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










Publication year range
1.
Clin Cancer Res ; 23(13): 3371-3384, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28003307

ABSTRACT

Purpose: Metastatic breast cancers continue to elude current therapeutic strategies, including those utilizing PI3K inhibitors. Given the prominent role of PI3Kα,ß in tumor growth and PI3Kγ,δ in immune cell function, we sought to determine whether PI3K inhibition altered antitumor immunity.Experimental Design: The effect of PI3K inhibition on tumor growth, metastasis, and antitumor immune response was characterized in mouse models utilizing orthotopic implants of 4T1 or PyMT mammary tumors into syngeneic or PI3Kγ-null mice, and patient-derived breast cancer xenografts in humanized mice. Tumor-infiltrating leukocytes were characterized by IHC and FACS analysis in BKM120 (30 mg/kg, every day) or vehicle-treated mice and PI3Kγnull versus PI3KγWT mice. On the basis of the finding that PI3K inhibition resulted in a more inflammatory tumor leukocyte infiltrate, the therapeutic efficacy of BKM120 (30 mg/kg, every day) and anti-PD1 (100 µg, twice weekly) was evaluated in PyMT tumor-bearing mice.Results: Our findings show that PI3K activity facilitates tumor growth and surprisingly restrains tumor immune surveillance. These activities could be partially suppressed by BKM120 or by genetic deletion of PI3Kγ in the host. The antitumor effect of PI3Kγ loss in host, but not tumor, was partially reversed by CD8+ T-cell depletion. Treatment with therapeutic doses of both BKM120 and antibody to PD-1 resulted in consistent inhibition of tumor growth compared with either agent alone.Conclusions: PI3K inhibition slows tumor growth, enhances antitumor immunity, and heightens susceptibility to immune checkpoint inhibitors. We propose that combining PI3K inhibition with anti-PD1 may be a viable therapeutic approach for triple-negative breast cancer. Clin Cancer Res; 23(13); 3371-84. ©2016 AACR.


Subject(s)
Cell Proliferation/drug effects , Class Ib Phosphatidylinositol 3-Kinase/genetics , Mammary Neoplasms, Animal/drug therapy , Triple Negative Breast Neoplasms/drug therapy , Aminopyridines/administration & dosage , Animals , Cell Line, Tumor , Female , Humans , Immunity, Cellular/drug effects , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/immunology , Mammary Neoplasms, Animal/pathology , Mice , Morpholines/administration & dosage , Neoplasm Metastasis , Phosphoinositide-3 Kinase Inhibitors , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Protein Kinase Inhibitors/administration & dosage , Signal Transduction/drug effects , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
2.
Mol Cancer Ther ; 15(2): 313-22, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26719579

ABSTRACT

T cells recognize cancer cells via HLA/peptide complexes, and when disease overtakes these immune mechanisms, immunotherapy can exogenously target these same HLA/peptide surface markers. We previously identified an HLA-A2-presented peptide derived from macrophage migration inhibitory factor (MIF) and generated antibody RL21A against this HLA-A2/MIF complex. The objective of the current study was to assess the potential for targeting the HLA-A2/MIF complex in ovarian cancer. First, MIF peptide FLSELTQQL was eluted from the HLA-A2 of the human cancerous ovarian cell lines SKOV3, A2780, OV90, and FHIOSE118hi and detected by mass spectrometry. By flow cytometry, RL21A was shown to specifically stain these four cell lines in the context of HLA-A2. Next, partially matched HLA-A*02:01+ ovarian cancer (n = 27) and normal fallopian tube (n = 24) tissues were stained with RL21A by immunohistochemistry to assess differential HLA-A2/MIF complex expression. Ovarian tumor tissues revealed significantly increased RL21A staining compared with normal fallopian tube epithelium (P < 0.0001), with minimal staining of normal stroma and blood vessels (P < 0.0001 and P < 0.001 compared with tumor cells) suggesting a therapeutic window. We then demonstrated the anticancer activity of toxin-bound RL21A via the dose-dependent killing of ovarian cancer cells. In summary, MIF-derived peptide FLSELTQQL is HLA-A2-presented and recognized by RL21A on ovarian cancer cell lines and patient tumor tissues, and targeting of this HLA-A2/MIF complex with toxin-bound RL21A can induce ovarian cancer cell death. These results suggest that the HLA-A2/MIF complex should be further explored as a cell-surface target for ovarian cancer immunotherapy.


Subject(s)
Biomarkers, Tumor/metabolism , HLA-A2 Antigen/metabolism , Intramolecular Oxidoreductases/metabolism , Macrophage Migration-Inhibitory Factors/metabolism , Ovarian Neoplasms/metabolism , Peptides/metabolism , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Antibody Affinity , Antibody Specificity , Cell Line, Tumor , Female , HLA-A2 Antigen/immunology , Humans , Ovarian Neoplasms/pathology
3.
Clin Cancer Res ; 21(23): 5338-48, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26152738

ABSTRACT

PURPOSE: Preclinical studies show that inhibition of aurora kinases in melanoma tumors induces senescence and reduces tumor growth, but does not cause tumor regression. Additional preclinical models are needed to identify agents that will synergize with aurora kinase inhibitors to induce tumor regression. EXPERIMENTAL DESIGN: We combined treatment with an aurora kinase A inhibitor, MLN8237, with agents that activate death receptors (Apo2L/TRAIL or death receptor 5 agonists) and monitored the ability of this treatment to induce tumor apoptosis and melanoma tumor regression using human cell lines and patient-derived xenograft (PDX) mouse models. RESULTS: We found that this combined treatment led to apoptosis and markedly reduced cell viability. Mechanistic analysis showed that the induction of tumor cell senescence in response to the AURKA inhibitor resulted in a decreased display of Apo2L/TRAIL decoy receptors and increased display of one Apo2L/TRAIL receptor (death receptor 5), resulting in enhanced response to death receptor ligand/agonists. When death receptors were activated in senescent tumor cells, both intrinsic and extrinsic apoptotic pathways were induced independent of BRAF, NRAS, or p53 mutation status. Senescent tumor cells exhibited BID-mediated mitochondrial depolarization in response to Apo2L/TRAIL treatment. In addition, senescent tumor cells had a lower apoptotic threshold due to decreased XIAP and survivin expression. Melanoma tumor xenografts of one human cell line and one PDX displayed total blockage of tumor growth when treated with MLN8237 combined with DR5 agonist antibody. CONCLUSIONS: These findings provide a strong rationale for combining senescence-inducing therapeutics with death receptor agonists for improved cancer treatment.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aurora Kinases/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Receptors, Death Domain/agonists , Animals , Apoptosis/genetics , Azepines/pharmacology , Caspases/metabolism , Cell Line, Tumor , Cellular Senescence/drug effects , Disease Models, Animal , Drug Evaluation, Preclinical , Female , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma/metabolism , Melanoma/pathology , Mice , Pyrimidines/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, Tumor Necrosis Factor, Member 10c/metabolism , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Xenograft Model Antitumor Assays
4.
J Vis Exp ; (97)2015 Mar 24.
Article in English | MEDLINE | ID: mdl-25867039

ABSTRACT

According to the American Cancer Society, more than 200,000 women will be diagnosed with invasive breast cancer each year and approximately 40,000 will die from the disease. The human leukocyte antigen (HLA) class I samples peptides derived from proteasomal degradation of cellular proteins and presents these fragments on the cell surface for interrogation by circulating cytotoxic T lymphocytes (CTL). Generation of T-cell receptor mimic (TCRm) monoclonal antibodies (mAbs) which recognize breast cancer specific peptide/HLA-A*02:01 complexes such as those derived from macrophage migration inhibitory factor (MIF19-27) and NY-ESO-1157-165 enable detection and destruction of breast cancer cells in the absence of an effective anti-tumor CTL response. Intact class I HLA/peptide complexes are shed by breast cancer cells and represent potentially relevant cancer biomarkers. In this work, a breakthrough biomarker screening system for cancer diagnostics incorporating T-cell receptor mimic monoclonal antibodies combined with a novel, label-free biosensor utilizing guided-mode resonance (GMR) sensor technology is presented. Detection of shed MIF/HLA-A*02:01 complexes in MDA-MB-231 cell supernatants, spiked human serum, and patient plasma is demonstrated. The impact of this work could revolutionize personalized medicine through development of companion disease diagnostics for targeted immunotherapies.


Subject(s)
Biomarkers, Tumor/analysis , Biosensing Techniques/methods , Breast Neoplasms/chemistry , HLA-A2 Antigen/analysis , Antibodies, Monoclonal/chemistry , Antibodies, Monoclonal/immunology , Biomarkers, Tumor/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Cell Line, Tumor , Female , HLA-A2 Antigen/chemistry , HLA-A2 Antigen/immunology , Humans , Intramolecular Oxidoreductases/chemistry , Intramolecular Oxidoreductases/immunology , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , T-Lymphocytes, Cytotoxic/immunology
5.
Cancer Res ; 75(1): 181-93, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25398437

ABSTRACT

Therapeutics that induce cancer cell senescence can block cell proliferation and promote immune rejection. However, the risk of tumor relapse due to senescence escape may remain high due to the long lifespan of senescent cells that are not cleared. Here, we show how combining a senescence-inducing inhibitor of the mitotic kinase Aurora A (AURKA) with an MDM2 antagonist activates p53 in senescent tumors harboring wild-type 53. In the model studied, this effect is accompanied by proliferation arrest, mitochondrial depolarization, apoptosis, and immune clearance of cancer cells by antitumor leukocytes in a manner reliant upon Ccl5, Ccl1, and Cxcl9. The AURKA/MDM2 combination therapy shows adequate bioavailability and low toxicity to the host. Moreover, the prominent response of patient-derived melanoma tumors to coadministered MDM2 and AURKA inhibitors offers a sound rationale for clinical evaluation. Taken together, our work provides a preclinical proof of concept for a combination treatment that leverages both senescence and immune surveillance to therapeutic ends.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Aurora Kinase A/antagonists & inhibitors , Melanoma, Experimental/drug therapy , Melanoma/drug therapy , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Animals , Apoptosis/drug effects , Aurora Kinase A/metabolism , Azepines/administration & dosage , Azepines/pharmacology , Cell Proliferation/drug effects , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Melanoma/metabolism , Melanoma/pathology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Piperazines/administration & dosage , Piperazines/pharmacology , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , Pyrimidines/administration & dosage , Pyrimidines/pharmacology
6.
Cancer Res ; 74(24): 7274-84, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25336190

ABSTRACT

Myeloid cells are capable of promoting or eradicating tumor cells and the nodal functions that contribute to their different roles are still obscure. Here, we show that mice with myeloid-specific genetic loss of the NF-κB pathway regulatory kinase IKKß exhibit more rapid growth of cutaneous and lung melanoma tumors. In a BRAF(V600E/PTEN(-/-)) allograft model, IKKß loss in macrophages reduced recruitment of myeloid cells into the tumor, lowered expression of MHC class II molecules, and enhanced production of the chemokine CCL11, thereby negatively regulating dendritic-cell maturation. Elevated serum and tissue levels of CCL11 mediated suppression of dendritic-cell differentiation/maturation within the tumor microenvironment, skewing it toward a Th2 immune response and impairing CD8(+) T cell-mediated tumor cell lysis. Depleting macrophages or CD8(+) T cells in mice with wild-type IKKß myeloid cells enhanced tumor growth, where the myeloid cell response was used to mediate antitumor immunity against melanoma tumors (with less dependency on a CD8(+) T-cell response). In contrast, myeloid cells deficient in IKKß were compromised in tumor cell lysis, based on their reduced ability to phagocytize and digest tumor cells. Thus, mice with continuous IKKß signaling in myeloid-lineage cells (IKKß(CA)) exhibited enhanced antitumor immunity and reduced melanoma outgrowth. Collectively, our results illuminate new mechanisms through which NF-κB signaling in myeloid cells promotes innate tumor surveillance.


Subject(s)
I-kappa B Kinase/genetics , Immunity, Innate , Melanoma, Experimental/genetics , Tumor Microenvironment/immunology , Animals , CD8-Positive T-Lymphocytes/metabolism , Dendritic Cells , Humans , I-kappa B Kinase/metabolism , Macrophages/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism , NF-kappa B/metabolism , Proto-Oncogene Proteins B-raf/genetics , Signal Transduction/immunology , Tumor Microenvironment/genetics
7.
Eur J Immunol ; 44(7): 1936-46, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24723377

ABSTRACT

We used a newly generated T-cell receptor mimic monoclonal antibody (TCRm MAb) that recognizes a known nonself immunodominant peptide epitope from West Nile virus (WNV) NS4B protein to investigate epitope presentation after virus infection in C57BL/6 mice. Previous studies suggested that peptides of different length, either SSVWNATTAI (10-mer) or SSVWNATTA (9-mer) in complex with class I MHC antigen H-2D(b) , were immunodominant after WNV infection. Our data establish that both peptides are presented on the cell surface after WNV infection and that CD8(+) T cells can detect 10- and 9-mer length variants similarly. This result varies from the idea that a given T-cell receptor (TCR) prefers a single peptide length bound to its cognate class I MHC. In separate WNV infection studies with the TCRm MAb, we show that in vivo the 10-mer was presented on the surface of uninfected and infected CD8α(+) CD11c(+) dendritic cells, which suggests the use of direct and cross-presentation pathways. In contrast, CD11b(+) CD11c(-) cells bound the TCRm MAb only when they were infected. Our study demonstrates that TCR recognition of peptides is not limited to certain peptide lengths and that TCRm MAbs can be used to dissect the cell-type specific mechanisms of antigen presentation in vivo.


Subject(s)
Dendritic Cells/immunology , Immunodominant Epitopes , Receptors, Antigen, T-Cell/physiology , West Nile virus/immunology , Animals , CD11b Antigen/analysis , CD11c Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Mice , Mice, Inbred C57BL , Viral Nonstructural Proteins/immunology
8.
Traffic ; 15(4): 451-69, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24450359

ABSTRACT

The chemokine receptor CXCR2 is vital for inflammation, wound healing, angiogenesis, cancer progression and metastasis. Adaptor protein 2 (AP2), a clathrin binding heterotetrameric protein comprised of α, ß2, µ2 and σ2 subunits, facilitates clathrin-mediated endocytosis. Mutation of the LLKIL motif in the CXCR2 carboxyl-terminal domain (CTD) results in loss of AP2 binding to the receptor and loss of ligand-mediated receptor internalization and chemotaxis. AP2 knockdown also results in diminished ligand-mediated CXCR2 internalization, polarization and chemotaxis. Using knockdown/rescue approaches with AP2-µ2 mutants, the binding domains were characterized in reference to CXCR2 internalization and chemotaxis. When in an open conformation, µ2 Patch 1 and Patch 2 domains bind tightly to membrane PIP2 phospholipids. When AP2-µ2, is replaced with µ2 mutated in Patch 1 and/or Patch 2 domains, ligand-mediated receptor binding and internalization are not lost. However, chemotaxis requires AP2-µ2 Patch 1, but not Patch 2. AP2-σ2 has been demonstrated to bind dileucine motifs to facilitate internalization. Expression of AP2-σ2 V88D and V98S dominant negative mutants resulted in loss of CXCR2 mediated chemotaxis. Thus, AP2 binding to both membrane phosphatidylinositol phospholipids and dileucine motifs is crucial for directional migration or chemotaxis. Moreover, AP2-mediated receptor internalization can be dissociated from AP2-mediated chemotaxis.


Subject(s)
Adaptor Protein Complex 2/physiology , Chemotaxis/physiology , Receptors, Interleukin-8B/physiology , Adaptor Protein Complex 2/genetics , Base Sequence , DNA Primers , Endocytosis , HEK293 Cells , HL-60 Cells , Humans , Mutagenesis, Site-Directed
9.
FASEB J ; 27(7): 2788-98, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23568779

ABSTRACT

Bone marrow-derived human mesenchymal stem cells (hMSCs) either promote or inhibit cancer progression, depending on factors that heretofore have been undefined. Here we have utilized extreme hypoxia (0.5% O2) and concurrent treatment with metal carcinogen (nickel) to evaluate the passage-dependent response of hMSCs toward cancerous transformation. Effects of hypoxia and nickel treatment on hMSC proliferation, apoptosis, gene and protein expression, replicative senescence, reactive oxygen species (ROS), redox mechanisms, and in vivo tumor growth were analyzed. The behavior of late passage hMSCs in a carcinogenic hypoxia environment follows a profile similar to that of transformed cancer cells (i.e., increased expression of oncogenic proteins, decreased expression of tumor suppressor protein, increased proliferation, decreased apoptosis, and aberrant redox mechanisms), but this effect was not observed in earlier passage control cells. These events resulted in accumulated intracellular ROS in vitro and excessive proliferation in vivo. We suggest a mechanism by which carcinogenic hypoxia modulates the activity of three critical transcription factors (c-MYC, p53, and HIF1), resulting in accumulated ROS and causing hMSCs to undergo cancer-like behavioral changes. This is the first study to utilize carcinogenic hypoxia as an environmentally relevant experimental model for studying the age-dependent cancerous transformation of hMSCs.


Subject(s)
Cell Transformation, Neoplastic , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Nickel/pharmacology , Animals , Blotting, Western , Cell Differentiation/drug effects , Cell Hypoxia , Cell Proliferation/drug effects , Cells, Cultured , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Female , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Gene Expression/drug effects , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Knockout , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation, Heterologous , Tumor Burden/genetics , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
EMBO Mol Med ; 5(1): 149-66, 2013 01.
Article in English | MEDLINE | ID: mdl-23180582

ABSTRACT

Oncogene-induced senescence can provide a protective mechanism against tumour progression. However, production of cytokines and growth factors by senescent cells may contribute to tumour development. Thus, it is unclear whether induction of senescence represents a viable therapeutic approach. Here, using a mouse model with orthotopic implantation of metastatic melanoma tumours taken from 19 patients, we observed that targeting aurora kinases with MLN8054/MLN8237 impaired mitosis, induced senescence and markedly blocked proliferation in patient tumour implants. Importantly, when a subset of tumour-bearing mice were monitored for tumour progression after pausing MLN8054 treatment, 50% of the tumours did not progress over a 12-month period. Mechanistic analyses revealed that inhibition of aurora kinases induced polyploidy and the ATM/Chk2 DNA damage response, which mediated senescence and a NF-κB-related, senescence-associated secretory phenotype (SASP). Blockade of IKKß/NF-κB led to reversal of MLN8237-induced senescence and SASP. Results demonstrate that removal of senescent tumour cells by infiltrating myeloid cells is crucial for inhibition of tumour re-growth. Altogether, these data demonstrate that induction of senescence, coupled with immune surveillance, can limit melanoma growth.


Subject(s)
Azepines/pharmacology , Benzazepines/pharmacology , Melanoma, Experimental/drug therapy , NF-kappa B/antagonists & inhibitors , Protein Serine-Threonine Kinases/antagonists & inhibitors , Pyrimidines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins , Aurora Kinases , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Checkpoint Kinase 2 , DNA Damage , DNA-Binding Proteins/metabolism , Humans , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/secondary , Mice , Mice, Nude , Polyploidy , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism , Xenograft Model Antitumor Assays
11.
Cancer Res ; 72(18): 4682-95, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22836752

ABSTRACT

Although human angiosarcoma has been associated frequently with mutational inactivation of the tumor suppressor gene Ink4a/Arf, the underlying mechanisms have not been delineated. Here we report that malignant angiosarcoma is associated with high levels of RelA/NF-κB and IL-6 in contrast to normal vessels or benign hemagiomas. Studies of Ink4a/Arf deficient mice not only recapitulate genetic traits observed in human angiosarcoma, but also unveil a possible therapeutic link comprised of the NF-kB/IL-6/Stat3 signaling axis. In Ink4a/Arf(-/-) cells, NF-κB controlled Stat3 signaling by transcriptionally controlling the expression of IL-6, gp130, and Jak2. Further, IL-6 mediated Stat3 signaling through the sIL-6R. Inhibition of Ikkß solely in myeloid cells was insufficient to block angiosarcoma development; in contrast, systemic inhibition of Ikkß, IL-6, or Stat3 markedly inhibited angiosarcoma growth. Our findings offer clinical implications for targeting the NF-kB/IL-6/STAT3 pathway as a rational strategy to treat angiosarcoma.


Subject(s)
Hemangiosarcoma/metabolism , I-kappa B Kinase/metabolism , Interleukin-6/metabolism , NF-kappa B/metabolism , Signal Transduction/physiology , Animals , Blotting, Western , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Mice , Mice, Knockout , Real-Time Polymerase Chain Reaction , Tissue Array Analysis , Transcription Factor RelA/metabolism
12.
Clin Cancer Res ; 18(8): 2184-98, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22351689

ABSTRACT

PURPOSE: The purpose of this preclinical study was to determine the effectiveness of RAF265, a multikinase inhibitor, for treatment of human metastatic melanoma and to characterize traits associated with drug response. EXPERIMENTAL DESIGN: Advanced metastatic melanoma tumors from 34 patients were orthotopically implanted to nude mice. Tumors that grew in mice (17 of 34) were evaluated for response to RAF265 (40 mg/kg, every day) over 30 days. The relation between patient characteristics, gene mutation profile, global gene expression profile, and RAF265 effects on tumor growth, mitogen-activated protein/extracellular signal-regulated kinase (MEK)/extracellular signal-regulated kinase (ERK) phosphorylation, proliferation, and apoptosis markers was evaluated. RESULTS: Nine of the 17 tumors that successfully implanted (53%) were mutant BRAF (BRAF(V600E/K)), whereas eight of 17 (47%) tumors were BRAF wild type (BRAF(WT)). Tumor implants from 7 of 17 patients (41%) responded to RAF265 treatment with more than 50% reduction in tumor growth. Five of the 7 (71%) responders were BRAF(WT), of which 1 carried c-KIT(L576P) and another N-RAS(Q61R) mutation, while only 2 (29%) of the responding tumors were BRAF(V600E/K). Gene expression microarray data from nonimplanted tumors revealed that responders exhibited enriched expression of genes involved in cell growth, proliferation, development, cell signaling, gene expression, and cancer pathways. Although response to RAF265 did not correlate with pERK1/2 reduction, RAF265 responders did exhibit reduced pMEK1, reduced proliferation based upon reduced Ki-67, cyclin D1 and polo-like kinase1 levels, and induction of the apoptosis mediator BCL2-like 11. CONCLUSIONS: Orthotopic implants of patient tumors in mice may predict prognosis and treatment response for melanoma patients. A subpopulation of human melanoma tumors responds to RAF265 and can be characterized by gene mutation and gene expression profiles.


Subject(s)
Imidazoles/pharmacology , Imidazoles/therapeutic use , Melanoma/drug therapy , Pyridines/pharmacology , Pyridines/therapeutic use , Animals , Apoptosis/drug effects , Base Sequence , Cell Cycle Proteins/biosynthesis , Cell Proliferation/drug effects , Cyclin D1/biosynthesis , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Gene Expression Profiling , Humans , Ki-67 Antigen/biosynthesis , MAP Kinase Signaling System/drug effects , Male , Melanoma/metabolism , Melanoma/pathology , Melanoma/secondary , Mice , Mice, Nude , Mutation , Oligonucleotide Array Sequence Analysis , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/biosynthesis , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Sequence Analysis, DNA , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , bcl-X Protein/biosynthesis , Polo-Like Kinase 1
13.
Breast Cancer Res ; 14(1): 103, 2012 Jan 26.
Article in English | MEDLINE | ID: mdl-22293321

ABSTRACT

The contribution of CXCR7 to the tumor microenvironment has introduced a new level of complexity to CXCL12 signaling in breast cancer. In the previous issue of Breast Cancer Research, Hernandez and colleagues delineate the roles of CXCR4 and CXCR7 in tumor invasion and metastasis. The authors demonstrate that co-expression of CXCR7 and CXCR4 results in inhibition of CXCL12-mediated invasion, reduced intravasation of tumor cells into the vasculature, and fewer lung metastases compared with parental tumors. The results of this study suggest the combination of small molecule inhibitors of CXCR4 and CXCR7 could dramatically reduce invasion, intravasation, and metastasis and could be highly beneficial for the treatment of invasive breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, CXCR4/metabolism , Receptors, CXCR/metabolism , Animals , Female , Humans
14.
Int Rev Immunol ; 30(5-6): 328-40, 2011.
Article in English | MEDLINE | ID: mdl-22053972

ABSTRACT

The human leukocyte antigen (HLA; also called major histocompatibility, or MHC) class I system presents peptides that distinguish healthy from diseased cells. Therefore, the discovery of peptide/MHC class I markers can provide highly specific targets for immunotherapy. Over the course of almost two decades, various strategies have been used, with mixed success, to produce antibodies that have recognition specificity for unique peptide/MHC class I complexes that mark infected and cancerous cells. Using these antibody reagents, novel peptide/MHC class I targets have been directly validated on diseased cells and new insight has been gained into the mechanisms of antigen presentation. More recently, these antibodies have shown promise for clinical applications such as therapeutic targeting of cancerous and infected cells and diagnosis and imaging of diseased cells. In this review, the authors comprehensively describe the methods used to identify disease-specific peptide/MHC class I epitopes and generate antibodies to these markers. Finally, they offer several examples that illustrate the promise of using these antibodies as anti-cancer agents.


Subject(s)
Antibodies, Monoclonal/immunology , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/immunology , Peptides/immunology , Receptors, Antigen, T-Cell/immunology , Animals , Cell Line , Cell Line, Tumor , Histocompatibility Antigens Class I/metabolism , Humans , Immunotherapy/methods , Peptides/metabolism , Receptors, Antigen, T-Cell/metabolism
15.
BMC Immunol ; 12: 39, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21762519

ABSTRACT

BACKGROUND: MHC class I proteins are partly responsible for shaping the magnitude and focus of the adaptive cellular immune response. In humans, conventional wisdom suggests that the HLA-A, -B, and -C alleles are equally expressed on the majority of cell types. While we currently have a thorough understanding of how total MHC class I expression varies in different tissues, it has been difficult to examine expression of single MHC class I alleles due to the homogeneity of MHC class I sequences. It is unclear how cDNA species are expressed in distinct cell subsets in humans and particularly in macaques which transcribe upwards of 20 distinct MHC class I alleles at variable levels. RESULTS: We examined MHC gene expression in human and macaque leukocyte subsets. In humans, while we detected overall differences in locus transcription, we found that transcription of MHC class I genes was consistent across the leukocyte subsets we studied with only small differences detected. In contrast, transcription of certain MHC cDNA species in macaques varied dramatically by up to 45% between different subsets. Although the Mafa-B134:02 RNA is virtually undetectable in CD4+ T cells, it represents over 45% of class I transcripts in CD14+ monocytes. We observed parallel MHC transcription differences in rhesus macaques. Finally, we analyzed expression of select MHC proteins at the cell surface using fluorescent peptides. This technique confirmed results from the transcriptional analysis and demonstrated that other MHC proteins, known to restrict SIV-specific responses, are also differentially expressed among distinct leukocyte subsets. CONCLUSIONS: We assessed MHC class I transcription and expression in human and macaque leukocyte subsets. Until now, it has been difficult to examine MHC class I allele expression due to the similarity of MHC class I sequences. Using two novel techniques we showed that expression varies among distinct leukocyte subsets of macaques but does not vary dramatically in the human cell subsets we examined. These findings suggest pathogen tropism may have a profound impact on the shape and focus of the MHC class I restricted CD8+ T cell response in macaques.


Subject(s)
Histocompatibility Antigens Class I/metabolism , Leukocytes/immunology , Alleles , Animals , CD8-Positive T-Lymphocytes/immunology , Gene Expression Regulation , Haplotypes , Histocompatibility Antigens Class I/genetics , Humans , Leukocytes/metabolism , Macaca , Transcription, Genetic
16.
J Immunol ; 186(11): 6607-16, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21515791

ABSTRACT

This report describes a novel HLA/peptide complex with potential prognostic and therapeutic roles for invasive breast cancer. Macrophage migration inhibitory factor (MIF) mediates inflammation and immunity, and MIF overexpression is observed in breast cancer. We hypothesized that the HLA class I of cancerous breast epithelial cells would present MIF-derived peptides. Consistent with this hypothesis, the peptide FLSELTQQL (MIF(19-27)) was eluted from the HLA-A*0201 (HLA-A2) of breast cancer cell lines. We posited that if this MIF(19-27)/HLA-A2 complex was exclusively found in invasive breast cancer, it could be a useful prognostic indicator. To assess the presentation of MIF peptides by the HLA of various cells and tissues, mice were immunized with the MIF(19-27)/HLA-A2 complex. The resulting mAb (RL21A) stained invasive ductal carcinoma (IDC) but not ductal carcinoma in situ, fibroadenoma, or normal breast tissues. RL21A did not stain WBCs (total WBCs) or normal tissues from deceased HLA-A2 donors, substantiating the tumor-specific nature of this MIF/HLA complex. As this MIF/HLA complex appeared specific to the surface of IDC, RL21A was tested as an immunotherapeutic for breast cancer in vitro and in vivo. In vitro, RL21A killed the MDA-MB-231 cell line via complement and induction of apoptosis. In an in vivo orthotopic mouse model, administration of RL21A reduced MDA-MB-231 and BT-20 tumor burden by 5-fold and by >2-fold, respectively. In summary, HLA-presented MIF peptides show promise as prognostic cell surface indicators for IDC and as targets for immunotherapeutic intervention.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/immunology , HLA-A Antigens/immunology , Macrophage Migration-Inhibitory Factors/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/therapeutic use , Antibody Affinity/immunology , Antibody Specificity/immunology , Apoptosis/drug effects , Apoptosis/immunology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/pathology , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Dose-Response Relationship, Drug , Female , HLA-A Antigens/metabolism , HLA-A2 Antigen , Humans , Kinetics , Macrophage Migration-Inhibitory Factors/chemistry , Macrophage Migration-Inhibitory Factors/metabolism , Mice , Mice, Nude , Peptides/immunology , Peptides/metabolism , Prognosis , Protein Binding/immunology , Xenograft Model Antitumor Assays
17.
Immunogenetics ; 63(5): 275-90, 2011 May.
Article in English | MEDLINE | ID: mdl-21274527

ABSTRACT

The Simian immunodeficiency virus (SIV)-infected Indian rhesus macaque (Macaca mulatta) is the most established model of HIV infection and AIDS-related research, despite the potential that macaques of Chinese origin is a more relevant model. Ongoing efforts to further characterize the Chinese rhesus macaques' major histocompatibility complex (MHC) for composition and function should facilitate greater utilization of the species. Previous studies have demonstrated that Chinese-origin M. mulatta (Mamu) class I alleles are more polymorphic than their Indian counterparts, perhaps inferring a model more representative of human MHC, human leukocyte antigen (HLA). Furthermore, the Chinese rhesus macaque class I allele Mamu-A1*02201, the most frequent allele thus far identified, has recently been characterized and shown to be an HLA-B7 supertype analog, the most frequent supertype in human populations. In this study, we have characterized two additional alleles expressed with high frequency in Chinese rhesus macaques, Mamu-A1*02601 and Mamu-B*08301. Upon the development of MHC-peptide-binding assays and definition of their associated motifs, we reveal that these Mamu alleles share peptide-binding characteristics with the HLA-A2 and HLA-A3 supertypes, respectively, the next most frequent human supertypes after HLA-B7. These data suggest that Chinese rhesus macaques may indeed be a more representative model of HLA gene diversity and function as compared to the species of Indian origin and therefore a better model for investigating human immune responses.


Subject(s)
Disease Models, Animal , Histocompatibility Antigens Class I/immunology , Macaca mulatta/immunology , Amino Acid Sequence , Animals , Base Sequence , Cells, Cultured , Gene Frequency , HLA-A2 Antigen/genetics , HLA-A2 Antigen/immunology , HLA-A3 Antigen/genetics , HLA-A3 Antigen/immunology , Histocompatibility Antigens Class I/genetics , Macaca mulatta/genetics , Molecular Sequence Data , Simian Immunodeficiency Virus/genetics , Simian Immunodeficiency Virus/immunology
18.
Cancer Immunol Immunother ; 59(4): 563-73, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19779714

ABSTRACT

The identification and validation of new cancer-specific T cell epitopes continues to be a major area of research interest. Nevertheless, challenges remain to develop strategies that can easily discover and validate epitopes expressed in primary cancer cells. Regarded as targets for T cells, peptides presented in the context of the major histocompatibility complex (MHC) are recognized by monoclonal antibodies (mAbs). These mAbs are of special importance as they lend themselves to the detection of epitopes expressed in primary tumor cells. Here, we use an approach that has been successfully utilized in two different infectious disease applications (WNV and influenza). A direct peptide-epitope discovery strategy involving mass spectrometric analysis led to the identification of peptide YLLPAIVHI in the context of MHC A*02 allele (YLL/A2) from human breast carcinoma cell lines. We then generated and characterized an anti-YLL/A2 mAb designated as RL6A TCRm. Subsequently, the TCRm mAb was used to directly validate YLL/A2 epitope expression in human breast cancer tissue, but not in normal control breast tissue. Moreover, mice implanted with human breast cancer cells grew tumors, yet when treated with RL6A TCRm showed a marked reduction in tumor size. These data demonstrate for the first time a coordinated direct discovery and validation strategy that identified a peptide/MHC complex on primary tumor cells for antibody targeting and provide a novel approach to cancer immunotherapy.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/therapy , Epitopes, T-Lymphocyte/immunology , Peptide Fragments/therapeutic use , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , Antibodies, Monoclonal/immunology , Antigens, Neoplasm/immunology , Blotting, Western , Breast/metabolism , Breast/pathology , Breast Neoplasms/immunology , Cancer Vaccines/therapeutic use , DEAD-box RNA Helicases/immunology , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Humans , Immunoenzyme Techniques , Mice , Mice, Nude , Molecular Mimicry , Peptide Fragments/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Surface Plasmon Resonance
19.
J Immunol ; 182(12): 7763-75, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19494300

ABSTRACT

HLA-B27- and -B57-positive HIV-infected humans have long been associated with control of HIV replication, implying that CD8(+) T cell responses contribute to control of viral replication. In a similar fashion, 50% of Mamu-B*08-positive Indian rhesus macaques control SIVmac239 replication and become elite controllers with chronic-phase viremia <1000 viral RNA copies/ml. Interestingly, Mamu-B*08-restricted SIV-derived epitopes appeared to match the peptide binding profile for HLA-B*2705 in humans. We therefore defined a detailed peptide-binding motif for Mamu-B*08 and investigated binding similarities between the macaque and human MHC class I molecules. Analysis of a panel of approximately 900 peptides revealed that despite substantial sequence differences between Mamu-B*08 and HLA-B*2705, the peptide-binding repertoires of these two MHC class I molecules share a remarkable degree of overlap. Detailed knowledge of the Mamu-B*08 peptide-binding motif enabled us to identify six additional novel Mamu-B*08-restricted SIV-specific CD8(+) T cell immune responses directed against epitopes in Gag, Vpr, and Env. All 13 Mamu-B*08-restricted epitopes contain an R at the position 2 primary anchor and 10 also possess either R or K at the N terminus. Such dibasic peptides are less prone to cellular degradation. This work highlights the relevance of the Mamu-B*08-positive SIV-infected Indian rhesus macaque as a model to examine elite control of immunodeficiency virus replication. The remarkable similarity of the peptide-binding motifs and repertoires for Mamu-B*08 and HLA-B*2705 suggests that the nature of the peptide bound by the MHC class I molecule may play an important role in control of immunodeficiency virus replication.


Subject(s)
Histocompatibility Antigens Class I/immunology , Peptides/immunology , Peptides/metabolism , Simian Immunodeficiency Virus/immunology , Virus Replication , Amino Acid Sequence , Animals , DNA-Directed DNA Polymerase , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , Histocompatibility Antigens Class I/chemistry , Humans , Ligands , Macaca mulatta , Molecular Sequence Data , Peptide Library , Protein Binding , Sequence Homology, Amino Acid
20.
J Proteome Res ; 7(4): 1445-57, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18345606

ABSTRACT

Cellular immune mechanisms detect and destroy cancerous and infected cells via the human leukocyte antigen (HLA) class I molecules that present peptides of intracellular origin on the surface of all nucleated cells. The identification of novel, tumor-specific epitopes is a critical step in the development of immunotherapeutics for breast cancer. To directly identify peptide epitopes unique to cancerous cells, secreted human class I HLA molecules (sHLA) were constructed by deletion of the transmembrane and cytoplasmic domain of HLA A*0201. The resulting sHLA-A*0201 was transferred and expressed in breast cancer cell lines MCF-7, MDA-MB-231, and BT-20 as well as in the immortal, nontumorigenic cell line MCF10A. Stable transfectants were seeded into bioreactors for production of > 25 mg of sHLA-A*0201. Peptides eluted from affinity purified sHLA were analyzed by mass spectroscopy. Comparative analysis of HLA-A*0201 peptides revealed 5 previously uncharacterized epitopes uniquely presented on breast cancer cells. These peptides were derived from intracellular proteins with either well-defined or putative roles in breast cancer development and progression: Cyclin Dependent Kinase 2 (Cdk2), Ornithine Decarboxylase (ODC1), Kinetochore Associated 2 (KNTC2 or HEC1), Macrophage Migration Inhibitory Factor (MIF), and Exosome Component 6 (EXOSC6). Cellular recognition of the MIF, KNTC2, EXOSC6, and Cdk2 peptides by circulating CD8+ cells was demonstrated by tetramer staining and IFN-gamma ELISPOT. The identification and characterization of peptides unique to the class I of breast cancer cells provide putative targets for the development of immune diagnostic tools and therapeutics.


Subject(s)
Breast Neoplasms/immunology , Epitopes, T-Lymphocyte/analysis , HLA-A Antigens/immunology , Amino Acid Sequence , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Chromatography, High Pressure Liquid , Cyclin-Dependent Kinase 2/immunology , Cyclin-Dependent Kinase 2/metabolism , Cytoskeletal Proteins , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/isolation & purification , Exoribonucleases/immunology , Exoribonucleases/metabolism , Female , HLA-A Antigens/chemistry , HLA-A2 Antigen , Humans , Interferon-gamma/metabolism , Intramolecular Oxidoreductases/immunology , Intramolecular Oxidoreductases/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Macrophage Migration-Inhibitory Factors/immunology , Macrophage Migration-Inhibitory Factors/metabolism , Mass Spectrometry , Nuclear Proteins/immunology , Nuclear Proteins/metabolism , Ornithine Decarboxylase/immunology , Ornithine Decarboxylase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL