We report here that intranasal vaccination with these peptides and the adjuvant alpha-galactosylceramide elicits systemic and mucosal T-cell responses leading to reduced HPV(+) TC-1 tumor growth and prolonged survival in mice. We hypothesized that the inability of these T cells to fully reject established tumors resulted from suppression in the tumor microenvironment which could be ameliorated through checkpoint modulation. Combining this E6/E7 peptide vaccine with checkpoint blockade produced only modest benefit; however, coadministration with a 4-1BB agonist antibody promoted durable regression of established genital TC-1 tumors. Relative to other therapies tested, this combination of vaccine and α4-1BB promoted the highest CD8(+) versus regulatory FoxP3(+) T-cell ratios, elicited 2- to 5-fold higher infiltration by E7-specific CTL, and evoked higher densities of highly cytotoxic TcEO (T cytotoxic Eomesodermin) CD8 (>70-fold) and ThEO (T helper Eomesodermin) CD4 (>17-fold) T cells. These findings have immediate clinical relevance both in terms of the direct clinical utility of the vaccine studied and in illustrating the potential of 4-1BB antibody to convert therapeutic E6/E7 vaccines already in clinical trials into curative therapies.


Subject(s)
Antibodies/chemistry , Papillomavirus Vaccines/chemistry , Tumor Necrosis Factor Receptor Superfamily, Member 9/agonists , Animals , Cell Separation , Cytokines/metabolism , Female , Flow Cytometry , Immunotherapy/methods , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Oncogene Proteins, Viral/chemistry , Papillomaviridae , Papillomavirus E7 Proteins/chemistry , Papillomavirus Vaccines/immunology , Peptides/chemistry , Spleen/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology , Vaccines, Subunit/chemistry , Vaccines, Subunit/immunology , Vagina/pathology
4.
Cancer Immunol Immunother ; 64(7): 885-92, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25555570

ABSTRACT

The discovery that antibody blockade of the T cell co-inhibitory receptor cytotoxic T lymphocyte-associated protein 4 (CTLA-4) can restore tumor immunity against many murine transplantable tumors leading to complete rejection of established cancer forever changed the field of immunotherapy. In more robust murine models as well as human cancer, however, CTLA-4 blockade alone can slow tumor growth and extend patient survival, but is rarely curative. Subsequent studies have revealed a large family of T cell immune checkpoint receptors which tumors engage to shield themselves from host immunity. As with CTLA-4, blockade of one of these additional inhibitory receptors, programmed death 1, has led to remarkable therapeutic responses against tumors of multiple lineages. Checkpoint monotherapy has demonstrated that durable, immune-mediated cures of established metastatic cancers are possible, yet the percentage of patients experiencing these outcomes remains low due to both redundant mechanisms of immune suppression in the tumor and limiting toxicity associated with some therapies. Thus, extending the curative potential of immunotherapy to a larger percentage of patients with a broader spectrum of malignancies will likely require combinations of co-inhibitory blockade and co-stimulatory activation designed to peel back multiple layers of tumor immune suppression while at the same time minimizing immune-mediated toxicity. As over a dozen T cell immune checkpoints and an additional dozen more co-stimulatory receptors have now been described, the challenge before us is to identify the most advantageous combinations of these agents based on the knowledge of their underlying biology and preclinical studies in murine tumor models.


Subject(s)
Antigens, Neoplasm/immunology , CTLA-4 Antigen/antagonists & inhibitors , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , 4-1BB Ligand/metabolism , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, Neoplasm/genetics , CTLA-4 Antigen/genetics , CTLA-4 Antigen/immunology , Humans , Immunotherapy , Ipilimumab , Mice , Neoplasms/genetics , Neoplasms/therapy , T-Lymphocytes/immunology
5.
Spine (Phila Pa 1976) ; 39(15): E865-9, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-25171072

ABSTRACT

STUDY DESIGN: We used optogenetic techniques in spinal cord and dorsal root ganglion (DRG) neuron studies. OBJECTIVE: This study investigated changes in channelrhodopsin-2 (ChR2) expression in the spinal cord and DRG neurons using optogenetic techniques. The results show the possibility of using optogenetics to treat neuropathic pain. SUMMARY OF BACKGROUND DATA: Previous studies have shown that activated ChR2 induces an increase in DRG neuron action potential. METHODS: Western blot analysis was used to measure ChR2 protein levels in the spinal cord and DRG neurons or rats intrathecally injected with ChR2 lentivirus. Electrophysiology recording was used to detect differences in action potential levels in the spinal cord and calcium channel currents in the DRG neurons. RESULTS: Our studies showed that ChR2 expression increased the action potential in the spinal cord and increased calcium channel currents in DRG neurons. CONCLUSION: We successfully expressed the ChR2 protein in the spinal cord and DRG neurons. We also found that ChR2 increased the action potential in the spinal cord and activated the calcium channel in DRG neurons. These findings support the research possibilities of using optogenetic studies to improve treatment for neuropathic pain. LEVEL OF EVIDENCE: N/A.


Subject(s)
Action Potentials/physiology , Calcium Channels/physiology , Neurons/physiology , Spinal Cord/physiology , Animals , Blotting, Western , Cell Line, Tumor , Channelrhodopsins , Ganglia, Spinal/cytology , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Injections, Spinal , Lentivirus/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Microscopy, Confocal , Neurons/metabolism , Optogenetics/methods , Patch-Clamp Techniques , Rats, Sprague-Dawley , Spinal Cord/metabolism , Transfection/methods
6.
Cell Signal ; 25(9): 1754-61, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23707532

ABSTRACT

Breast tumor kinase (Brk), also known as protein kinase-6 (PTK6), is a nonreceptor protein-tyrosine kinase that has a close functional relationship with the human epidermal growth factor receptor 2 (HER2). High levels of Brk were found in HER2-positive tumor specimens from patients with invasive ductal breast cancer; however, the underlying mechanism of the co-overexpression of Brk and HER2 remains elusive. In the current study, we explored the mechanism of HER2 and Brk co-overexpression in breast cancer cells by investigating the effect of overexpression and knockdown of HER2 on the level of Brk in breast cancer cells. We found that Brk was more stable in HER2-elevated cells than in control vector-transfected cells and was less stable in HER2 siRNA-treated cells than in control siRNA-treated cells, suggesting that HER2 regulates Brk protein stability. Further studies indicated that degradation of Brk involved a calpain-1-mediated proteolytic pathway and indicated an inverse relationship between the level of HER2 expression and calpain-1 activity. We found that HER2 inhibited calpain-1 activity through upregulating calpastatin, an endogenous calpain inhibitor. Silencing of HER2 downregulated calpastatin, and the downregulation could be rescued by overexpression of constitutively active MEK. Together, these data offer novel mechanistic insights into the functional relationship between Brk and HER2.


Subject(s)
Breast Neoplasms/metabolism , Breast/metabolism , Calcium-Binding Proteins/metabolism , Calpain/metabolism , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Receptor, ErbB-2/metabolism , Breast/pathology , Breast Neoplasms/genetics , Calcium-Binding Proteins/genetics , Enzyme Stability , Female , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Proteins/chemistry , Neoplasm Proteins/genetics , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Proteolysis , RNA Interference , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Up-Regulation
7.
J Biol Chem ; 288(13): 8826-37, 2013 Mar 29.
Article in English | MEDLINE | ID: mdl-23382387

ABSTRACT

Induction of tumor cell apoptosis has been recognized as a valid anticancer strategy. However, therapeutic selectivity between tumor and normal cells has always been a challenge. Here, we report a novel anti-cancer compound methyl 3-(4-nitrophenyl) propiolate (NPP) preferentially induces apoptosis in tumor cells through P450-catalyzed reactive oxygen species (ROS) production. A compound sensitivity study on multiple cell lines shows that tumor cells with high basal ROS levels, low antioxidant capacities, and p53 mutations are especially sensitive to NPP. Knockdown of p53 sensitized non-transformed cells to NPP-induced cell death. Additionally, by comparing NPP with other ROS inducers, we show that the susceptibility of tumor cells to the ROS-induced cell death is influenced by the mode, amount, duration, and perhaps location of ROS production. Our studies not only discovered a unique anticancer drug candidate but also shed new light on the understanding of ROS generation and function and the potential application of a ROS-promoting strategy in cancer treatment.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Phenylpropionates/chemistry , Reactive Oxygen Species/metabolism , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antioxidants/metabolism , Apoptosis , Cell Line, Tumor , Cell Survival , Cytochromes c/metabolism , Gene Expression Regulation, Neoplastic , Genes, p53 , Hep G2 Cells , Humans , Janus Kinase 1/metabolism , Magnetic Resonance Spectroscopy/methods , Mass Spectrometry/methods , Mutation , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidation-Reduction , Phenylpropionates/pharmacology , Propionates/pharmacology , RNA Interference , Tumor Suppressor Protein p53/metabolism
8.
Cancer Biol Ther ; 14(3): 237-45, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23291984

ABSTRACT

Breast tumor kinase (Brk)/protein tyrosine kinase-6 (PTK-6) is a nonreceptor PTK commonly expressed at high levels in breast cancer. Brk interacts closely with members of the human epidermal growth factor receptor (HER) family in breast cancer but the functional role of this interaction remains to be determined. Here, we provide novel mechanistic insights into the role of Brk in regulating cell survival and epithelial-to-mesenchymal transition (EMT) in the context of HER2-positive breast cancer cells. Overexpression of HER2 in MCF7 breast cancer cells (MCF7HER2) led to a higher level of Brk protein and concomitantly reduced Src Y416-phosphorylation, and the cells became mesenchymal in morphology. An in vivo selection of MCF7HER2 cells in nude mice resulted in a subline, termed EMT1, that exhibited not only mesenchymal morphology but also enhanced migration potential. Compared with MCF7HER2 cells, EMT1 cells maintained a similar level of HER2 protein but had much higher level of activated HER2, and the increase in Brk protein and the decrease in Src Y416-phosphorylation were less in EMT1 cells. EMT1 cells exhibited increased sensitivity to both pharmacological inhibition of HER2 and knockdown of Brk than did MCF7HER2 cells. Knockdown of Brk induced apoptosis and partially reversed the EMT phenotype in EMT1 cells. Overexpression of a constitutively active STAT3, a known substrate of Brk, overcame Brk knockdown-induced effects in EMT1 cells. Together, our findings support a new paradigm wherein Brk plays both a complementary and a counterbalancing role in cooperating with HER2 and Src to regulate breast cancer cell survival and EMT.


Subject(s)
Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Neoplasm Proteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Receptor, ErbB-2/metabolism , Animals , Breast Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Survival/genetics , Disease Models, Animal , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Gene Knockdown Techniques , Humans , MCF-7 Cells , Mice , Neoplasm Proteins/genetics , Protein-Tyrosine Kinases/genetics , Receptor, ErbB-2/genetics , STAT3 Transcription Factor/metabolism , Transplantation, Heterologous
9.
PLoS One ; 5(5): e10499, 2010 May 05.
Article in English | MEDLINE | ID: mdl-20463925

ABSTRACT

Many mitosis inhibitors are powerful anticancer drugs. Tremendous efforts have been made to identify new anti-mitosis compounds for developing more effective and less toxic anti-cancer drugs. We have identified LJK-11, a synthetic analog of 5, 8-disubstituted quinazolines, as a novel mitotic blocker. LJK-11 inhibited growth and induced apoptosis of many different types of tumor cells. It prevented mitotic spindle formation and arrested cells at early phase of mitosis. Detailed in vitro analysis demonstrated that LJK-11 inhibited microtubule polymerization. In addition, LJK-11 had synergistic effect with another microtubule inhibitor colchicine on blocking mitosis, but not with vinblastine or nocodazole. Therefore, LJK-11 represents a novel anti-microtubule structure. Understanding the function and mechanism of LJK-11 will help us to better understand the action of anti-microtubule agents and to design better anti-cancer drugs.


Subject(s)
Apoptosis/drug effects , Microtubules/drug effects , Microtubules/metabolism , Mitosis/drug effects , Quinazolines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Colchicine/pharmacology , Drug Screening Assays, Antitumor , Drug Synergism , G2 Phase/drug effects , Humans , Phosphorylation/drug effects , Phosphotyrosine/metabolism , Quinazolines/chemistry , Spindle Apparatus/drug effects , Spindle Apparatus/metabolism , Tubulin/metabolism
10.
Zhonghua Zhong Liu Za Zhi ; 27(4): 204-8, 2005 Apr.
Article in Chinese | MEDLINE | ID: mdl-15949416

ABSTRACT

OBJECTIVE: To investigate effect of AP-1 and Ets binding site adjacent to matrix metalloproteinase-9 (MMP-9) promoter on activation of MMP-9 transcription of nasopharyngeal carcinoma cells transfected with EBV-encoded latent membrane protein 1 (LMP1), and to ascertain if cross-talk between c-Jun and Ets1 is involved in LMP1-regulating expression of MMP-9. METHODS: Site-directed mutagenesis technique was used to establish a series of mutants, including MMP-9-CAT-Ets(-540)mt, MMP-9-CAT-AP-1(-533)mt and MMP-9-CAT-AP-1(-533)/Ets(-540)mt. After the mutants were transfected into LMP1-expressing NPC HNE2 cells regulated by Tet-on system (pTet-on-LMP1 HNE2), CAT activity of these mutants were assayed with induction of LMP1. With blockade of c-Jun or Ets1 antisense oligonucleotides, the activity of MMP-9 induced by LMP1 was assayed with gelatin zymography. RESULTS: The CAT activity of MMP-9-Ets(-540)mt-CAT, MMP-9-AP-1(-533)mt-CAT, MMP-9-AP-1(-533)/Ets(-540) mt-CAT decreased significantly compared to MMP-9-CAT wt. After blockade with c-Jun or Ets1 antisense oligonucleotides, activity of MMP-9 induced by LMP1 decreased significantly, especially with combined blockade of c-Jun and Ets1. CONCLUSION: The results suggest that transcription factor AP-1 and Ets play an crucial role in activation of MMP-9 transcription induced by LMP1, and cross-talk between c-Jun/Ets1 is involved in expression of MMP-9 mediated by LMP1.


Subject(s)
Matrix Metalloproteinase 9/biosynthesis , Nasopharyngeal Neoplasms/metabolism , Proto-Oncogene Protein c-ets-1/genetics , Proto-Oncogene Proteins c-jun/genetics , Viral Matrix Proteins/genetics , Herpesvirus 4, Human/genetics , Humans , Matrix Metalloproteinase 9/genetics , Nasopharyngeal Neoplasms/virology , Transfection , Tumor Cells, Cultured
11.
Chin Med J (Engl) ; 116(7): 1022-8, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12890376

ABSTRACT

OBJECTIVES: To identify whether Epstein-Barr virus (EBV) encoded latent membrane protein 1 (LMP1) can induce tumor necrosis factor receptor-associated factor 1 (TRAF1) expression and promote its anti-apoptosis activity via the NF-kappaB signaling pathway, and assess that LMP1 suppresses apoptosis in nasopharyngeal carcinoma (NPC). METHODS: A stable transfected cell line HNE2-LMP1 was established by introducing LMP1 cDNA into HNE2 cells. Transactivation of TRAF1 was determined by luciferase reporter assay, while expression of TRAF1 mRNA was detected by RT-PCR and expression of TRAF1 protein and caspase 3 by Western blot analysis. Apoptosis activity was observed through fluorescence staining. RESULTS: LMP1 induced TRAF1 expression in NPC cells and caused a decrease in apoptosis. This induction could be blocked by antisense LMP1. Moreover, LMP1-mediated induction of a TRAF1 promoter-driven reporter gene was significantly impaired when the kappaB site kappaB1 or kappaB5 was disrupted, whereas mutation of kappaB3 had only a minor effect on LMP1 dependent up-regulation of the reporter gene. CONCLUSION: LMP1 induces TRAF1 expression and promotes its anti-apoptosis activity via the NF-kappaB signaling pathway, which may be one of the mechanisms that LMP1 uses to suppress apoptosis in NPC cells.


Subject(s)
Apoptosis/physiology , NF-kappa B/physiology , Nasopharyngeal Neoplasms/physiopathology , Protein Biosynthesis , Signal Transduction/physiology , Viral Matrix Proteins/physiology , Humans , TNF Receptor-Associated Factor 1 , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...