19 results on '"Knutson KL"'
Search Results
2. Abstract P2-11-02: Robust generation of T cell immunity to HER2 in HER2+ breast cancer patients with a degenerate subdominant HLA-DR epitope vaccine
- Author
-
Knutson, KL, primary, Kalli, KR, additional, Block, MS, additional, Hobday, TJ, additional, Padley, DJ, additional, Erskine, CL, additional, Dockter, T, additional, Suman, VJ, additional, Wilson, G, additional, and Degnim, AC, additional
- Published
- 2016
- Full Text
- View/download PDF
3. Abstract P5-03-07: Targeting Hedgehog pathway to reverse chemoresistance in breast cancer stem cells
- Author
-
Mañu, A, primary, Fresquet, V, additional, Mena, M, additional, Sánchez, S, additional, Espinós, J, additional, Fernandez, Hidalgo OA, additional, Fernández-Zapico, M, additional, Knutson, KL, additional, Martínez-Climent, JA, additional, and Santisteban, M, additional
- Published
- 2012
- Full Text
- View/download PDF
4. IL10 Release upon PD-1 Blockade Sustains Immunosuppression in Ovarian Cancer.
- Author
-
Lamichhane P, Karyampudi L, Shreeder B, Krempski J, Bahr D, Daum J, Kalli KR, Goode EL, Block MS, Cannon MJ, and Knutson KL
- Subjects
- Animals, B-Lymphocytes immunology, Cell Line, Tumor, Dendritic Cells immunology, Drug Resistance, Neoplasm, Female, Humans, Mice, Mice, Inbred C57BL, Ovarian Neoplasms drug therapy, RNA Interference, RNA, Small Interfering genetics, STAT3 Transcription Factor antagonists & inhibitors, STAT3 Transcription Factor genetics, T-Lymphocytes immunology, Tumor Microenvironment immunology, Interleukin-10 metabolism, Interleukin-10 pharmacology, Ovarian Neoplasms immunology, Programmed Cell Death 1 Receptor antagonists & inhibitors, Programmed Cell Death 1 Receptor biosynthesis
- Abstract
Ligation of programmed cell death-1 (PD-1) in the tumor microenvironment is known to inhibit effective adaptive antitumor immunity. Blockade of PD-1 in humans has resulted in impressive, durable regression responses in select tumor types. However, durable responses have been elusive in ovarian cancer patients. PD-1 was recently shown to be expressed on and thereby impair the functions of tumor-infiltrating murine and human myeloid dendritic cells (TIDC) in ovarian cancer. In the present work, we characterize the regulation of PD-1 expression and the effects of PD-1 blockade on TIDC. Treatment of TIDC and bone marrow-derived dendritic cells (DC) with IL10 led to increased PD-1 expression. Both groups of DCs also responded to PD-1 blockade by increasing production of IL10. Similarly, treatment of ovarian tumor-bearing mice with PD-1 blocking antibody resulted in an increase in IL10 levels in both serum and ascites. While PD-1 blockade or IL10 neutralization as monotherapies were inefficient, combination of these two led to improved survival and delayed tumor growth; this was accompanied by augmented antitumor T- and B-cell responses and decreased infiltration of immunosuppressive MDSC. Taken together, our findings implicate compensatory release of IL10 as one of the adaptive resistance mechanisms that undermine the efficacy of anti-PD-1 (or anti-PD-L1) monotherapies and prompt further studies aimed at identifying such resistance mechanisms. Cancer Res; 77(23); 6667-78. ©2017 AACR ., (©2017 American Association for Cancer Research.)
- Published
- 2017
- Full Text
- View/download PDF
5. Improved Survival of HER2+ Breast Cancer Patients Treated with Trastuzumab and Chemotherapy Is Associated with Host Antibody Immunity against the HER2 Intracellular Domain.
- Author
-
Knutson KL, Clynes R, Shreeder B, Yeramian P, Kemp KP, Ballman K, Tenner KS, Erskine CL, Norton N, Northfelt D, Tan W, Calfa C, Pegram M, Mittendorf EA, and Perez EA
- Subjects
- Breast Neoplasms drug therapy, Breast Neoplasms metabolism, Capecitabine administration & dosage, Carboplatin administration & dosage, Case-Control Studies, Female, Follow-Up Studies, Humans, Immunization, Neoplasm Staging, Paclitaxel administration & dosage, Prognosis, Prospective Studies, Receptor, ErbB-2 metabolism, Survival Rate, Trastuzumab administration & dosage, Vinblastine administration & dosage, Vinblastine analogs & derivatives, Vinorelbine, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Biomarkers, Tumor metabolism, Breast Neoplasms immunology, Breast Neoplasms mortality, Receptor, ErbB-2 immunology
- Abstract
The addition of trastuzumab to chemotherapy extends survival among patients with HER2(+) breast cancer. Prior work showed that trastuzumab and chemotherapy augments HER2 extracellular domain (ECD)-specific antibodies. The current study investigated whether combination therapy induced immune responses beyond HER2-ECD and, importantly, whether those immune responses were associated with survival. Pretreatment and posttreatment sera were obtained from 48 women with metastatic HER2(+) breast cancer on NCCTG (now Alliance for Clinical Trials in Oncology) studies, N0337 and N983252. IgG to HER2 intracellular domain (ICD), HER2-ECD, p53, IGFBP2, CEA, and tetanus toxoid were examined. Sera from 25 age-matched controls and 26 surgically resected HER2(+) patients were also examined. Prior to therapy, some patients with metastatic disease had elevated antibodies to IGFBP2, p53, HER2-ICD, HER2-ECD, and CEA, but not to tetanus toxin, relative to controls and surgically resected patients. Treatment augmented antibody responses to HER2-ICD in 69% of metastatic patients, which was highly associated with improved progression-free survival (PFS; HR = 0.5, P = 0.0042) and overall survival (OS; HR = 0.7, P = 0.038). Augmented antibody responses to HER2-ICD also correlated (P = 0.03) with increased antibody responses to CEA, IGFBP2, and p53, indicating that treatment induces epitope spreading. Paradoxically, patients who already had high preexisting immunity to HER2-ICD did not respond to therapy with increased antibodies to HER2-ICD and demonstrated poorer PFS (HR = 1.6, P < 0.0001) and OS (HR = 1.4, P = 0.0006). Overall, the findings further demonstrate the importance of the adaptive immune system in the efficacy of trastuzumab-containing regimens. Cancer Res; 76(13); 3702-10. ©2016 AACR., (©2016 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
6. PD-1 Blunts the Function of Ovarian Tumor-Infiltrating Dendritic Cells by Inactivating NF-κB.
- Author
-
Karyampudi L, Lamichhane P, Krempski J, Kalli KR, Behrens MD, Vargas DM, Hartmann LC, Janco JM, Dong H, Hedin KE, Dietz AB, Goode EL, and Knutson KL
- Subjects
- Animals, Female, Humans, Mice, Mice, Inbred C57BL, Ovarian Neoplasms pathology, Signal Transduction, Dendritic Cells immunology, NF-kappa B metabolism, Ovarian Neoplasms immunology, Programmed Cell Death 1 Receptor immunology
- Abstract
The PD-1:PD-L1 immune signaling axis mediates suppression of T-cell-dependent tumor immunity. PD-1 expression was recently found to be upregulated on tumor-infiltrating murine (CD11c(+)CD11b(+)CD8(-)CD209a(+)) and human (CD1c(+)CD19(-)) myeloid dendritic cells (TIDC), an innate immune cell type also implicated in immune escape. However, there is little knowledge concerning how PD-1 regulates innate immune cells. In this study, we examined the role of PD-1 in TIDCs derived from mice bearing ovarian tumors. Similar to lymphocytes, TIDC expression of PD-1 was associated with expression of the adapter protein SHP-2, which signals to NF-κB; however, in contrast to its role in lymphocytes, we found that expression of PD-1 in TIDC tonically paralyzed NF-κB activation. Further mechanistic investigations showed that PD-1 blocked NF-κB-dependent cytokine release in a SHP-2-dependent manner. Conversely, inhibition of NF-κB-mediated antigen presentation by PD-1 occurred independently of SHP-2. Collectively, our findings revealed that PD-1 acts in a distinct manner in innate immune cells compared with adaptive immune cells, prompting further investigations of the signaling pathways controlled by this central mediator of immune escape in cancer., (©2015 American Association for Cancer Research.)
- Published
- 2016
- Full Text
- View/download PDF
7. Oncolytic measles virus expressing the sodium iodide symporter to treat drug-resistant ovarian cancer.
- Author
-
Galanis E, Atherton PJ, Maurer MJ, Knutson KL, Dowdy SC, Cliby WA, Haluska P Jr, Long HJ, Oberg A, Aderca I, Block MS, Bakkum-Gamez J, Federspiel MJ, Russell SJ, Kalli KR, Keeney G, Peng KW, and Hartmann LC
- Subjects
- Animals, Cohort Studies, Drug Resistance, Neoplasm, Female, Humans, Measles virus genetics, Measles virus metabolism, Mice, Middle Aged, Oncolytic Viruses genetics, Oncolytic Viruses metabolism, Ovarian Neoplasms pathology, Ovarian Neoplasms virology, Symporters genetics, Transgenes, Xenograft Model Antitumor Assays, Measles virus physiology, Oncolytic Virotherapy methods, Oncolytic Viruses physiology, Ovarian Neoplasms therapy, Symporters biosynthesis
- Abstract
Edmonston vaccine strains of measles virus (MV) have significant antitumor activity in mouse xenograft models of ovarian cancer. MV engineered to express the sodium iodide symporter gene (MV-NIS) facilitates localization of viral gene expression and offers a tool for tumor radiovirotherapy. Here, we report results from a clinical evaluation of MV-NIS in patients with taxol- and platinum-resistant ovarian cancer. MV-NIS was given intraperitoneally every 4 weeks for up to 6 cycles. Treatment was well tolerated and associated with promising median overall survival in these patients with heavily pretreated ovarian cancer; no dose-limiting toxicity was observed in 16 patients treated at high-dose levels (10(8)-10(9) TCID50), and their median overall survival of 26.5 months compared favorably with other contemporary series. MV receptor CD46 and nectin-4 expression was confirmed by immunohistochemistry in patient tumors. Sodium iodide symporter expression in patient tumors after treatment was confirmed in three patients by (123)I uptake on SPECT/CTs and was associated with long progression-free survival. Immune monitoring posttreatment showed an increase in effector T cells recognizing the tumor antigens IGFBP2 and FRα, indicating that MV-NIS treatment triggered cellular immunity against the patients' tumor and suggesting that an immune mechanism mediating the observed antitumor effect. Our findings support further clinical evaluation of MV-NIS as an effective immunovirotherapy., (©2014 American Association for Cancer Research.)
- Published
- 2015
- Full Text
- View/download PDF
8. Accumulation of memory precursor CD8 T cells in regressing tumors following combination therapy with vaccine and anti-PD-1 antibody.
- Author
-
Karyampudi L, Lamichhane P, Scheid AD, Kalli KR, Shreeder B, Krempski JW, Behrens MD, and Knutson KL
- Subjects
- Animals, Antigens, Neoplasm immunology, B7-H1 Antigen immunology, CD8-Positive T-Lymphocytes immunology, Cell Line, Tumor, Dendritic Cells drug effects, Dendritic Cells immunology, Disease-Free Survival, Female, Immunologic Memory drug effects, Mastocytoma immunology, Mastocytoma therapy, Mice, Mice, Inbred BALB C, Receptors, Interleukin-7 immunology, Tumor Microenvironment drug effects, Tumor Microenvironment immunology, Antibodies immunology, Antibodies pharmacology, CD8-Positive T-Lymphocytes drug effects, Cancer Vaccines immunology, Cancer Vaccines pharmacology, Immunologic Memory immunology, Programmed Cell Death 1 Receptor immunology
- Abstract
Immunosuppression in the tumor microenvironment blunts vaccine-induced immune effectors. PD-1/B7-H1 is an important inhibitory axis in the tumor microenvironment. Our goal in this study was to determine the effect of blocking this inhibitory axis during and following vaccination against breast cancer. We observed that using anti-PD-1 antibody and a multipeptide vaccine (consisting of immunogenic peptides derived from breast cancer antigens, neu, legumain, and β-catenin) as a combination therapy regimen for the treatment of breast cancer-bearing mice prolonged the vaccine-induced progression-free survival period. This prolonged survival was associated with increase in number of Tc1 and Tc2 CD8 T cells with memory precursor phenotype, CD27+IL-7RhiT-betlo, and decrease in number of PD-1+ dendritic cells (DC) in regressing tumors and enhanced antigen reactivity of tumor-infiltrating CD8 T cells. It was also observed that blockade of PD-1 on tumor DCs enhanced IL-7R expression on CD8 T cells. Taken together, our results suggest that PD-1 blockade enhances breast cancer vaccine efficacy by altering both CD8 T cell and DC components of the tumor microenvironment. Given the recent success of anti-PD-1 monotherapy, our results are encouraging for developing combination therapies for the treatment of patients with cancer in which anti-PD-1 monotherapy alone may be ineffective (i.e., PD-L1-negative tumors)., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
9. Tumor hypomethylation at 6p21.3 associates with longer time to recurrence of high-grade serous epithelial ovarian cancer.
- Author
-
Wang C, Cicek MS, Charbonneau B, Kalli KR, Armasu SM, Larson MC, Konecny GE, Winterhoff B, Fan JB, Bibikova M, Chien J, Shridhar V, Block MS, Hartmann LC, Visscher DW, Cunningham JM, Knutson KL, Fridley BL, and Goode EL
- Subjects
- Adult, Carcinoma, Ovarian Epithelial, CpG Islands, Female, Humans, Neoplasm Recurrence, Local immunology, Neoplasms, Glandular and Epithelial immunology, Ovarian Neoplasms immunology, Up-Regulation, Young Adult, Chromosomes, Human, Pair 6, DNA Methylation, Neoplasm Recurrence, Local genetics, Neoplasms, Glandular and Epithelial genetics, Ovarian Neoplasms genetics
- Abstract
To reveal biologic mechanisms underlying clinical outcome of high-grade serous (HGS) epithelial ovarian carcinomas (EOC), we evaluated the association between tumor epigenetic changes and time to recurrence (TTR). We assessed methylation at approximately 450,000 genome-wide CpGs in tumors of 337 Mayo Clinic (Rochester, MN) patients. Semi-supervised clustering of discovery (n=168) and validation (n=169) sets was used to determine clinically relevant methylation classes. Clustering identified two methylation classes based on 60 informative CpGs, which differed in TTR in the validation set [R vs. L class, P=2.9×10(-3), HR=0.52; 95% confidence interval (CI), 0.34-0.80]. Follow-up analyses considered genome-wide tumor mRNA expression (n=104) and CD8 T-cell infiltration (n=89) in patient subsets. Hypomethylation of CpGs located in 6p21.3 in the R class associated with cis upregulation of genes enriched in immune response processes (TAP1, PSMB8, PSMB9, HLA-DQB1, HLA-DQB2, HLA-DMA, and HLA-DOA), increased CD8 T-cell tumor infiltration (P=7.6×10(-5)), and trans-regulation of genes in immune-related pathways (P=1.6×10(-32)). This is the most comprehensive assessment of clinical outcomes with regard to epithelial ovarian carcinoma tumor methylation to date. Collectively, these results suggest that an epigenetically mediated immune response is a predictor of recurrence and, possibly, treatment response for HGS EOC., (©2014 American Association for Cancer Research.)
- Published
- 2014
- Full Text
- View/download PDF
10. Risk of ovarian cancer and the NF-κB pathway: genetic association with IL1A and TNFSF10.
- Author
-
Charbonneau B, Block MS, Bamlet WR, Vierkant RA, Kalli KR, Fogarty Z, Rider DN, Sellers TA, Tworoger SS, Poole E, Risch HA, Salvesen HB, Kiemeney LA, Baglietto L, Giles GG, Severi G, Trabert B, Wentzensen N, Chenevix-Trench G, Whittemore AS, Sieh W, Chang-Claude J, Bandera EV, Orlow I, Terry K, Goodman MT, Thompson PJ, Cook LS, Rossing MA, Ness RB, Narod SA, Kupryjanczyk J, Lu K, Butzow R, Dörk T, Pejovic T, Campbell I, Le ND, Bunker CH, Bogdanova N, Runnebaum IB, Eccles D, Paul J, Wu AH, Gayther SA, Hogdall E, Heitz F, Kaye SB, Karlan BY, Anton-Culver H, Gronwald J, Hogdall CK, Lambrechts D, Fasching PA, Menon U, Schildkraut J, Pearce CL, Levine DA, Kjaer SK, Cramer D, Flanagan JM, Phelan CM, Brown R, Massuger LF, Song H, Doherty JA, Krakstad C, Liang D, Odunsi K, Berchuck A, Jensen A, Lubinski J, Nevanlinna H, Bean YT, Lurie G, Ziogas A, Walsh C, Despierre E, Brinton L, Hein A, Rudolph A, Dansonka-Mieszkowska A, Olson SH, Harter P, Tyrer J, Vitonis AF, Brooks-Wilson A, Aben KK, Pike MC, Ramus SJ, Wik E, Cybulski C, Lin J, Sucheston L, Edwards R, McGuire V, Lester J, du Bois A, Lundvall L, Wang-Gohrke S, Szafron LM, Lambrechts S, Yang H, Beckmann MW, Pelttari LM, Van Altena AM, van den Berg D, Halle MK, Gentry-Maharaj A, Schwaab I, Chandran U, Menkiszak J, Ekici AB, Wilkens LR, Leminen A, Modugno F, Friel G, Rothstein JH, Vergote I, Garcia-Closas M, Hildebrandt MA, Sobiczewski P, Kelemen LE, Pharoah PD, Moysich K, Knutson KL, Cunningham JM, Fridley BL, and Goode EL
- Subjects
- Case-Control Studies, Female, Genetic Association Studies, Humans, Ovarian Neoplasms pathology, Polymorphism, Single Nucleotide, Risk, Interleukin-1alpha genetics, NF-kappa B metabolism, Ovarian Neoplasms genetics, Ovarian Neoplasms metabolism, Signal Transduction, TNF-Related Apoptosis-Inducing Ligand genetics
- Abstract
A missense single-nucleotide polymorphism (SNP) in the immune modulatory gene IL1A has been associated with ovarian cancer risk (rs17561). Although the exact mechanism through which this SNP alters risk of ovarian cancer is not clearly understood, rs17561 has also been associated with risk of endometriosis, an epidemiologic risk factor for ovarian cancer. Interleukin-1α (IL1A) is both regulated by and able to activate NF-κB, a transcription factor family that induces transcription of many proinflammatory genes and may be an important mediator in carcinogenesis. We therefore tagged SNPs in more than 200 genes in the NF-κB pathway for a total of 2,282 SNPs (including rs17561) for genotype analysis of 15,604 cases of ovarian cancer in patients of European descent, including 6,179 of high-grade serous (HGS), 2,100 endometrioid, 1,591 mucinous, 1,034 clear cell, and 1,016 low-grade serous, including 23,235 control cases spanning 40 studies in the Ovarian Cancer Association Consortium. In this large population, we confirmed the association between rs17561 and clear cell ovarian cancer [OR, 0.84; 95% confidence interval (CI), 0.76-0.93; P = 0.00075], which remained intact even after excluding participants in the prior study (OR, 0.85; 95% CI, 0.75-0.95; P = 0.006). Considering a multiple-testing-corrected significance threshold of P < 2.5 × 10(-5), only one other variant, the TNFSF10 SNP rs6785617, was associated significantly with a risk of ovarian cancer (low malignant potential tumors OR, 0.85; 95% CI, 0.79-0.91; P = 0.00002). Our results extend the evidence that borderline tumors may have a distinct genetic etiology. Further investigation of how these SNPs might modify ovarian cancer associations with other inflammation-related risk factors is warranted.
- Published
- 2014
- Full Text
- View/download PDF
11. TGFbeta/TNF(alpha)-mediated epithelial-mesenchymal transition generates breast cancer stem cells with a claudin-low phenotype.
- Author
-
Asiedu MK, Ingle JN, Behrens MD, Radisky DC, and Knutson KL
- Subjects
- Animals, Cell Line, Epithelial-Mesenchymal Transition drug effects, Female, Humans, Mammary Glands, Human drug effects, Mammary Glands, Human pathology, Mice, Mice, Transgenic, Neoplastic Stem Cells drug effects, Phenotype, Mammary Neoplasms, Experimental pathology, Neoplastic Stem Cells pathology, Transforming Growth Factor alpha pharmacology, Transforming Growth Factor beta pharmacology
- Abstract
Breast cancer recurrence is believed to be caused by a subpopulation of cancer cells that possess the stem cell attribute of treatment resistance. Recently, we and others have reported the generation of breast cancer stem cells (BCSC) by epithelial-mesenchymal transition (EMT), although the physiologic process by which these cells may arise in vivo remains unclear. We show here that exposure of tumor cells to TGFβ and TNFα induces EMT and, more importantly, generates cells with a stable BCSC phenotype which is shown by increased self-renewing capacity, greatly increased tumorigenicity, and increased resistance to oxaliplatin, etoposide, and paclitaxel. Furthermore, gene expression analyses found that the TGFβ/TNFα-derived BCSCs showed downregulated expression of genes encoding claudin 3, 4, and 7 and the luminal marker, cytokeratin 18. These changes indicate a shift to the claudin-low molecular subtype, a recently identified breast cancer subtype characterized by the expression of mesenchymal and stem cell-associated markers and correlated with a poor prognosis. Taken together, the data show that cytokine exposure can be used to generate stable BCSCs ex vivo, and suggest that these cells may provide a valuable tool in the identification of stem cell-directed biomarkers and therapies in breast cancer., (©2011 AACR.)
- Published
- 2011
- Full Text
- View/download PDF
12. Immune promotion of epithelial-mesenchymal transition and generation of breast cancer stem cells.
- Author
-
Reiman JM, Knutson KL, and Radisky DC
- Subjects
- Animals, Breast Neoplasms metabolism, CD8-Positive T-Lymphocytes cytology, Disease Progression, Epithelium pathology, Female, Humans, Mesoderm pathology, Mice, Mice, Transgenic, Models, Biological, Neoplastic Stem Cells cytology, Rats, Breast Neoplasms immunology, Breast Neoplasms therapy, Immune System, Neoplastic Stem Cells metabolism
- Abstract
Elements of the immune system act as intimate regulators of cancer progression, inhibiting early stages of tumor growth, through immunosurveillance while facilitating later stages of tumor progression. Recent findings have revealed that activated CD8 T cells can stimulate mammary epithelial tumor cells to undergo epithelial-mesenchymal transition (EMT) and to acquire the greatly increased tumorigenic capability and chemotherapeutic resistance of breast cancer stem cells (BCSC). These studies provide a window to understanding how BCSC arise and are maintained within tumors, and how to best target these processes for therapeutic benefit., ((c) 2010 AACR.)
- Published
- 2010
- Full Text
- View/download PDF
13. Immune-induced epithelial to mesenchymal transition in vivo generates breast cancer stem cells.
- Author
-
Santisteban M, Reiman JM, Asiedu MK, Behrens MD, Nassar A, Kalli KR, Haluska P, Ingle JN, Hartmann LC, Manjili MH, Radisky DC, Ferrone S, and Knutson KL
- Subjects
- Animals, CD24 Antigen immunology, CD8-Positive T-Lymphocytes immunology, DNA Repair Enzymes biosynthesis, Epithelial Cells immunology, Epithelial Cells pathology, Female, Gene Expression, Genes, erbB-2, Hyaluronan Receptors immunology, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Mesoderm immunology, Mesoderm pathology, Mice, Mice, Transgenic, Mitoxantrone pharmacology, Neoplastic Stem Cells pathology, Mammary Neoplasms, Experimental immunology, Neoplastic Stem Cells immunology
- Abstract
The breast cancer stem cell (BCSC) hypotheses suggest that breast cancer is derived from a single tumor-initiating cell with stem-like properties, but the source of these cells is unclear. We previously observed that induction of an immune response against an epithelial breast cancer led in vivo to the T-cell-dependent outgrowth of a tumor, the cells of which had undergone epithelial to mesenchymal transition (EMT). The resulting mesenchymal tumor cells had a CD24(-/lo)CD44(+) phenotype, consistent with BCSCs. In the present study, we found that EMT was induced by CD8 T cells and the resulting tumors had characteristics of BCSCs, including potent tumorigenicity, ability to reestablish an epithelial tumor, and enhanced resistance to drugs and radiation. In contrast to the hierarchal cancer stem cell hypothesis, which suggests that breast cancer arises from the transformation of a resident tissue stem cell, our results show that EMT can produce the BCSC phenotype. These findings have several important implications related to disease progression and relapse.
- Published
- 2009
- Full Text
- View/download PDF
14. An HLA-DR-degenerate epitope pool detects insulin-like growth factor binding protein 2-specific immunity in patients with cancer.
- Author
-
Kalli KR, Krco CJ, Hartmann LC, Goodman K, Maurer MJ, Yu C, Johnson EM, Erskine CL, Disis ML, Wettstein PJ, Fikes JD, Beebe M, Ishioka G, and Knutson KL
- Subjects
- Adult, CD4-Positive T-Lymphocytes metabolism, CD8-Positive T-Lymphocytes metabolism, Case-Control Studies, Dendritic Cells immunology, Enzyme-Linked Immunosorbent Assay, Epitopes, Humans, Insulin-Like Growth Factor Binding Protein 2 metabolism, Peptide Fragments immunology, Peptide Fragments metabolism, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, HLA-DR1 Antigen genetics, HLA-DR1 Antigen immunology, Insulin-Like Growth Factor Binding Protein 2 immunology, Neoplasms immunology
- Abstract
Recent studies have shown the importance of helper CD4 T cells in initiating and sustaining tumor-specific CD8 T-cell immunity. This has paved the way for identifying MHC class II epitopes that could be incorporated into class I-based vaccines. In this study, the goal was to identify an HLA-DR-degenerate epitope pool derived from insulin-like growth factor binding protein 2 (IGFBP-2). IGFBP-2, a regulator of insulin-like growth factor action, is overexpressed in the majority of breast and ovarian cancers. Using algorithms, we predicted 29 HLA-DR1-binding epitopes. Binding assays targeting 15 different HLA-DRs revealed that 10 epitopes were degenerate, binding to at least four different HLA-DR variants. An IFN-gamma enzyme-linked immunosorbent spot assay was used to assess immunity to these 10 epitopes in 48 patients with either breast or ovarian cancer and 18 controls. Elevated T-cell immunity in patients was detected in 4 of the 10 epitopes (IGFBP2.17, IGFBP2.22, IGFBP2.249, and IGFBP2.293). The cumulative T-cell frequency of these four epitopes was elevated in patients relative to controls. All four peptides are naturally processed and presented to CD4 T-cells. The degenerate pool of peptides covers nearly 80% of patients and may be useful for augmenting CD4 T-cell immunity in patients undergoing immunization.
- Published
- 2008
- Full Text
- View/download PDF
15. Signatures associated with rejection or recurrence in HER-2/neu-positive mammary tumors.
- Author
-
Worschech A, Kmieciak M, Knutson KL, Bear HD, Szalay AA, Wang E, Marincola FM, and Manjili MH
- Subjects
- Animals, Apoptosis immunology, Biomarkers, Tumor biosynthesis, Biomarkers, Tumor immunology, Cell Line, Tumor, Female, Immune Tolerance, Immunotherapy, Adoptive, Interleukin-10 biosynthesis, Interleukin-10 immunology, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental pathology, Mammary Neoplasms, Experimental therapy, Mice, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Rats, Receptor, ErbB-2 genetics, Receptors, Interleukin-10 biosynthesis, Receptors, Interleukin-10 immunology, Suppressor of Cytokine Signaling 1 Protein, Suppressor of Cytokine Signaling Proteins biosynthesis, Suppressor of Cytokine Signaling Proteins immunology, T-Lymphocytes immunology, Mammary Neoplasms, Experimental immunology, Neoplasm Recurrence, Local immunology, Receptor, ErbB-2 immunology
- Abstract
We have previously shown T-cell-mediated rejection of the neu-overexpressing mammary carcinoma cells (MMC) in wild-type FVB mice. However, following rejection of primary tumors, a fraction of animals experienced a recurrence of a neu antigen-negative variant (ANV) of MMC (tumor evasion model) after a long latency period. In the present study, we determined that T cells derived from wild-type FVB mice can specifically recognize MMC by secreting IFN-gamma and can induce apoptosis of MMC in vitro. Neu transgenic (FVBN202) mice develop spontaneous tumors and cannot reject it (tumor tolerance model). To dissect the mechanisms associated with rejection or tolerance of MMC tumors, we compared transcriptional patterns within the tumor microenvironment of MMC undergoing rejection with those that resisted it either because of tumor evasion/antigen loss recurrence (ANV tumors) or because of intrinsic tolerance mechanisms displayed by the transgenic mice. Gene profiling confirmed that immune rejection is primarily mediated through activation of IFN-stimulated genes and T-cell effector mechanisms. The tumor evasion model showed combined activation of Th1 and Th2 with a deviation toward Th2 and humoral immune responses that failed to achieve rejection likely because of lack of target antigen. Interestingly, the tumor tolerance model instead displayed immune suppression pathways through activation of regulatory mechanisms that included in particular the overexpression of interleukin-10 (IL-10), IL-10 receptor, and suppressor of cytokine signaling (SOCS)-1 and SOCS-3. These data provide a road map for the identification of novel biomarkers of immune responsiveness in clinical trials.
- Published
- 2008
- Full Text
- View/download PDF
16. Peptide vaccine given with a Toll-like receptor agonist is effective for the treatment and prevention of spontaneous breast tumors.
- Author
-
Nava-Parada P, Forni G, Knutson KL, Pease LR, and Celis E
- Subjects
- Animals, Cancer Vaccines immunology, Cancer Vaccines pharmacology, Epitopes, T-Lymphocyte immunology, Female, Interleukin-2 Receptor alpha Subunit immunology, Mammary Neoplasms, Experimental genetics, Mice, Mice, Inbred BALB C, Mice, Transgenic, Receptor, ErbB-2 biosynthesis, Receptor, ErbB-2 genetics, Receptor, ErbB-2 immunology, T-Lymphocytes, Cytotoxic immunology, T-Lymphocytes, Regulatory immunology, Toll-Like Receptors immunology, Mammary Neoplasms, Experimental immunology, Mammary Neoplasms, Experimental prevention & control, Peptides immunology, Toll-Like Receptors agonists
- Abstract
Our goal is to develop peptide vaccines that stimulate tumor antigen-specific T-cell responses against frequently found cancers. Previous work has shown that to generate effective T-cell responses, peptides have to be administered in combination with strong adjuvants such as Toll-like receptor agonists. However, most animal tumor model systems used to study peptide vaccines were not truly representative of malignant diseases in humans because they solely used transplantable tumor lines, and instead of true tumor antigens, they used highly immunogenic foreign proteins. Here, we describe a peptide vaccination strategy, which is highly effective in delaying or preventing the occurrence of spontaneous breast tumors. Transgenic female BALB-neuT mice that carry the activated rat HER-2/neu oncogene were vaccinated with a synthetic peptide from the rat HER-2/neu gene product, which represents an epitope for CTLs in combination with a Toll-like receptor agonist adjuvant. Our results show that to obtain tumor antigen-specific CTL responses and antitumor effects, the vaccine had to be administered repetitively, or the function of CD4/CD25 T regulatory cells had to be blocked with anti-CD25 antibody therapy. Mice that were vaccinated with this approach remained tumor-free or were able to control spontaneous tumor growth and exhibited long-lasting CTL responses, not only against the immunizing peptide but also against other peptides derived from rat HER-2/neu product (i.e., epitope spreading). These results suggest that similar strategies should be followed for conducting clinical studies in patients.
- Published
- 2007
- Full Text
- View/download PDF
17. The tumor antigen repertoire identified in tumor-bearing neu transgenic mice predicts human tumor antigens.
- Author
-
Lu H, Knutson KL, Gad E, and Disis ML
- Subjects
- Animals, Antibodies, Neoplasm biosynthesis, Antibodies, Neoplasm blood, Antibodies, Neoplasm immunology, Antigens, Neoplasm genetics, Antigens, Neoplasm immunology, DNA, Complementary genetics, Disease Progression, Female, Gene Expression Regulation, Neoplastic, Gene Library, Glycoproteins genetics, Glycoproteins physiology, Humans, Male, Mammary Neoplasms, Experimental genetics, Mice, Mice, Transgenic, Neoplasm Transplantation, RNA, Messenger biosynthesis, RNA, Neoplasm biosynthesis, Rats, Receptor, ErbB-2, Specific Pathogen-Free Organisms, Testis metabolism, Antigens, Neoplasm analysis, Genes, erbB-2, Mammary Neoplasms, Experimental immunology
- Abstract
FVB/N mice transgenic for nontransforming rat neu develop spontaneous breast cancers that are neu positive and estrogen receptor negative, mimicking premenopausal human breast cancer. These animals have been widely used as a model for immunobased therapies targeting HER-2/neu. In this study, we used serological analysis of recombinant cDNA expression libraries to characterize the antigenic repertoire of neu transgenic (neu-tg) mice and questioned the ability of this murine model to predict potential human tumor antigens. After screening 3 x 10(6) clones from 3 different cDNA libraries, 15 tumor antigens were identified, including cytokeratin 2-8, glutamyl-prolyl-tRNA synthetase, complement C3, galectin 8, and serine/threonine-rich protein kinase 1. Multiple proteins involved in the Rho/Rho-associated, coiled coil-containing protein kinase (Rock) signal transduction pathway were found to be immunogenic, including Rock1, Rho/Rac guanine nucleotide exchange factor 2, and schistosoma mansoni adult worm antigen preparation 70. All of the identified antigens are self-proteins that are expressed in normal tissues in addition to breast tumors and the majority of the antigens are intracellular proteins. More than half of the mouse tumor antigens have human homologues that have been reported previously as tumor antigens. Finally, the tumor-specific antibody immunity and marked immune cell infiltration that was observed in mice with spontaneous tumors were not observed in mice with transplanted tumors. Our results indicate that neu-tg mice bearing spontaneous tumors develop humoral immunity to their tumors similar to cancer patients and that tumor antigens identified in transgenic mouse may predict immunogenic human homologues.
- Published
- 2006
- Full Text
- View/download PDF
18. Tumor cells transduced with the MHC class II Transactivator and CD80 activate tumor-specific CD4+ T cells whether or not they are silenced for invariant chain.
- Author
-
Thompson JA, Dissanayake SK, Ksander BR, Knutson KL, Disis ML, and Ostrand-Rosenberg S
- Subjects
- Animals, Antigens, Neoplasm biosynthesis, B7-1 Antigen immunology, Female, Gene Expression Regulation, Genetic Vectors, Humans, Lymphocyte Activation, Mice, Nuclear Proteins biosynthesis, Nuclear Proteins physiology, RNA, Small Interfering, Retroviridae, Trans-Activators biosynthesis, Trans-Activators physiology, Transduction, Genetic, Tumor Cells, Cultured immunology, Breast Neoplasms immunology, CD4-Positive T-Lymphocytes immunology, Cancer Vaccines immunology, Nuclear Proteins genetics, Trans-Activators genetics
- Abstract
The specificity and potency of the immune system make immunotherapy a potential strategy for the treatment of cancer. To exploit this potential, we have developed cell-based cancer vaccines consisting of tumor cells expressing syngeneic MHC class II and costimulatory molecules. The vaccines mediate tumor regression in mice and activate human CD4+ T cells in vitro. Previous vaccines were generated by transducing MHC II negative tumor cells with a single HLA-DR allele. Because expression of multiple MHC II alleles would facilitate presentation of a broader repertoire of tumor antigens, we have now transduced tumor cells with the MHC class II transactivator (CIITA), a regulatory gene that coordinately increases expression of all MHC II alleles. Previous studies in mice indicated that coexpression of the MHC II accessory molecule invariant chain (Ii) inhibited presentation of endogenously synthesized tumor antigens and reduced vaccine efficacy. To determine if Ii expression affects presentation of MHC class II-restricted endogenously synthesized tumor antigens in human tumor cells, HLA-DR-MCF10 breast cancer cells were transduced with the CIITA, CD80 costimulatory molecule gene, and with or without small interfering RNAs (siRNA) specific for Ii. Ii expression is silenced >95% in CIITA/CD80/siRNA transductants; down-regulation of Ii does not affect HLA-DR expression or stability; and Ii(+) and Ii(-) transductants activate human CD4+ T cells to DRB1*0701-restricted HER-2/neu epitopes. Therefore, tumor cells transduced with the CIITA, CD80, and with or without Ii siRNA present endogenously synthesized tumor antigens and are potential vaccines for activating tumor-specific CD4+ T cells.
- Published
- 2006
- Full Text
- View/download PDF
19. Neu antigen-negative variants can be generated after neu-specific antibody therapy in neu transgenic mice.
- Author
-
Knutson KL, Almand B, Dang Y, and Disis ML
- Subjects
- Animals, Antibody Specificity, Cell Division immunology, Cell Line, Tumor, Female, Mice, Mice, Transgenic, Receptor, ErbB-2 biosynthesis, Antibodies, Monoclonal immunology, Antibodies, Monoclonal pharmacology, Immunization, Passive methods, Mammary Neoplasms, Experimental immunology, Mammary Neoplasms, Experimental therapy, Receptor, ErbB-2 immunology
- Abstract
Prolonged administration of HER-2/neu-specific monoclonal antibody therapy is now widely used for the treatment of HER-2/neu-overexpressing tumors in advanced-stage breast cancer patients. Monoclonal antibody therapy has the potential to promote reduced tumor expression of HER-2/neu by receptor down-modulation and/or the generation of antigen-negative variants. Loss of antigen by either mechanism could potentially impact subsequent therapeutic strategies targeting HER-2/neu. In this study, the effects of chronic neu-specific monoclonal antibody therapy on tumor growth and neu protein expression were examined in a murine model of neu-overexpressing breast cancer. Treatment of neu-overexpressing tumors with neu-specific antibody, in vitro or in vivo, resulted in significant tumor growth inhibition. When neu antibody was used to treat neu-overexpressing tumor cells both in vitro and in vivo in tumor-bearing mice, neu receptor expression was not diminished after cessation of therapy. However, in the setting of clinically undetectable disease in a fraction of animals, antigen-negative variants were generated. An understanding of the effects of monoclonal antibodies on target antigen expression is critical for the future design and testing of novel HER-2/neu-targeted therapies administered in combination with or after HER-2/neu-specific monoclonal antibody therapy.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.