Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
J Texture Stud ; 54(2): 222-236, 2023 04.
Article in English | MEDLINE | ID: mdl-36790799

ABSTRACT

Undesired dough adhesion is still a challenge during the production of baked goods. There are various methods for determining the adhesive texture properties of dough. In the majority of scientific papers, dough stickiness is measured analytically by the force-distance recording of dough detachment. In this study, we describe a new multi-scale approach to compare dough adhesion phenomena in a laboratory, pilot sale and human sensory assessment. In it, the adhesive material properties of dough were investigated using a pilot scale toppling device representing dough adhesion behavior in the production process, in the laboratory by texture analysis with the Chen-Hoseney method and furthermore with a new, implemented non-oral human sensory analysis. To simulate different dough adhesion behavior, the dough mechanical and adhesion properties were varied by applying dough-modifying enzymes and different dough storage times. The structural changes in the different wheat dough system were compared by rheological characterization. By characterizing the different adhesion phenomena of the doughs, the sample with bacterial xylanase showed the highest values after 80 min of storage time in all three methods. Correlation analysis revealed a strong relationship between the detachment time (pilot scale) and human sensory assessment attributes (Force R = 0.81, Time R = 0.87, Distance R = 0.92, Stickiness R = 0.80) after 80 min of storage time. Even though human sensory assessment showed limits in the detectability of differences in dough adhesion behavior compared to the Chen-Hoseney method, it was better suited to predict machinability.


Subject(s)
Food , Mechanical Phenomena
2.
Clin Cancer Res ; 28(12): 2555-2566, 2022 06 13.
Article in English | MEDLINE | ID: mdl-35421231

ABSTRACT

PURPOSE: Immunotherapy for hepatocellular carcinoma (HCC) shows considerable promise in improving clinical outcomes. HepaVac-101 represents a single-arm, first-in-human phase I/II multicenter cancer vaccine trial for HCC (NCT03203005). It combines multipeptide antigens (IMA970A) with the TLR7/8/RIG I agonist CV8102. IMA970A includes 5 HLA-A*24 and 7 HLA-A*02 as well as 4 HLA-DR restricted peptides selected after mass spectrometric identification in human HCC tissues or cell lines. CV8102 is an RNA-based immunostimulator inducing a balanced Th1/Th2 immune response. PATIENTS AND METHODS: A total of 82 patients with very early- to intermediate-stage HCCs were enrolled and screened for suitable HLA haplotypes and 22 put on study treatment. This consisted in a single infusion of low-dose cyclophosphamide followed by nine intradermal coadministrations of IMA970A and CV8102. Only patients with no disease relapse after standard-of-care treatments were vaccinated. The primary endpoints of the HepaVac-101 clinical trial were safety, tolerability, and antigen-specific T-cell responses. Secondary or exploratory endpoints included additional immunologic parameters and survival endpoints. RESULTS: The vaccination showed a good safety profile. Transient mild-to-moderate injection-site reactions were the most frequent IMA970A/CV8102-related side effects. Immune responses against ≥1 vaccinated HLA class I tumor-associated peptide (TAA) and ≥1 vaccinated HLA class II TAA were respectively induced in 37% and 53% of the vaccinees. CONCLUSIONS: Immunotherapy may provide a great improvement in treatment options for HCC. HepaVac-101 is a first-in-human clinical vaccine trial with multiple novel HLA class I- and class II-restricted TAAs against HCC. The results are initial evidence for the safety and immunogenicity of the vaccine. Further clinical evaluations are warranted.


Subject(s)
Cancer Vaccines , Carcinoma, Hepatocellular , Liver Neoplasms , Adjuvants, Immunologic , Cancer Vaccines/adverse effects , Carcinoma, Hepatocellular/drug therapy , HLA-A Antigens , Humans , Immunotherapy/methods , Liver Neoplasms/drug therapy , Peptides
3.
A A Pract ; 14(9): e01295, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32909725

ABSTRACT

We report for the first time therapy-resistant hypernatremia (plasma sodium concentration ≥150 mmol per liter) developing in 6 of 12 critically ill coronavirus disease 2019 (COVID-19) patients age 57-84 years requiring mechanical ventilation. There was no correlation between plasma sodium concentrations and sodium input. Plasma concentrations of chloride were elevated, those of potassium decreased. These findings are consistent with abnormally increased renal sodium reabsorption, possibly caused by increased angiotensin II activity secondary to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-induced downregulation of angiotensin-converting enzyme 2 (ACE2) receptors. As hypernatremia was associated with increased length of intensive care unit stay, special attention should be paid to the electrolyte status of COVID-19 patients.


Subject(s)
Coronavirus Infections/complications , Fluid Therapy/methods , Hypernatremia/complications , Natriuretic Agents/therapeutic use , Pneumonia, Viral/complications , Aged , Aged, 80 and over , Betacoronavirus , COVID-19 , Case-Control Studies , Chlorides/blood , Cohort Studies , Coronavirus Infections/blood , Female , Fluid Therapy/statistics & numerical data , Hospital Mortality , Humans , Hypernatremia/blood , Hypernatremia/epidemiology , Hypernatremia/therapy , Length of Stay/statistics & numerical data , Male , Middle Aged , Pandemics , Pneumonia, Viral/blood , Renal Dialysis , Respiration, Artificial , Respiratory Insufficiency/etiology , Respiratory Insufficiency/therapy , Retrospective Studies , SARS-CoV-2
4.
J Immunother Cancer ; 7(1): 38, 2019 02 08.
Article in English | MEDLINE | ID: mdl-30736848

ABSTRACT

BACKGROUND: Preclinical studies demonstrate synergism between cancer immunotherapy and local radiation, enhancing anti-tumor effects and promoting immune responses. BI1361849 (CV9202) is an active cancer immunotherapeutic comprising protamine-formulated, sequence-optimized mRNA encoding six non-small cell lung cancer (NSCLC)-associated antigens (NY-ESO-1, MAGE-C1, MAGE-C2, survivin, 5T4, and MUC-1), intended to induce targeted immune responses. METHODS: We describe a phase Ib clinical trial evaluating treatment with BI1361849 combined with local radiation in 26 stage IV NSCLC patients with partial response (PR)/stable disease (SD) after standard first-line therapy. Patients were stratified into three strata (1: non-squamous NSCLC, no epidermal growth factor receptor (EGFR) mutation, PR/SD after ≥4 cycles of platinum- and pemetrexed-based treatment [n = 16]; 2: squamous NSCLC, PR/SD after ≥4 cycles of platinum-based and non-platinum compound treatment [n = 8]; 3: non-squamous NSCLC, EGFR mutation, PR/SD after ≥3 and ≤ 6 months EGFR-tyrosine kinase inhibitor (TKI) treatment [n = 2]). Patients received intradermal BI1361849, local radiation (4 × 5 Gy), then BI1361849 until disease progression. Strata 1 and 3 also had maintenance pemetrexed or continued EGFR-TKI therapy, respectively. The primary endpoint was evaluation of safety; secondary objectives included assessment of clinical efficacy (every 6 weeks during treatment) and of immune response (on Days 1 [baseline], 19 and 61). RESULTS: Study treatment was well tolerated; injection site reactions and flu-like symptoms were the most common BI1361849-related adverse events. Three patients had grade 3 BI1361849-related adverse events (fatigue, pyrexia); there was one grade 3 radiation-related event (dysphagia). In comparison to baseline, immunomonitoring revealed increased BI1361849 antigen-specific immune responses in the majority of patients (84%), whereby antigen-specific antibody levels were increased in 80% and functional T cells in 40% of patients, and involvement of multiple antigen specificities was evident in 52% of patients. One patient had a partial response in combination with pemetrexed maintenance, and 46.2% achieved stable disease as best overall response. Best overall response was SD in 57.7% for target lesions. CONCLUSION: The results support further investigation of mRNA-based immunotherapy in NSCLC including combinations with immune checkpoint inhibitors. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT01915524 .


Subject(s)
Adjuvants, Immunologic/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy , Lung Neoplasms/radiotherapy , Lung Neoplasms/therapy , Pemetrexed/therapeutic use , Protamines/therapeutic use , RNA, Messenger/therapeutic use , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Combined Modality Therapy , Female , Humans , Lung Neoplasms/immunology , Male , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Middle Aged , Mucin-1/genetics , Neoplasm Proteins/genetics , Survivin/genetics
5.
Cancer Immunol Immunother ; 68(5): 799-812, 2019 May.
Article in English | MEDLINE | ID: mdl-30770959

ABSTRACT

CV9201 is an RNActive®-based cancer immunotherapy encoding five non-small cell lung cancer-antigens: New York esophageal squamous cell carcinoma-1, melanoma antigen family C1/C2, survivin, and trophoblast glycoprotein. In a phase I/IIa dose-escalation trial, 46 patients with locally advanced (n = 7) or metastatic (n = 39) NSCLC and at least stable disease after first-line treatment received five intradermal CV9201 injections (400-1600 µg of mRNA). The primary objective of the trial was to assess safety. Secondary objectives included assessment of antibody and ex vivo T cell responses against the five antigens, and changes in immune cell populations. All CV9201 dose levels were well-tolerated and the recommended dose for phase IIa was 1600 µg. Most AEs were mild-to-moderate injection site reactions and flu-like symptoms. Three (7%) patients had grade 3 related AEs. No related grade 4/5 or related serious AEs occurred. In phase IIa, antigen-specific immune responses against ≥ 1 antigen were detected in 63% of evaluable patients after treatment. The frequency of activated IgD+CD38hi B cells increased > twofold in 18/30 (60%) evaluable patients. 9/29 (31%) evaluable patients in phase IIa had stable disease and 20/29 (69%) had progressive disease. Median progression-free and overall survival were 5.0 months (95% CI 1.8-6.3) and 10.8 months (8.1-16.7) from first administration, respectively. Two- and 3-year survival rates were 26.7% and 20.7%, respectively. CV9201 was well-tolerated and immune responses could be detected after treatment supporting further clinical investigation.


Subject(s)
B-Lymphocytes/immunology , Cancer Vaccines/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy/methods , Lung Neoplasms/therapy , RNA, Messenger/therapeutic use , T-Lymphocytes/immunology , Aged , Aged, 80 and over , Antigens, Neoplasm/genetics , Cancer Vaccines/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Cells, Cultured , Female , Humans , Immunotherapy/adverse effects , Injection Site Reaction/etiology , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lymphocyte Activation , Male , Middle Aged , Neoplasm Staging , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , RNA, Messenger/immunology , Survival Analysis
6.
Vaccine ; 37(13): 1819-1826, 2019 03 22.
Article in English | MEDLINE | ID: mdl-30797640

ABSTRACT

BACKGROUND: We report the first-in-concept human trial of the safety, tolerability and immunogenicity when a novel TLR 7/8/RIG I agonist RNA-based adjuvant, CV8102, was administered alone or mixed with fractional doses of a licensed rabies vaccine (Rabipur®) as model antigen. METHODS: The primary objective was to assess the safety and reactogenicity of various dose levels of CV8102 alone or mixed with Rabipur® in healthy 18-40 year-old male volunteers. A secondary objective was to assess the immune-enhancing potential of bedside-mixes of CV8102 with fractional doses of Rabipur® by measuring induction of rabies virus neutralising titres (VNTs). RESULTS: Fifty-six volunteers received 50-100 µg CV8102 alone (n = 11), bedside-mixed CV8102 and Rabipur® (n = 20), or Rabipur® alone (n = 25; control). When given alone or mixed with Rabipur® CV8102 caused mostly Grade 1 or 2 local or systemic reactogenicity, but no related SAEs. As 100 µg CV8102 was associated with marked CRP increases further dose escalation was stopped. Combining 25-50 µg of CV8102 with fractional doses of Rabipur® significantly improved the kinetics of VNT responses; 50 µg CV8102 also improved the magnitude of VNT responses to 1/10 Rabipur® but caused severe but self-limiting influenza-like symptoms in 2 of 14 subjects. CONCLUSIONS: Doses of 25 and 50 µg CV8102 appeared safe and with an acceptable reactogenicity profile while significantly enhancing the immunogenicity of fractional doses of rabies vaccine. EudraCT No. 2013-004514-18.


Subject(s)
Adjuvants, Immunologic , Immunogenicity, Vaccine , Rabies Vaccines/immunology , Rabies virus/immunology , Rabies/immunology , Rabies/prevention & control , Adult , Antibodies, Neutralizing , Antibodies, Viral , Healthy Volunteers , Humans , Immunization Schedule , Male , Outcome Assessment, Health Care , Rabies Vaccines/administration & dosage , Rabies Vaccines/adverse effects , Vaccination , Young Adult
7.
Lancet ; 390(10101): 1511-1520, 2017 Sep 23.
Article in English | MEDLINE | ID: mdl-28754494

ABSTRACT

BACKGROUND: Vaccines based on mRNA coding for antigens have been shown to be safe and immunogenic in preclinical models. We aimed to report results of the first-in-human proof-of-concept clinical trial in healthy adults of a prophylactic mRNA-based vaccine encoding rabies virus glycoprotein (CV7201). METHODS: We did an open-label, uncontrolled, prospective, phase 1 clinical trial at one centre in Munich, Germany. Healthy male and female volunteers (aged 18-40 years) with no history of rabies vaccination were sequentially enrolled. They received three doses of CV7201 intradermally or intramuscularly by needle-syringe or one of three needle-free devices. Escalating doses were given to subsequent cohorts, and one cohort received a booster dose after 1 year. The primary endpoint was safety and tolerability. The secondary endpoint was to determine the lowest dose of CV7201 to elicit rabies virus neutralising titres equal to or greater than the WHO-specified protective antibody titre of 0·5 IU/mL. The study is continuing for long-term safety and immunogenicity follow-up. This trial is registered with ClinicalTrials.gov, number NCT02241135. FINDINGS: Between Oct 21, 2013, and Jan 11, 2016, we enrolled and vaccinated 101 participants with 306 doses of mRNA (80-640 µg) by needle-syringe (18 intradermally and 24 intramuscularly) or needle-free devices (46 intradermally and 13 intramuscularly). In the 7 days post vaccination, 60 (94%) of 64 intradermally vaccinated participants and 36 (97%) of 37 intramuscularly vaccinated participants reported solicited injection site reactions, and 50 (78%) of 64 intradermally vaccinated participants and 29 (78%) of 37 intramuscularly vaccinated participants reported solicited systemic adverse events, including ten grade 3 events. One unexpected, possibly related, serious adverse reaction that occurred 7 days after a 640 µg intramuscular dose resolved without sequelae. mRNA vaccination by needle-free intradermal or intramuscular device injection induced virus neutralising antibody titres of 0·5 IU/mL or more across dose levels and schedules in 32 (71%) of 45 participants given 80 µg or 160 µg CV7201 doses intradermally and six (46%) of 13 participants given 200 µg or 400 µg CV7201 doses intramuscularly. 1 year later, eight (57%) of 14 participants boosted with an 80 µg needle-free intradermal dose of CV7201 achieved titres of 0·5 IU/mL or more. Conversely, intradermal or intramuscular needle-syringe injection was ineffective, with only one participant (who received 320 µg intradermally) showing a detectable immune response. INTERPRETATION: This first-ever demonstration in human beings shows that a prophylactic mRNA-based candidate vaccine can induce boostable functional antibodies against a viral antigen when administered with a needle-free device, although not when injected by a needle-syringe. The vaccine was generally safe with a reasonable tolerability profile. FUNDING: CureVac AG.


Subject(s)
Immunogenicity, Vaccine , RNA, Messenger/immunology , Rabies Vaccines/administration & dosage , Rabies/prevention & control , Adolescent , Adult , Antibodies, Neutralizing/blood , Antibodies, Viral/blood , Double-Blind Method , Drug Administration Routes , Drug Administration Schedule , Drug-Related Side Effects and Adverse Reactions , Female , Germany , Humans , Male , Prospective Studies , Rabies Vaccines/immunology , Young Adult
8.
Oncoimmunology ; 5(12): e1249560, 2016.
Article in English | MEDLINE | ID: mdl-28123889

ABSTRACT

We recently completed a phase I/IIa trial of RNActive® CV9201, a novel mRNA-based therapeutic vaccine targeting five tumor-associated antigens in non-small cell lung cancer (NSCLC) patients. The aim of the study presented here was to comprehensively analyze changes in peripheral blood during the vaccination period and to generate hypotheses facilitating the identification of potential biomarkers correlating with differential clinical outcomes post RNActive® immunotherapy. We performed whole-genome expression profiling in a subgroup of 22 stage IV NSCLC patients before and after initiation of treatment with CV9201. Utilizing an analytic approach based on blood transcriptional modules (BTMs), a previously described, sensitive tool for blood transcriptome data analysis, patients segregated into two major clusters based on transcriptional changes post RNActive® treatment. The first group of patients was characterized by the upregulation of an expression signature associated with myeloid cells and inflammation, whereas the other group exhibited an expression signature associated with T and NK cells. Patients with an enrichment of T and NK cell modules after treatment compared to baseline exhibited significantly longer progression-free and overall survival compared to patients with an upregulation of myeloid cell and inflammatory modules. Notably, these gene expression signatures were mutually exclusive and inversely correlated. Furthermore, our findings correlated with phenotypic data derived by flow cytometry as well as the neutrophil-to-lymphocyte ratio. Our study thus demonstrates non-overlapping, distinct transcriptional profiles correlating with survival warranting further validation for the development of biomarker candidates for mRNA-based immunotherapy.

9.
Cancer Immunol Res ; 4(1): 18-25, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26563311

ABSTRACT

Previous cancer vaccination trials often aimed to activate CD8(+) cytotoxic T-cell (CTL) responses with short (8-10mer) peptides and targeted CD4(+) helper T cells (TH) with HLA class II-binding longer peptides (12-16 mer) that were derived from tumor antigens. Accordingly, a study of immunomonitoring focused on the detection of CTL responses to the short, and TH responses to the long, peptides. The possible induction of concurrent TH responses to short peptides was widely neglected. In a recent phase I vaccination trial, 53 patients with different solid cancers were vaccinated with EMD640744, a cocktail of five survivin-derived short (9- or 10-mer) peptides in Montanide ISA 51VG. We monitored 49 patients and found strong CD8(+) T-cell responses in 63% of the patients. In addition, we unexpectedly found CD4(+) TH cell responses against at least two of the five short peptides in 61% (23/38) of the patients analyzed. The two peptides were recognized by HLA-DP4- and HLA-DR-restricted TH1 cells. Some short peptide-reactive (sp)CD4 T cells showed high functional avidity. Here, we show that a short peptide vaccine is able to activate a specific CD4(+) T-cell repertoire in many patients, facilitating a strong combined CD4(+)/CD8(+) T-cell response.


Subject(s)
Adjuvants, Immunologic/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Neoplasms/therapy , Vaccines, Subunit/administration & dosage , CD8-Positive T-Lymphocytes/immunology , Cell Line , Humans , Mannitol/administration & dosage , Mannitol/analogs & derivatives , Neoplasms/immunology , Oleic Acids/administration & dosage , Treatment Outcome
10.
J Immunother Cancer ; 3: 26, 2015.
Article in English | MEDLINE | ID: mdl-26082837

ABSTRACT

BACKGROUND: CV9103 is a prostate-cancer vaccine containing self-adjuvanted mRNA (RNActive®) encoding the antigens PSA, PSCA, PSMA, and STEAP1. This phase I/IIa study evaluated safety and immunogenicity of CV9103 in patients with advanced castration-resistant prostate-cancer. METHODS: 44 Patients received up to 5 intra-dermal vaccinations. Three dose levels of total mRNA were tested in Phase I in cohorts of 3-6 patients to determine a recommended dose. In phase II, 32 additional patients were treated at the recommended dose. The primary endpoint was safety and tolerability, the secondary endpoint was induction of antigen specific immune responses monitored at baseline and at weeks 5, 9 and 17. RESULTS: The most frequent adverse events were grade 1/2 injection site erythema, injection site reactions, fatigue, pyrexia, chills and influenza-like illness. Possibly treatment related urinary retention occurred in 3 patients. The recommended dose was 1280 µg. A total of 26/33 evaluable patients treated at 1280 µg developed an immune response, directed against multiple antigens in 15 out of 33 patients. One patient showed a confirmed PSA response. In the subgroup of 36 metastatic patients, the Kaplan-Meier estimate of median overall survival was 31.4 months [95 % CI: 21.2; n.a]. CONCLUSIONS: The self-adjuvanted RNActive® vaccine CV9103 was well tolerated and immunogenic. The technology is a versatile, fast and cost-effective platform allowing for creation of vaccines. The follow-up vaccine CV9104 including the additional antigens prostatic acid phosphatase (PAP) and Muc1 is currently being tested in a randomized phase IIb trial to assess the clinical benefit induced by this new vaccination approach. TRIAL REGISTRATION: EU Clinical Trials Register: EudraCT number 2008-003967-37, registered 27 Jan 2009.

11.
BMC Cancer ; 14: 748, 2014 Oct 06.
Article in English | MEDLINE | ID: mdl-25288198

ABSTRACT

BACKGROUND: Advanced non-small cell lung cancer (NSCLC) represents a significant unmet medical need. Despite advances with targeted therapies in a small subset of patients, fewer than 20% of patients survive for more than two years after diagnosis. Cancer vaccines are a promising therapeutic approach that offers the potential for durable responses through the engagement of the patient's own immune system. CV9202 is a self-adjuvanting mRNA vaccine that targets six antigens commonly expressed in NSCLC (NY-ESO-1, MAGEC1, MAGEC2, 5 T4, survivin, and MUC1). METHODS/DESIGN: The trial will assess the safety and tolerability of CV9202 vaccination combined with local radiation designed to enhance immune responses and will include patients with stage IV NSCLC and a response or stable disease after first-line chemotherapy or therapy with an EGFR tyrosine kinase inhibitor. Three histological and molecular subtypes of NSCLC will be investigated (squamous and non-squamous cell with/without EGFR mutations). All patients will receive two initial vaccinations with CV9202 prior to local radiotherapy (5 GY per day for four successive days) followed by further vaccinations until disease progression. The primary endpoint of the study is the number of patients experiencing Grade >3 treatment-related adverse events. Pharmacodynamic analyses include the assessment of immune responses to the antigens encoded by CV9202 and others not included in the panel (antigen spreading) and standard efficacy assessments. DISCUSSION: RNActive self-adjuvanted mRNA vaccines offer the potential for simultaneously inducing immune responses to a wide panel of antigens commonly expressed in tumors. This trial will assess the feasibility of this approach in combination with local radiotherapy in NSCLC patients. TRIAL REGISTRATION: Clinicaltrials.gov: NCT01915524/EudraCT No.: 2012-004230-41.


Subject(s)
Cancer Vaccines/genetics , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , RNA, Messenger/administration & dosage , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/pathology , Chemotherapy, Adjuvant , Combined Modality Therapy , Humans , Lung Neoplasms/immunology , Lung Neoplasms/pathology , RNA, Messenger/adverse effects , RNA, Messenger/therapeutic use , Radiation Dosage , Radiotherapy , Treatment Outcome
12.
Cancer Immunol Immunother ; 63(4): 381-94, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24487961

ABSTRACT

PURPOSE: Survivin is a member of the inhibitor-of-apoptosis family. Essential for tumor cell survival and overexpressed in most cancers, survivin is a promising target for anti-cancer immunotherapy. Immunogenicity has been demonstrated in multiple cancers. Nonetheless, few clinical trials have demonstrated survivin-vaccine-induced immune responses. EXPERIMENTAL DESIGN: This phase I trial was conducted to test whether vaccine EMD640744, a cocktail of five HLA class I-binding survivin peptides in Montanide(®) ISA 51 VG, promotes anti-survivin T-cell responses in patients with solid cancers. The primary objective was to compare immunologic efficacy of EMD640744 at doses of 30, 100, and 300 µg. Secondary objectives included safety, tolerability, and clinical efficacy. RESULTS: In total, 49 patients who received ≥2 EMD640744 injections with available baseline- and ≥1 post-vaccination samples [immunologic-diagnostic (ID)-intention-to-treat] were analyzed by ELISpot- and peptide/MHC-multimer staining, revealing vaccine-activated peptide-specific T-cell responses in 31 patients (63 %). This cohort included the per study protocol relevant ID population for the primary objective, i.e., T-cell responses by ELISpot in 17 weeks following first vaccination, as well as subjects who discontinued the study before week 17 but showed responses to the treatment. No dose-dependent effects were observed. In the majority of patients (61 %), anti-survivin responses were detected only after vaccination, providing evidence for de novo induction. Best overall tumor response was stable disease (28 %). EMD640744 was well tolerated; local injection-site reactions constituted the most frequent adverse event. CONCLUSIONS: Vaccination with EMD640744 elicited T-cell responses against survivin peptides in the majority of patients, demonstrating the immunologic efficacy of EMD640744.


Subject(s)
Cancer Vaccines/immunology , Epitopes, T-Lymphocyte/immunology , Inhibitor of Apoptosis Proteins/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Vaccination , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Cancer Vaccines/therapeutic use , Dose-Response Relationship, Immunologic , Female , HLA-A Antigens/immunology , HLA-B7 Antigen/immunology , Humans , Interferon-gamma Release Tests , Male , Middle Aged , Neoplasm Proteins/immunology , Neoplasms/immunology , Peptide Fragments/immunology , Survivin , T-Cell Antigen Receptor Specificity
13.
J Transl Med ; 11: 5, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23294527

ABSTRACT

BACKGROUND: EMD 521873 (Selectikine or NHS-IL2LT) is a fusion protein consisting of modified human IL-2 which binds specifically to the high-affinity IL-2 receptor, and an antibody specific for both single- and double-stranded DNA, designed to facilitate the enrichment of IL-2 in tumor tissue. METHODS: An extensive analysis of pharmacodynamic (PD) markers associated with target modulation was assessed during a first-in-human phase I dose-escalation trial of Selectikine. RESULTS: Thirty-nine patients with metastatic or locally advanced tumors refractory to standard treatments were treated with increasing doses of Selectikine, and nine further patients received additional cyclophosphamide. PD analysis, assessed during the first two treatment cycles, revealed strong activation of both CD4+ and CD8+ T-cells and only weak NK cell activation. No dose response was observed. As expected, Treg cells responded actively to Selectikine but remained at lower frequency than effector CD4+ T-cells. Interestingly, patient survival correlated positively with both high lymphocyte counts and low levels of activated CD8+ T-cells at baseline, the latter of which was associated with enhanced T-cell responses to the treatment. CONCLUSIONS: The results confirm the selectivity of Selectikine with predominant T-cell and low NK cell activation, supporting follow-up studies assessing the clinical efficacy of Selectikine for cancer patients.


Subject(s)
Antineoplastic Agents/therapeutic use , DNA/immunology , Interleukin-2/immunology , Lymphocyte Activation , Neoplasms/drug therapy , Recombinant Fusion Proteins/therapeutic use , T-Lymphocytes/cytology , Cell Proliferation , Flow Cytometry , Humans , Immunohistochemistry , Lymphocyte Count , Survival Analysis
14.
Eur J Cancer ; 49(1): 35-44, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22918078

ABSTRACT

BACKGROUND: EMD 521873 (Selectikine), an immunocytokine comprising a DNA-targeting antibody, aimed at tumour necrosis, fused with a genetically modified interleukin-2 (IL-2) moiety, was investigated in this first-in-human phase I study. METHODS: Patients had metastatic or locally advanced solid tumours failing previous standard therapy. Selectikine was administered as a 1-hour intravenous infusion on 3 consecutive days, every 3 weeks. A subgroup of patients also received 300 mg/m(2) cyclophosphamide on day 1 of each cycle. Escalating doses of Selectikine were investigated with the primary objective of determining the maximum tolerated dose (MTD). RESULTS: Thirty-nine patients were treated with Selectikine alone at dose levels from 0.075 to 0.9 mg/kg, and nine were treated at doses of 0.45 and 0.6 mg/kg in combination with cyclophosphamide. A dose-dependent linear increase of peak serum concentrations and area under curve was found. The dose-limiting toxicity was grade 3 skin rash at the 0.9 mg/kg dose-level; the MTD was 0.6 mg/kg. Rash and flu-like symptoms were the most frequent side-effects. No severe cardiovascular side-effects (hypotension or vascular leak) were observed. At all dose-levels, transient increases in total lymphocyte, eosinophil and monocyte counts were recorded. No objective tumour responses, but long periods of disease stabilisation were observed. Transient and non-neutralising Selectikine antibodies were detected in 69% of patients. CONCLUSIONS: The MTD of Selectikine with or without cyclophosphamide administered under this schedule was 0.6 mg/kg. The recommended phase II dose was 0.45-0.6 mg/kg. Selectikine had a favourable safety profile and induced biological effects typical for IL-2.


Subject(s)
Antineoplastic Agents/administration & dosage , Interleukin-2/administration & dosage , Neoplasms/drug therapy , Recombinant Fusion Proteins/administration & dosage , Adult , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Dose-Response Relationship, Drug , Female , Humans , Interleukin-2/adverse effects , Interleukin-2/pharmacokinetics , Interleukin-2/therapeutic use , Male , Maximum Tolerated Dose , Middle Aged , Recombinant Fusion Proteins/adverse effects , Recombinant Fusion Proteins/pharmacokinetics , Recombinant Fusion Proteins/therapeutic use , Young Adult
15.
J Thorac Dis ; 3(2): 79-81, 2011 Jun.
Article in English | MEDLINE | ID: mdl-22263067
16.
Anticancer Drugs ; 16(1): 39-45, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15613902

ABSTRACT

The efficacy of combination therapy with irinotecan and capecitabine has been demonstrated for the first-line treatment of metastatic colorectal cancer (MCRC). The aim of this trial was to evaluate the efficacy and safety of this combination in MCRC as second-line treatment after failure of 24-h infusional 5-fluorouracil (5-FU24h) and folinic acid (FA). Patients pre-treated with 5-FU24h/FA were recruited at two institutions to receive 6 x weekly irinotecan 70 mg/m2 and capecitabine (1000 mg/m2 b.i.d. days 1-14 and 22-35). Courses were repeated on day 50. In elderly patients (>65 years) a 20% dose reduction of both drugs was scheduled. Twenty-eight patients [M/F 20/8; median age 65 years (range 44-79); median ECOG score 1] were enrolled. The most frequent sites of metastases were liver, n=20, lymph nodes and lungs, n=10, respectively. Half of the patients had two or more metastatic sites. A total of 71 treatment courses (median 2, range 1-8) were administered. Main toxicities [worst per patient (%); CTC grade 1/2/3/4] were: anaemias 18/14/-/-; leukocytopenia 11/21/-/-; thrombocytopenia 11/-/-/-; diarrhea 18/36/21/-; nausea/vomiting 43/29/4/-; mucositis 4/11/-/-; alopecia 7/25/-/-; hand-foot syndrome 7/21/-/-; fatigue 14/14/-/-; renal insufficiency (caused by diarrhea and exsiccosis) -/-/-/7. Dose intensity in the first course was [median/mean (%)]: irinotecan 92/83; capecitabine 88/82. Twenty-three patients are evaluable for response analysis (five did not complete the first course): three patients showed partial remissions (13%) and 11 patients had stable disease (48%). Median time to progression was 3.0 months for the total population (range 1.4-17.3) and 6.5 months for responders (partial response plus no change). Seventy-four percent of the patients received a third-line therapy. Overall survival was 15.7 months calculated from the start of study treatment. Second-line therapy with irinotecan and capecitabine yielded a tumor control in 61% of patients with MCRC. Efficacy and toxicity data are comparable to 5-FU/irinotecan combinations, although the likelihood of severe diarrhea appears to be higher with capecitabine/irinotecan.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Camptothecin/analogs & derivatives , Colorectal Neoplasms/drug therapy , Deoxycytidine/analogs & derivatives , Adenocarcinoma/pathology , Adult , Aged , Camptothecin/administration & dosage , Capecitabine , Colorectal Neoplasms/pathology , Deoxycytidine/administration & dosage , Female , Fluorouracil/administration & dosage , Humans , Infusions, Intravenous , Irinotecan , Leucovorin/administration & dosage , Male , Middle Aged , Neoplasm Staging , Survival Analysis , Treatment Failure , Treatment Outcome
17.
Plant J ; 38(1): 107-18, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15053764

ABSTRACT

Viroid infection is associated with the production of short interfering RNAs (siRNAs), a hallmark of post-transcriptional gene silencing (PTGS). However, viroid RNAs autonomously replicating in the nucleus have not been shown to trigger the degradation of homologous RNA in the cytoplasm. To investigate the potential of viroids for the induction of gene silencing, non-infectious fragments of potato spindle tuber viroid (PSTVd) cDNA were transcriptionally fused to the 3' end of the green fluorescent protein (GFP)-coding region. Introduction of such constructs into tobacco plants resulted in stable transgene expression. Upon PSTVd infection, transgene expression was suppressed and partial de novo methylation of the transgene was observed. PSTVd-specific siRNA was detected but none was found corresponding to the gfp gene. Methylation was restricted almost entirely to the PSTVd-specific part of the transgene. Neither a gfp transgene construct lacking viroid-specific elements was silenced nor was de novo methylation detected, when it was introduced into the genetic background of the PSTVd-infected plant lines containing silenced GFP:PSTVd transgenes. The absence of gfp-specific siRNAs and of significant methylation within the gfp-coding region demonstrated that neither silencing nor DNA methylation spread from the initiator region into adjacent 5' regions.


Subject(s)
Nicotiana/genetics , Base Sequence , DNA Methylation , DNA, Complementary/genetics , DNA, Plant/chemistry , DNA, Plant/genetics , DNA, Viral/genetics , Gene Expression , Green Fluorescent Proteins , Luminescent Proteins/genetics , Phenotype , Plants, Genetically Modified , RNA Interference , RNA, Small Interfering/genetics , RNA, Viral/genetics , Recombinant Fusion Proteins/genetics , Nicotiana/metabolism , Nicotiana/virology , Viroids/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...