Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Clin Cancer Res ; 24(6): 1315-1325, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29301826

ABSTRACT

Purpose: To conduct a phase I trial of a Modified Vaccinia Ankara vaccine delivering wild-type human p53 (p53MVA) in combination with gemcitabine chemotherapy in patients with platinum-resistant ovarian cancer.Experimental Design: Patients received gemcitabine on days 1 and 8 and p53MVA vaccine on day 15, during the first 3 cycles of chemotherapy. Toxicity was classified using the NCI Common Toxicity Criteria and clinical response assessed by CT scan. Peripheral blood samples were collected for immunophenotyping and monitoring of anti-p53 immune responses.Results: Eleven patients were evaluated for p53MVA/gemcitabine toxicity, clinical outcome, and immunologic response. TOXICITY: there were no DLTs, but 3 of 11 patients came off study early due to gemcitabine-attributed adverse events (AE). Minimal AEs were attributed to p53MVA vaccination. Immunologic and clinical response: enhanced in vitro recognition of p53 peptides was detectable after immunization in both the CD4+ and CD8+ T-cell compartments in 5 of 11 and 6 of 11 patients, respectively. Changes in peripheral T regulatory cells (Tregs) and myeloid-derived suppressor cells (MDSC) did not correlate significantly with vaccine response or progression-free survival (PFS). Patients with the greatest expansion of p53-reactive T cells had significantly longer PFS than patients with lower p53-reactivity after therapy. Tumor shrinkage or disease stabilization occurred in 4 patients.Conclusions: p53MVA was well tolerated, but gemcitabine without steroid pretreatment was intolerable in some patients. However, elevated p53-reactive CD4+ and CD8+ T-cell responses after therapy correlated with longer PFS. Therefore, if responses to p53MVA can be enhanced with alternative agents, superior clinical responses may be achievable. Clin Cancer Res; 24(6); 1315-25. ©2018 AACR.


Subject(s)
Cancer Vaccines/immunology , Carcinoma, Ovarian Epithelial/immunology , Carcinoma, Ovarian Epithelial/therapy , Deoxycytidine/analogs & derivatives , Drug Resistance, Neoplasm , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tumor Suppressor Protein p53/immunology , Adult , Aged , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Cancer Vaccines/therapeutic use , Carcinoma, Ovarian Epithelial/diagnosis , Carcinoma, Ovarian Epithelial/mortality , Combined Modality Therapy , Deoxycytidine/therapeutic use , Female , Humans , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Platinum/pharmacology , Platinum/therapeutic use , Treatment Outcome , Tumor Suppressor Protein p53/metabolism , Gemcitabine
2.
Cancer Immunol Immunother ; 64(11): 1357-67, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26438084

ABSTRACT

The use of peptide vaccines, enhanced by adjuvants, has shown some efficacy in clinical trials. However, responses are often short-lived and rarely induce notable memory responses. The reason is that self-antigens have already been presented to the immune system as the tumor develops, leading to tolerance or some degree of host tumor cell destruction. To try to break tolerance against self-antigens, one of the methods employed has been to modify peptides at the anchor residues to enhance their ability to bind major histocompatibility complex molecules, extending their exposure to the T-cell receptor. These modified or analogue peptides have been investigated as stimulators of the immune system in patients with different cancers with variable but sometimes notable success. In this review we describe the background and recent developments in the use of analogue peptides for the immunotherapy of acute myeloid leukemia describing knowledge useful for the application of analogue peptide treatments for other malignancies.


Subject(s)
Cancer Vaccines/therapeutic use , Leukemia, Myeloid, Acute/therapy , Vaccines, Subunit/therapeutic use , Antigens, Neoplasm/classification , Antigens, Neoplasm/immunology , Clinical Trials as Topic , Histocompatibility Antigens Class II/immunology , Humans , Leukemia, Myeloid, Acute/immunology , Nuclear Proteins/immunology , Nucleophosmin , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/immunology , Vaccination
3.
Clin Cancer Res ; 20(17): 4459-70, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-24987057

ABSTRACT

PURPOSE: To conduct a phase I trial of a modified vaccinia Ankara (MVA) vaccine delivering wild-type human p53 (p53MVA) in patients with refractory gastrointestinal cancers. EXPERIMENTAL DESIGN: Three patients were vaccinated with 1.0×10(8) plaque-forming unit (pfu) p53MVA followed by nine patients at 5.6×10(8) pfu. Toxicity was classified using the NCI Common Toxicity Criteria and clinical responses were assessed by CT scan. Peripheral blood samples were collected pre- and post-immunization for immunophenotyping, monitoring of p53MVA-induced immune response, and examination of PD1 checkpoint inhibition in vitro. RESULTS: p53MVA immunization was well tolerated at both doses, with no adverse events above grade 2. CD4+ and CD8+ T cells showing enhanced recognition of a p53 overlapping peptide library were detectable after the first immunization, particularly in the CD8+ T-cell compartment (P=0.03). However, in most patients, this did not expand further with the second and third immunization. The frequency of PD1+ T cells detectable in patients' peripheral blood mononuclear cells (PBMC) was significantly higher than in healthy controls. Furthermore, the frequency of PD1+ CD8+ T cells showed an inverse correlation with the peak CD8+ p53 response (P=0.02) and antibody blockade of PD1 in vitro increased the p53 immune responses detected after the second or third immunizations. Induction of strong T-cell and antibody responses to the MVA backbone were also apparent. CONCLUSION: p53MVA was well tolerated and induced robust CD8+ T-cell responses. Combination of p53MVA with immune checkpoint inhibition could help sustain immune responses and lead to enhanced clinical benefit.


Subject(s)
Cancer Vaccines/administration & dosage , Gastrointestinal Neoplasms/immunology , Tumor Suppressor Protein p53/genetics , Aged , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Female , Gastrointestinal Neoplasms/genetics , Gastrointestinal Neoplasms/prevention & control , Humans , Male , Middle Aged , Mutation , Tumor Suppressor Protein p53/administration & dosage , Tumor Suppressor Protein p53/immunology
4.
J Transl Med ; 7: 18, 2009 Mar 19.
Article in English | MEDLINE | ID: mdl-19298672

ABSTRACT

BACKGROUND: Optimal techniques for DC generation for immunotherapy in cancer are yet to be established. Study aims were to evaluate: (i) DC activation/maturation milieu (TNF-alpha +/- IFN-alpha) and its effects on CD8+ hTERT-specific T cell responses to class I epitopes (p540 or p865), (ii) CD8+ hTERT-specific T cell responses elicited by vaccination with class I alone or both class I and II epitope (p766 and p672)-pulsed DCs, prepared without IFN-alpha, (iii) association between circulating T regulatory cells (Tregs) and clinical responses. METHODS: Autologous DCs were generated from 10 patients (HLA-0201) with advanced cancer by culturing CD14+ blood monocytes in the presence of GM-CSF and IL-4 supplemented with TNF-alpha [DCT] or TNF-alpha and IFN-alpha [DCTI]. The capacity of the DCs to induce functional CD8+ T cell responses to hTERT HLA-0201 restricted nonapeptides was assessed by MHC tetramer binding and peptide-specific cytotoxicity. Each DC preparation (DCT or DCTI) was pulsed with only one type of hTERT peptide (p540 or p865) and both preparations were injected into separate lymph node draining regions every 2-3 weeks. This vaccination design enabled comparison of efficacy between DCT and DCTI in generating hTERT peptide specific CD8+ T cells and comparison of class I hTERT peptide (p540 or p865)-loaded DCT with or without class II cognate help (p766 and p672) in 6 patients. T regulatory cells were evaluated in 8 patients. RESULTS: (i) DCTIs and DCTs, pulsed with hTERT peptides, were comparable (p = 0.45, t-test) in inducing peptide-specific CD8+ T cell responses. (ii) Class II cognate help, significantly enhanced (p < 0.05, t-test) peptide-specific CD8+T cell responses, compared with class I pulsed DCs alone. (iii) Clinical responders had significantly lower (p < 0.05, Mann-Whitney U test) T regs, compared with non-responders. 4/16 patients experienced partial but transient clinical responses during vaccination. Vaccination was well tolerated with minimal toxicity. CONCLUSION: Addition of IFN-alpha to ex vivo monocyte-derived DCs, did not significantly enhance peptide-specific T cell responses in vivo, compared with TNF-alpha alone. Class II cognate help significantly augments peptide-specific T cell responses. Clinically favourable responses were seen in patients with low levels of circulating T regs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/immunology , HLA-D Antigens/immunology , Histocompatibility Antigens Class I/immunology , Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Cancer Vaccines/therapeutic use , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/immunology , Dinoprostone/therapeutic use , Humans , Interferon-alpha/therapeutic use , Interleukin-1/therapeutic use , Interleukin-6/therapeutic use , Lymph Nodes/immunology , Macrophages/immunology , Peptide Fragments/immunology , Telomerase/immunology , Tumor Necrosis Factor-alpha/therapeutic use
6.
Cancer Immunol Immunother ; 55(8): 1017-24, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16450142

ABSTRACT

Cell based therapies for acute myeloid leukaemia (AML) have made significant progress in the last decade benefiting the prognosis and survival of patients with this aggressive form of leukaemia. Due to advances in haematopoietic stem cell transplantation (HSCT) and particularly the advent of reduced intensity conditioning (RIC), the scope of transplantation has now extended to those patients previously ineligible due to age and health restrictions and has been associated with a decrease in transplant related mortality. The apparent graft versus leukaemia (GvL) effect observed following HSCT demonstrates the potential of the immune system to target and eradicate AML cells. Building on previously published pre-clinical studies by ourselves and others, we are now initiating a Phase I clinical study in which lentiviral vectors are used to genetically modify AML cells to express B7.1 (CD80) and IL-2. By combining allogeneic HSCT with immunisation, using the autologous AML cells expressing B7.1 and IL-2, we hope to stimulate immune eradication of residual AML cells in poor prognosis patients that have achieved donor chimerism. In this report we describe the background to cell therapy based approaches for AML, and discuss difficulties associated with the deployment of a chronically stimulated, hence exhausted/depleted immune system to eradicate tumour cells that have already escaped immune surveillance.


Subject(s)
Immunotherapy/methods , Leukemia, Myeloid/immunology , Leukemia, Myeloid/therapy , Acute Disease , Clinical Trials as Topic , Female , Genetic Vectors , Humans , Lentivirus/genetics , Tumor Escape/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...