Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Clin Cancer Res ; 24(15): 3611-3631, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29703821

ABSTRACT

Purpose: Conventional therapy for malignant glioma fails to specifically target tumor cells. In contrast, substantial evidence indicates that if appropriately redirected, T cells can precisely eradicate tumors. Here we report the rational development of a fully human bispecific antibody (hEGFRvIII-CD3 bi-scFv) that redirects human T cells to lyse malignant glioma expressing a tumor-specific mutation of the EGFR (EGFRvIII).Experimental Design: We generated a panel of bispecific single-chain variable fragments and optimized design through successive rounds of screening and refinement. We tested the ability of our lead construct to redirect naïve T cells and induce target cell-specific lysis. To test for efficacy, we evaluated tumor growth and survival in xenogeneic and syngeneic models of glioma. Tumor penetrance following intravenous drug administration was assessed in highly invasive, orthotopic glioma models.Results: A highly expressed bispecific antibody with specificity to CD3 and EGFRvIII was generated (hEGFRvIII-CD3 bi-scFv). Antibody-induced T-cell activation, secretion of proinflammatory cytokines, and proliferation was robust and occurred exclusively in the presence of target antigen. hEGFRvIII-CD3 bi-scFv was potent and target-specific, mediating significant lysis of multiple malignant glioma cell lines and patient-derived malignant glioma samples that heterogeneously express EGFRvIII. In both subcutaneous and orthotopic models, well-engrafted, patient-derived malignant glioma was effectively treated despite heterogeneity of EGFRvIII expression; intravenous hEGFRvIII-CD3 bi-scFv administration caused significant regression of tumor burden (P < 0.0001) and significantly extended survival (P < 0.0001). Similar efficacy was obtained in highly infiltrative, syngeneic glioma models, and intravenously administered hEGFRvIII-CD3 bi-scFv localized to these orthotopic tumors.Conclusions: We have developed a clinically translatable bispecific antibody that redirects human T cells to safely and effectively treat malignant glioma. On the basis of these results, we have developed a clinical study of hEGFRvIII-CD3 bi-scFv for patients with EGFRvIII-positive malignant glioma. Clin Cancer Res; 24(15); 3611-31. ©2018 AACR.


Subject(s)
CD3 Complex/antagonists & inhibitors , ErbB Receptors/antagonists & inhibitors , Glioma/drug therapy , Immunotherapy , Animals , Antibodies, Bispecific/pharmacology , CD3 Complex/immunology , Cell Line, Tumor , ErbB Receptors/immunology , Female , Gene Expression Regulation, Neoplastic/immunology , Glioma/immunology , Glioma/pathology , Humans , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Male , Mice , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Single-Chain Antibodies/immunology , Single-Chain Antibodies/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Xenograft Model Antitumor Assays
2.
Cancer Res ; 78(1): 256-264, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29093005

ABSTRACT

Median survival for glioblastoma (GBM) remains <15 months. Human cytomegalovirus (CMV) antigens have been identified in GBM but not normal brain, providing an unparalleled opportunity to subvert CMV antigens as tumor-specific immunotherapy targets. A recent trial in recurrent GBM patients demonstrated the potential clinical benefit of adoptive T-cell therapy (ATCT) of CMV phosphoprotein 65 (pp65)-specific T cells. However, ex vivo analyses from this study found no change in the capacity of CMV pp65-specific T cells to gain multiple effector functions or polyfunctionality, which has been associated with superior antitumor efficacy. Previous studies have shown that dendritic cells (DC) could further enhance tumor-specific CD8+ T-cell polyfunctionality in vivo when administered as a vaccine. Therefore, we hypothesized that vaccination with CMV pp65 RNA-loaded DCs would enhance the frequency of polyfunctional CMV pp65-specific CD8+ T cells after ATCT. Here, we report prospective results of a pilot trial in which 22 patients with newly diagnosed GBM were initially enrolled, of which 17 patients were randomized to receive CMV pp65-specific T cells with CMV-DC vaccination (CMV-ATCT-DC) or saline (CMV-ATCT-saline). Patients who received CMV-ATCT-DC vaccination experienced a significant increase in the overall frequencies of IFNγ+, TNFα+, and CCL3+ polyfunctional, CMV-specific CD8+ T cells. These increases in polyfunctional CMV-specific CD8+ T cells correlated (R = 0.7371, P = 0.0369) with overall survival, although we cannot conclude this was causally related. Our data implicate polyfunctional T-cell responses as a potential biomarker for effective antitumor immunotherapy and support a formal assessment of this combination approach in a larger randomized study.Significance: A randomized pilot trial in patients with GBM implicates polyfunctional T-cell responses as a biomarker for effective antitumor immunotherapy. Cancer Res; 78(1); 256-64. ©2017 AACR.


Subject(s)
Brain Neoplasms/therapy , Dendritic Cells/immunology , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , T-Lymphocytes/immunology , Adoptive Transfer , Adult , Aged , CD8-Positive T-Lymphocytes/immunology , Cytomegalovirus , Dendritic Cells/metabolism , Female , Humans , Male , Middle Aged , Phosphoproteins/metabolism , T-Lymphocytes/transplantation , Treatment Outcome , Viral Matrix Proteins/metabolism
3.
Clin Cancer Res ; 23(8): 1898-1909, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28411277

ABSTRACT

Purpose: Patients with glioblastoma have less than 15-month median survival despite surgical resection, high-dose radiation, and chemotherapy with temozolomide. We previously demonstrated that targeting cytomegalovirus pp65 using dendritic cells (DC) can extend survival and, in a separate study, that dose-intensified temozolomide (DI-TMZ) and adjuvant granulocyte macrophage colony-stimulating factor (GM-CSF) potentiate tumor-specific immune responses in patients with glioblastoma. Here, we evaluated pp65-specific cellular responses following DI-TMZ with pp65-DCs and determined the effects on long-term progression-free survival (PFS) and overall survival (OS).Experimental Design: Following standard-of-care, 11 patients with newly diagnosed glioblastoma received DI-TMZ (100 mg/m2/d × 21 days per cycle) with at least three vaccines of pp65 lysosome-associated membrane glycoprotein mRNA-pulsed DCs admixed with GM-CSF on day 23 ± 1 of each cycle. Thereafter, monthly DI-TMZ cycles and pp65-DCs were continued if patients had not progressed.Results: Following DI-TMZ cycle 1 and three doses of pp65-DCs, pp65 cellular responses significantly increased. After DI-TMZ, both the proportion and proliferation of regulatory T cells (Tregs) increased and remained elevated with serial DI-TMZ cycles. Median PFS and OS were 25.3 months [95% confidence interval (CI), 11.0-∞] and 41.1 months (95% CI, 21.6-∞), exceeding survival using recursive partitioning analysis and matched historical controls. Four patients remained progression-free at 59 to 64 months from diagnosis. No known prognostic factors [age, Karnofsky performance status (KPS), IDH-1/2 mutation, and MGMT promoter methylation] predicted more favorable outcomes for the patients in this cohort.Conclusions: Despite increased Treg proportions following DI-TMZ, patients receiving pp65-DCs showed long-term PFS and OS, confirming prior studies targeting cytomegalovirus in glioblastoma. Clin Cancer Res; 23(8); 1898-909. ©2017 AACR.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Dendritic Cells/transplantation , Glioblastoma/therapy , Phosphoproteins/therapeutic use , Viral Matrix Proteins/therapeutic use , Adjuvants, Immunologic , Aged , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Combined Modality Therapy , Dacarbazine/administration & dosage , Dacarbazine/analogs & derivatives , Dendritic Cells/immunology , Disease-Free Survival , Female , Glioblastoma/immunology , Glioblastoma/mortality , Granulocyte-Macrophage Colony-Stimulating Factor , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Phosphoproteins/immunology , T-Lymphocytes, Regulatory/immunology , Temozolomide , Viral Matrix Proteins/immunology
4.
Cancer Immunol Immunother ; 65(2): 205-11, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26759007

ABSTRACT

Regulatory B cells that secrete IL-10 (IL-10(+) Bregs) represent a suppressive subset of the B cell compartment with prominent anti-inflammatory capacity, capable of suppressing cellular and humoral responses to cancer and vaccines. B lymphocyte stimulator (BLyS) is a key regulatory molecule in IL-10(+) Breg biology with tightly controlled serum levels. However, BLyS levels can be drastically altered upon chemotherapeutic intervention. We have previously shown that serum BLyS levels are elevated, and directly associated, with increased antigen-specific antibody titers in patients with glioblastoma (GBM) undergoing lymphodepletive temozolomide chemotherapy and vaccination. In this study, we examined corresponding IL-10(+) Breg responses within this patient population and demonstrate that the IL-10(+) Breg compartment remains constant before and after administration of the vaccine, despite elevated BLyS levels in circulation. IL-10(+) Breg frequencies were not associated with serum BLyS levels, and ex vivo stimulation with a physiologically relevant concentration of BLyS did not increase IL-10(+) Breg frequency. However, BLyS stimulation did increase the frequency of the overall B cell compartment and promoted B cell proliferation upon B cell receptor engagement. Therefore, using BLyS as an adjuvant with therapeutic peptide vaccination could promote humoral immunity with no increase in immunosuppressive IL-10(+) Bregs. These results have implications for modulating humoral responses in human peptide vaccine trials in patients with GBM.


Subject(s)
B-Cell Activating Factor/blood , B-Lymphocytes, Regulatory/immunology , Glioblastoma/blood , Glioblastoma/immunology , Lymphocyte Count , Antibodies/blood , Antibodies/immunology , Antineoplastic Agents, Alkylating/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , B-Lymphocytes, Regulatory/metabolism , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Dacarbazine/therapeutic use , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/immunology , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Immunotherapy/methods , Interleukin-10/metabolism , Lymphocyte Activation/immunology , Temozolomide , Vaccines, Subunit/administration & dosage , Vaccines, Subunit/immunology
5.
Clin Cancer Res ; 20(4): 972-84, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24352643

ABSTRACT

PURPOSE: Chimeric antigen receptor (CAR) transduced T cells represent a promising immune therapy that has been shown to successfully treat cancers in mice and humans. However, CARs targeting antigens expressed in both tumors and normal tissues have led to significant toxicity. Preclinical studies have been limited by the use of xenograft models that do not adequately recapitulate the immune system of a clinically relevant host. A constitutively activated mutant of the naturally occurring epidermal growth factor receptor (EGFRvIII) is antigenically identical in both human and mouse glioma, but is also completely absent from any normal tissues. EXPERIMENTAL DESIGN: We developed a third-generation, EGFRvIII-specific murine CAR (mCAR), and performed tests to determine its efficacy in a fully immunocompetent mouse model of malignant glioma. RESULTS: At elevated doses, infusion with EGFRvIII mCAR T cells led to cures in all mice with brain tumors. In addition, antitumor efficacy was found to be dependent on lymphodepletive host conditioning. Selective blockade with EGFRvIII soluble peptide significantly abrogated the activity of EGFRvIII mCAR T cells in vitro and in vivo, and may offer a novel strategy to enhance the safety profile for CAR-based therapy. Finally, mCAR-treated, cured mice were resistant to rechallenge with EGFRvIII(NEG) tumors, suggesting generation of host immunity against additional tumor antigens. CONCLUSION: All together, these data support that third-generation, EGFRvIII-specific mCARs are effective against gliomas in the brain and highlight the importance of syngeneic, immunocompetent models in the preclinical evaluation of tumor immunotherapies.


Subject(s)
Adoptive Transfer , Astrocytoma/therapy , Brain Neoplasms/therapy , ErbB Receptors/metabolism , Receptors, Antigen, T-Cell/metabolism , Animals , Astrocytoma/immunology , Astrocytoma/metabolism , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , Cell Line, Tumor , ErbB Receptors/immunology , Humans , Mice , Neoplasm Transplantation , Single-Chain Antibodies/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/transplantation
6.
Cancer Immunol Immunother ; 62(6): 983-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23591978

ABSTRACT

B lymphocyte stimulator (BLyS) is a cytokine involved in differentiation and survival of follicular B cells along with humoral response potentiation. Lymphopenia is known to precipitate dramatic elevation in serum BLyS; however, the use of this effect to enhance humoral responses following vaccination has not been evaluated. We evaluated BLyS serum levels and antigen-specific antibody titers in 8 patients undergoing therapeutic temozolomide (TMZ)-induced lymphopenia, with concomitant vaccine against a tumor-specific mutation in the epidermal growth factor receptor (EGFRvIII). Our studies demonstrate that TMZ-induced lymphopenia corresponded with spikes in serum BLyS that directly preceded the induction of anti-EGFRvIII antigen-specific antibody titers, in some cases as high as 1:2,000,000. Our data are the first clinical observation of BLyS serum elevation and greatly enhanced humoral immune responses as a consequence of chemotherapy-induced lymphopenia. These observations should be considered for the development of future vaccination strategies in the setting of malignancy.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , B-Cell Activating Factor/blood , Cancer Vaccines/immunology , Dacarbazine/analogs & derivatives , Glioblastoma/immunology , Lymphocyte Depletion , Antibodies/blood , Antibody Specificity/immunology , Antineoplastic Agents, Alkylating/adverse effects , Dacarbazine/adverse effects , Dacarbazine/therapeutic use , Glioblastoma/therapy , Humans , Lymphopenia/blood , Lymphopenia/chemically induced , Temozolomide
7.
PLoS One ; 8(3): e59082, 2013.
Article in English | MEDLINE | ID: mdl-23527092

ABSTRACT

Temozolomide (TMZ) is an alkylating agent shown to prolong survival in patients with high grade glioma and is routinely used to treat melanoma brain metastases. A prominent side effect of TMZ is induction of profound lymphopenia, which some suggest may be incompatible with immunotherapy. Conversely, it has been proposed that recovery from chemotherapy-induced lymphopenia may actually be exploited to potentiate T-cell responses. Here, we report the first demonstration of TMZ as an immune host-conditioning regimen in an experimental model of brain tumor and examine its impact on antitumor efficacy of a well-characterized peptide vaccine. Our results show that high-dose, myeloablative (MA) TMZ resulted in markedly reduced CD4(+), CD8(+) T-cell and CD4(+)Foxp3(+) TReg counts. Adoptive transfer of naïve CD8(+) T cells and vaccination in this setting led to an approximately 70-fold expansion of antigen-specific CD8(+) T cells over controls. Ex vivo analysis of effector functions revealed significantly enhanced levels of pro-inflammatory cytokine secretion from mice receiving MA TMZ when compared to those treated with a lower lymphodepletive, non-myeloablative (NMA) dose. Importantly, MA TMZ, but not NMA TMZ was uniquely associated with an elevation of endogenous IL-2 serum levels, which we also show was required for optimal T-cell expansion. Accordingly, in a murine model of established intracerebral tumor, vaccination-induced immunity in the setting of MA TMZ-but not lymphodepletive, NMA TMZ-led to significantly prolonged survival. Overall, these results may be used to leverage the side-effects of a clinically-approved chemotherapy and should be considered in future study design of immune-based treatments for brain tumors.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , Dacarbazine/analogs & derivatives , Animals , Antigens/immunology , Antineoplastic Agents, Alkylating/adverse effects , Brain Neoplasms/mortality , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/immunology , Dacarbazine/adverse effects , Dacarbazine/pharmacology , Disease Models, Animal , Immunotherapy , Interleukin-2/blood , Interleukin-2/pharmacology , Lymphocyte Depletion , Lymphopenia/chemically induced , Mice , Mice, Transgenic , Temozolomide , Vaccines, Subunit/immunology
8.
PLoS One ; 7(2): e31046, 2012.
Article in English | MEDLINE | ID: mdl-22383993

ABSTRACT

BACKGROUND: Preclinical studies in mice have demonstrated that the prophylactic depletion of immunosuppressive regulatory T-cells (T(Regs)) through targeting the high affinity interleukin-2 (IL-2) receptor (IL-2Rα/CD25) can enhance anti-tumor immunotherapy. However, therapeutic approaches are complicated by the inadvertent inhibition of IL-2Rα expressing anti-tumor effector T-cells. OBJECTIVE: To determine if changes in the cytokine milieu during lymphopenia may engender differential signaling requirements that would enable unarmed anti-IL-2Rα monoclonal antibody (MAbs) to selectively deplete T(Regs) while permitting vaccine-stimulated immune responses. METHODOLOGY: A randomized placebo-controlled pilot study was undertaken to examine the ability of the anti-IL-2Rα MAb daclizumab, given at the time of epidermal growth factor receptor variant III (EGFRvIII) targeted peptide vaccination, to safely and selectively deplete T(Regs) in patients with glioblastoma (GBM) treated with lymphodepleting temozolomide (TMZ). RESULTS AND CONCLUSIONS: Daclizumab treatment (n = 3) was well-tolerated with no symptoms of autoimmune toxicity and resulted in a significant reduction in the frequency of circulating CD4+Foxp3+ TRegs in comparison to saline controls (n = 3)( p = 0.0464). A significant (p<0.0001) inverse correlation between the frequency of TRegs and the level of EGFRvIII specific humoral responses suggests the depletion of TRegs may be linked to increased vaccine-stimulated humoral immunity. These data suggest this approach deserves further study. TRIAL REGISTRATION: ClinicalTrials.gov NCT00626015.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Lymphopenia/metabolism , T-Lymphocytes, Regulatory/metabolism , Adult , Aged , Antibodies, Monoclonal, Humanized/pharmacology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Daclizumab , Female , Humans , Immune System , Immunoglobulin G/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-2 Receptor alpha Subunit/chemistry , Male , Middle Aged , Pilot Projects , T-Lymphocytes/cytology
9.
Blood ; 118(11): 3003-12, 2011 Sep 15.
Article in English | MEDLINE | ID: mdl-21768296

ABSTRACT

Lymphodepletion augments adoptive cell transfer during antitumor immunotherapy, producing dramatic clinical responses in patients with malignant melanoma. We report that the lymphopenia induced by the chemotherapeutic agent temozolomide (TMZ) enhances vaccine-driven immune responses and significantly reduces malignant growth in an established model of murine tumorigenesis. Unexpectedly, despite the improved antitumor efficacy engendered by TMZ-induced lymphopenia, there was a treatment related increase in the frequency of immunosuppressive regulatory T cells (T(Regs); P = .0006). Monoclonal antibody (mAb)-mediated inhibition of the high-affinity IL-2 receptor α (IL-2Rα/CD25) during immunotherapy in normal mice depleted T(Regs) (73% reduction; P = .0154) but also abolished vaccine-induced immune responses. However, during lymphodepletion, IL-2Rα blockade decreased T(Regs) (93% reduction; P = .0001) without impairing effector T-cell responses, to augment therapeutic antitumor efficacy (66% reduction in tumor growth; P = .0024). Of clinical relevance, we also demonstrate that anti-IL-2Rα mAb administration during recovery from lymphodepletive TMZ in patients with glioblastoma reduced T(Reg) frequency (48% reduction; P = .0061) while permitting vaccine-stimulated antitumor effector cell expansion. To our knowledge, this is the first report of systemic antibody-mediated T(Reg) depletion during lymphopenia and the consequent synergistic enhancement of vaccine-driven cellular responses, as well as the first demonstration that anti-IL-2Rα mAbs function differentially in nonlymphopenic versus lymphopenic contexts.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interleukin-2 Receptor alpha Subunit/antagonists & inhibitors , Lymphocyte Depletion/methods , Lymphopenia/immunology , T-Lymphocytes, Regulatory/drug effects , Adult , Animals , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Cells, Cultured , Combined Modality Therapy , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Daclizumab , Drug Evaluation, Preclinical , Glioblastoma/immunology , Glioblastoma/therapy , Humans , Immunoglobulin G/pharmacology , Immunoglobulin G/therapeutic use , Immunotherapy/methods , Interleukin-2 Receptor alpha Subunit/immunology , Lymphopenia/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Substrate Specificity/drug effects , Substrate Specificity/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Temozolomide , Young Adult
10.
J Clin Oncol ; 28(31): 4722-9, 2010 Nov 01.
Article in English | MEDLINE | ID: mdl-20921459

ABSTRACT

PURPOSE: Immunologic targeting of tumor-specific gene mutations may allow precise eradication of neoplastic cells without toxicity. Epidermal growth factor receptor variant III (EGFRvIII) is a constitutively activated and immunogenic mutation not expressed in normal tissues but widely expressed in glioblastoma multiforme (GBM) and other neoplasms. PATIENTS AND METHODS: A phase II, multicenter trial was undertaken to assess the immunogenicity of an EGFRvIII-targeted peptide vaccine and to estimate the progression-free survival (PFS) and overall survival (OS) of vaccinated patients with newly diagnosed EGFRvIII-expressing GBM with minimal residual disease. Intradermal vaccinations were given until toxicity or tumor progression was observed. Sample size was calculated to differentiate between PFS rates of 20% and 40% 6 months after vaccination. RESULTS: There were no symptomatic autoimmune reactions. The 6-month PFS rate after vaccination was 67% (95% CI, 40% to 83%) and after diagnosis was 94% (95% CI, 67% to 99%; n = 18). The median OS was 26.0 months (95% CI, 21.0 to 47.7 months). After adjustment for age and Karnofsky performance status, the OS of vaccinated patients was greater than that observed in a control group matched for eligibility criteria, prognostic factors, and temozolomide treatment (hazard ratio, 5.3; P = .0013; n = 17). The development of specific antibody (P = .025) or delayed-type hypersensitivity (P = .03) responses to EGFRvIII had a significant effect on OS. At recurrence, 82% (95% CI, 48% to 97%) of patients had lost EGFRvIII expression (P < .001). CONCLUSION: EGFRvIII-targeted vaccination in patients with GBM warrants investigation in a phase III, randomized trial.


Subject(s)
Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , ErbB Receptors/immunology , Glioblastoma/drug therapy , Glioblastoma/immunology , Adult , Aged , Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/diagnosis , Brain Neoplasms/mortality , Brain Neoplasms/radiotherapy , Brain Neoplasms/surgery , Chemotherapy, Adjuvant , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Dacarbazine/analogs & derivatives , Dacarbazine/therapeutic use , Disease-Free Survival , Drug Administration Schedule , Enzyme-Linked Immunosorbent Assay , Female , Gene Expression Regulation, Neoplastic , Glioblastoma/diagnosis , Glioblastoma/mortality , Glioblastoma/radiotherapy , Glioblastoma/surgery , Humans , Immunohistochemistry , Injections, Intradermal , Magnetic Resonance Imaging , Male , Middle Aged , Mutation , Predictive Value of Tests , Prognosis , Prospective Studies , Radiotherapy, Adjuvant , Sample Size , Temozolomide , Time Factors , Tumor Suppressor Proteins/genetics , United States
12.
Clin Cancer Res ; 14(18): 5759-68, 2008 Sep 15.
Article in English | MEDLINE | ID: mdl-18794085

ABSTRACT

PURPOSE: Activation of signal transducers and activators of transcription 3 (STAT3) has been identified as a central mediator of melanoma growth and metastasis. We hypothesized that WP1066, a novel STAT3 blockade agent, has marked antitumor activity, even against the melanoma metastasis to brain, a site typically refractory to therapies. EXPERIMENTAL DESIGN: The antitumor activities and related mechanisms of WP1066 were investigated both in vitro on melanoma cell lines and in vivo on mice with subcutaneously syngeneic melanoma or with intracerebral melanoma tumors. RESULTS: WP1066 achieved an IC(50) of 1.6, 2.3, and 1.5 mumol/L against melanoma cell line A375, B16, and B16EGFRvIII, respectively. WP1066 suppressed the phosphorylation of Janus-activated kinase 2 and STAT3 (Tyr705) in these cells. Tumor growth in mice with subcutaneously established syngeneic melanoma was markedly inhibited by WP1066 compared with that in controls. Long-term survival (>78 days) was observed in 80% of mice with established intracerebral syngeneic melanoma treated with 40 mg/kg of WP1066 in contrast to control mice who survived for a median of 15 days. Although WP1066 did not induce immunologic memory or enhance humoral responses to EGFRvIII, this compound reduced the production of immunosuppressive cytokines and chemokines (transforming growth factor-beta, RANTES, MCP-1, vascular endothelial growth factor), markedly inhibited natural and inducible Treg proliferation, and significantly increased cytotoxic immune responses of T cells. CONCLUSIONS: The antitumor cytotoxic effects of WP1066 and its ability to induce antitumor immune responses suggest that this compound has potential for the effective treatment of melanoma metastatic to brain.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Melanoma, Experimental/drug therapy , Pyridines/therapeutic use , STAT3 Transcription Factor/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Tyrphostins/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cytokines/metabolism , Female , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Pyridines/pharmacology , Tyrphostins/pharmacology
13.
J Immunol Methods ; 339(1): 74-81, 2008 Nov 30.
Article in English | MEDLINE | ID: mdl-18775433

ABSTRACT

The epidermal growth factor receptor variant III (EGFRvIII) is a consistent tumor-specific mutation that is widely expressed in glioblastoma multiforme (GBM) and other neoplasms. As such it represents a truly tumor-specific target for antitumor immunotherapy. Although endogenous humoral responses to EGFRvIII have been reported in patients with EGFRvIII-expressing breast cancer, it is not known whether de novo responses can be generated or endogenous responses enhanced with an EGFRvIII-specific vaccine. To assess this in clinical trials, we have developed and validated an immunoassay to measure and isolate anti-EGFRvIII and anti-KLH antibodies from the serum of patients vaccinated with an EGFRvIII-specific peptide (PEPvIII) conjugated to keyhole limpet hemocyanin (KLH). Using magnetic beads with immobilized antigen we captured and detected anti-EGFRvIII and anti-KLH antibodies in serum from patients before and after vaccinations. Using this assay, we found that significant levels of antibody for tumor-specific antigen EGFRvIII (>4 microg/mL) and KLH could be induced after vaccination with PEPvIII-KLH.


Subject(s)
Antibodies, Neoplasm/blood , Antibody Formation , Cancer Vaccines/therapeutic use , Glioblastoma/blood , Hemocyanins/therapeutic use , Receptor, Fibroblast Growth Factor, Type 3/therapeutic use , Adjuvants, Immunologic , Antibodies, Neoplasm/immunology , Antibody Formation/drug effects , Antibody Formation/immunology , Cancer Vaccines/immunology , Female , Glioblastoma/immunology , Glioblastoma/therapy , Hemocyanins/immunology , Humans , Male , Receptor, Fibroblast Growth Factor, Type 3/immunology , Vaccination/methods
14.
J Neuroimmunol ; 182(1-2): 63-72, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17137636

ABSTRACT

For the study of malignant glioma, we have previously characterized a highly tumorigenic murine astrocytoma, SMA-560, which arose spontaneously in an inbred, immunocompetent VM/Dk mouse. Using this cell line as a model of murine glioma, we performed DNA microarray analysis of autologous normal murine astroctyes (NMA) and SMA-560 tumor cells grown in monolayer culture or intracranially in syngeneic immunocompetent or immunocompromised hosts in order to determine whether tumors grown in vitro recreate the complex genetic regulation that occurs in vivo. Our findings support our hypothesis that glioma phenotype in vitro may be quite different in vivo and significantly altered by in situ growth factors and other invading cell populations.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/immunology , Gene Expression , Glioma/genetics , Glioma/immunology , Immunity/genetics , Animals , Astrocytes/metabolism , Astrocytoma/genetics , Astrocytoma/immunology , Astrocytoma/metabolism , Astrocytoma/pathology , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Cluster Analysis , Gene Expression Profiling , Glioma/metabolism , Glioma/pathology , Immunocompetence/genetics , Immunocompromised Host/genetics , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Principal Component Analysis , T-Lymphocytes/pathology
15.
Clin Cancer Res ; 12(24): 7306-15, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17189402

ABSTRACT

PURPOSE: Analyses of T-cell mRNA expression profiles in glioblastoma multiforme has not been previously reported but may help to define and characterize the immunosuppressed phenotype in patients with this type of cancer. EXPERIMENTAL DESIGN: We did microarray studies that have shown significant and fundamental differences in the expression profiles of CD4(+) and CD8(+) T cells and immunosuppressive CD4(+)CD25(+)CD45RO(+)FoxP3(+) regulatory T cells (T(reg)) from normal healthy volunteers compared with patients with newly diagnosed glioblastoma multiforme. For these investigations, we isolated total RNA from enriched CD4(+) and CD8(+) T cell or T(reg) cell populations from age-matched individuals and did microarray analyses. RESULTS: ANOVA and principal components analysis show that the various T cell compartments exhibit consistently similar mRNA expression profiles among individuals within either healthy or brain tumor groups but reflect significant differences between these groups. Compared with healthy volunteers, CD4(+) and CD8(+) T cells from patients with glioblastoma multiforme display coordinate down-regulation of genes involved in T cell receptor ligation, activation, and intracellular signaling. In contrast, T(regs) from patients with glioblastoma multiforme exhibit increased levels of transcripts involved in inhibiting host immunity. CONCLUSION: Our findings support the notion that key differences between expression profiles in T-cell populations from patients with glioblastoma multiforme results from differential expression of the immunologic transcriptome, such that a limited number of genes are principally important in producing the dysregulated T-cell phenotype.


Subject(s)
Brain Neoplasms/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Forkhead Transcription Factors/metabolism , Glioma/metabolism , Interleukin-2 Receptor alpha Subunit/metabolism , Leukocyte Common Antigens/metabolism , T-Lymphocytes/metabolism , Adult , Case-Control Studies , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Humans , Immunophenotyping/methods , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...