Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Clin Pharmacol Drug Dev ; 11(8): 997-1006, 2022 08.
Article in English | MEDLINE | ID: mdl-35509219

ABSTRACT

Preclinical studies in animal models of obesity and inflammation have shown that oral administration of ARD-101, a potential TAS2R agonist, reduced food intake and body weight and downregulated inflammatory cytokines. We present results from a first-in-human phase 1 randomized, placebo-controlled trial that evaluated safety, pharmacokinetics, and pharmacodynamics of single or multiple ascending doses of oral ARD-101 (40, 100, and 240 mg) in healthy adults. A total of 43 subjects were randomly assigned and dosed to ARD-101 or placebo with 42 subjects completing the study treatment. ARD-101 was found to be >99% restricted to the gut with minimal systemic exposure, demonstrated a favorable safety profile, and was well tolerated at all dose levels. Blood samples taken 1 hour after administration showed that subjects dosed with 240 mg of ARD-101 had elevated circulating levels of several gut peptide hormones. It is postulated that ARD-101 activates enteroendocrine cells to achieve its effects regulating metabolism and inflammation. The phase 1 clinical results demonstrated safety of ARD-101 and indicated activation of gut peptide hormone release in healthy adults. Further clinical trials will evaluate ARD-101 in patients with metabolic and inflammatory disorders.


Subject(s)
Acetates , Taste , Adult , Animals , Dose-Response Relationship, Drug , Humans , Inflammation , Quaternary Ammonium Compounds
2.
Oncoimmunology ; 4(4): e1001217, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26137397

ABSTRACT

VEGFR-2 is expressed on tumor vasculature and a target for anti-angiogenic intervention. VXM01 is a first in kind orally applied tumor vaccine based on live, attenuated Salmonella bacteria carrying an expression plasmid, encoding VEGFR-2. We here studied the safety, tolerability, T effector (Teff), T regulatory (Treg) and humoral responses to VEGFR2 and anti-angiogenic effects in advanced pancreatic cancer patients in a randomized, dose escalation phase I clinical trial. Results of the first 3 mo observation period are reported. Locally advanced or metastatic, pancreatic cancer patients were enrolled. In five escalating dose groups, 30 patients received VXM01 and 15 placebo on days 1, 3, 5, and 7. Treatment was well tolerated at all dose levels. No dose-limiting toxicities were observed. Salmonella excretion and salmonella-specific humoral immune responses occurred in the two highest dose groups. VEGFR2 specific Teff, but not Treg responses were overall increased in vaccinated patients. We furthermore observed a significant reduction of tumor perfusion after 38 d in vaccinated patients together with increased levels of serum biomarkers indicative of anti-angiogenic activity, VEGF-A, and collagen IV. Vaccine specific Teff responses significantly correlated with reductions of tumor perfusion and high levels of preexisting VEGFR2-specific Teff while those showing no antiangiogenic activity had low levels of preexisting VEGFR2 specific Teff, showed a transient early increase of VEGFR2-specific Treg and reduced levels of VEGFR2-specific Teff at later time points - pointing to the possibility that early anti-angiogenic activity might be based at least in part on specific reactivation of preexisting memory T cells.

3.
BMC Cancer ; 14: 290, 2014 Apr 25.
Article in English | MEDLINE | ID: mdl-24766732

ABSTRACT

BACKGROUND: The efficacy and safety of axitinib, a potent and selective second-generation inhibitor of vascular endothelial growth factor receptors 1, 2, and 3 in combination with pemetrexed and cisplatin was evaluated in patients with advanced non-squamous non-small-cell lung cancer (NSCLC). METHODS: Overall, 170 patients were randomly assigned to receive axitinib at a starting dose of 5-mg twice daily continuously plus pemetrexed 500 mg/m(2) and cisplatin 75 mg/m(2) on day 1 of up to six 21-day cycles (arm I); axitinib on days 2 through 19 of each cycle plus pemetrexed/cisplatin (arm II); or pemetrexed/cisplatin alone (arm III). The primary endpoint was progression-free survival (PFS). RESULTS: Median PFS was 8.0, 7.9, and 7.1 months in arms I, II, and III, respectively (hazard ratio: arms I vs. III, 0.89 [P = 0.36] and arms II vs. III, 1.02 [P = 0.54]). Median overall survival was 17.0 months (arm I), 14.7 months (arm II), and 15.9 months (arm III). Objective response rates (ORRs) for axitinib-containing arms were 45.5% (arm I) and 39.7% (arm II) compared with 26.3% for pemetrexed/cisplatin alone (arm III). Gastrointestinal disorders and fatigue were frequently reported across all treatment arms. The most common all-causality grade ≥3 adverse events were hypertension in axitinib-containing arms (20% and 17%, arms I and II, respectively) and fatigue with pemetrexed/cisplatin alone (16%). CONCLUSION: Axitinib in combination with pemetrexed/cisplatin was generally well tolerated. Axitinib combinations resulted in non-significant differences in PFS and numerically higher ORR compared with chemotherapy alone in advanced NSCLC. TRIAL REGISTRATION: ClinicalTrials.gov: NCT00768755 (October 7, 2008).


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Cisplatin/administration & dosage , Glutamates/administration & dosage , Guanine/analogs & derivatives , Imidazoles/administration & dosage , Indazoles/administration & dosage , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols , Axitinib , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Female , Guanine/administration & dosage , Humans , Male , Middle Aged , Pemetrexed , Vascular Endothelial Growth Factor A/genetics
4.
J Clin Oncol ; 32(8): 752-9, 2014 Mar 10.
Article in English | MEDLINE | ID: mdl-24297945

ABSTRACT

PURPOSE: To prospectively determine the efficacy of combination therapy with temsirolimus plus bevacizumab versus interferon alfa (IFN) plus bevacizumab in metastatic renal cell carcinoma (mRCC). PATIENTS AND METHODS: In a randomized, open-label, multicenter, phase III study, patients with previously untreated predominantly clear-cell mRCC were randomly assigned, stratified by prior nephrectomy and Memorial Sloan-Kettering Cancer Center prognostic group, to receive the combination of either temsirolimus (25 mg intravenously, weekly) or IFN (9 MIU subcutaneously thrice weekly) with bevacizumab (10 mg/kg intravenously, every 2 weeks). The primary end point was independently assessed progression-free survival (PFS). RESULTS: Median PFS in patients treated with temsirolimus/bevacizumab (n = 400) versus IFN/bevacizumab (n = 391) was 9.1 and 9.3 months, respectively (hazard ratio [HR], 1.1; 95% CI, 0.9 to 1.3; P = .8). There were no significant differences in overall survival (25.8 ν 25.5 months; HR, 1.0; P = .6) or objective response rate (27.0% ν 27.4%) with temsirolimus/bevacizumab versus IFN/bevacizumab, respectively. Patients receiving temsirolimus/bevacizumab reported significantly higher overall mean scores in the Functional Assessment of Cancer Therapy-Kidney Symptom Index (FKSI) -15 and FKSI-Disease Related Symptoms subscale compared with IFN/bevacizumab (indicating improvement); however, no differences in global health outcome measures were observed. Treatment-emergent all-causality grade ≥ 3 adverse events more common (P < .001) with temsirolimus/bevacizumab were mucosal inflammation, stomatitis, hypophosphatemia, hyperglycemia, and hypercholesterolemia, whereas neutropenia was more common with IFN/bevacizumab. Incidence of pneumonitis with temsirolimus/bevacizumab was 4.8%, mostly grade 1 or 2. CONCLUSION: Temsirolimus/bevacizumab combination therapy was not superior to IFN/bevacizumab for first-line treatment in clear-cell mRCC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab , Carcinoma, Renal Cell/mortality , Carcinoma, Renal Cell/secondary , Chemotherapy, Adjuvant , Disease-Free Survival , Drug Administration Schedule , Female , Humans , Interferon-alpha/administration & dosage , Kaplan-Meier Estimate , Kidney Neoplasms/mortality , Kidney Neoplasms/pathology , Male , Middle Aged , Nephrectomy , Proportional Hazards Models , Prospective Studies , Sirolimus/administration & dosage , Sirolimus/analogs & derivatives , Time Factors , Treatment Outcome , Young Adult
5.
BMC Cancer ; 12: 361, 2012 Aug 20.
Article in English | MEDLINE | ID: mdl-22906006

ABSTRACT

BACKGROUND: The investigational oral DNA vaccine VXM01 targets the vascular endothelial growth factor receptor 2 (VEGFR-2) and uses Salmonella typhi Ty21a as a vector. The immune reaction elicited by VXM01 is expected to disrupt the tumor neovasculature and, consequently, inhibit tumor growth. VXM01 potentially combines the advantages of anti-angiogenic therapy and active immunotherapy. METHODS/DESIGN: This phase I trial examines the safety, tolerability, and immunological and clinical responses to VXM01. The randomized, placebo-controlled, double blind dose-escalation study includes up to 45 patients with locally advanced and stage IV pancreatic cancer. The patients will receive four doses of VXM01 or placebo in addition to gemcitabine as standard of care. Doses from 106 cfu up to 1010 cfu of VXM01 will be evaluated in the study. An independent data safety monitoring board (DSMB) will be involved in the dose-escalation decisions. In addition to safety as primary endpoint, the VXM01-specific immune reaction, as well as clinical response parameters will be evaluated. DISCUSSION: The results of this study shall provide the first data regarding the safety and immunogenicity of the oral anti-VEGFR-2 vaccine VXM01 in cancer patients. They will also define the recommended dose for phase II and provide the basis for further clinical evaluation, which may also include additional cancer indications. TRIAL REGISTRATION: EudraCT No.: 2011-000222-29, NCT01486329, ISRCTN68809279.


Subject(s)
Cancer Vaccines/administration & dosage , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/immunology , Vaccines, DNA/administration & dosage , Vascular Endothelial Growth Factor Receptor-2/immunology , Administration, Oral , Adult , Antimetabolites, Antineoplastic/pharmacology , Antimetabolites, Antineoplastic/therapeutic use , Cancer Vaccines/adverse effects , Cancer Vaccines/immunology , Clinical Trials, Phase I as Topic/methods , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Double-Blind Method , Humans , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/prevention & control , Pancreatic Neoplasms/blood supply , Pancreatic Neoplasms/prevention & control , Placebos , Randomized Controlled Trials as Topic/methods , Salmonella typhi/genetics , Vaccines, DNA/adverse effects , Vaccines, DNA/immunology , Vascular Endothelial Growth Factor Receptor-2/genetics , Gemcitabine
6.
Clin Cancer Res ; 17(23): 7462-9, 2011 Dec 01.
Article in English | MEDLINE | ID: mdl-21976544

ABSTRACT

PURPOSE: This multicenter, open-label, phase II study evaluated the safety and clinical activity of axitinib, a potent and selective second-generation inhibitor of vascular endothelial growth factor receptors (VEGFR)-1, 2, and 3, in patients with metastatic melanoma. EXPERIMENTAL DESIGN: Thirty-two patients with a maximum of one prior systemic therapy received axitinib at a starting dose of 5 mg twice daily. The primary endpoint was objective response rate. RESULTS: Objective response rate was 18.8% [95% confidence interval (CI), 7.2-36.4], comprising one complete and five partial responses with a median response duration of 5.9 months (95% CI, 5.0-17.0). Stable disease at 16 weeks was noted in six patients (18.8%), with an overall clinical benefit rate of 37.5%. Six-month progression-free survival rate was 33.9%, 1-year overall survival rate was 28.1%, and median overall survival was 6.6 months (95% CI, 5.2-9.0). The most frequently (>15%) reported nonhematologic, treatment-related adverse events were fatigue, hypertension, hoarseness, and diarrhea. Treatment-related fatal bowel perforation, a known class effect, occurred in one patient. Axitinib selectively decreased plasma concentrations of soluble VEGFR (sVEGFR)-2 and sVEGFR-3 compared with soluble stem cell factor receptor (sKIT). No significant association was noted between plasma levels of axitinib and response. However, post hoc analyses indicated potential relationships between efficacy endpoints and diastolic blood pressure of 90 mm Hg or higher as well as baseline serum lactate dehydrogenase levels. CONCLUSIONS: Axitinib was well tolerated, showed a selective VEGFR-inhibitory profile, and showed single-agent activity in metastatic melanoma. Further evaluations of axitinib, alone and combined with chemotherapy, are ongoing.


Subject(s)
Imidazoles/therapeutic use , Indazoles/therapeutic use , Melanoma/drug therapy , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/therapeutic use , Axitinib , Disease-Free Survival , Female , Humans , Imidazoles/administration & dosage , Imidazoles/adverse effects , Indazoles/administration & dosage , Indazoles/adverse effects , Male , Melanoma/secondary , Middle Aged , Neoplasm Metastasis , Proto-Oncogene Proteins c-kit/blood , Receptors, Vascular Endothelial Growth Factor/blood
7.
Immunol Rev ; 222: 117-28, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18363997

ABSTRACT

Four novel oral DNA vaccines provide protection against melanoma, colon, breast, and lung carcinoma in mouse models. Vaccines are delivered by attenuated Salmonella typhimurium to secondary lymphoid organs and respectively target vascular endothelial growth factor receptor-2, transcription factor Fos-related antigen-1, anti-apoptosis protein survivin and Legumain, an asparaginyl endopeptidase specifically overexpressed on tumor-associated macrophages (TAMs) in the tumor microenvironment (TME). These vaccines are all capable of inducing potent cell-mediated protective immunity against self-antigens, resulting in marked suppression of tumor growth and dissemination. Key mechanisms induced by these DNA vaccines include efficient suppression of angiogenesis in the tumor vasculature and marked activation of cytotoxic T cells, natural killer cells, and antigen-presenting dendritic cells. The vaccine targeting Legumain establishes the new paradigm whereby a reduction in the density of TAMs in the TME decreases the release of factors potentiating tumor growth and angiogenesis. This, in turn, remodels the TME and decreases its immunosuppressive milieu and thereby potentiates the DNA vaccine's ability to effectively suppress tumor cell proliferation, vascularization, and metastasis. It is anticipated that such research efforts will lead to novel DNA-based vaccines that will be effective for the treatment of cancer.


Subject(s)
Immunotherapy , Neoplasms, Experimental/therapy , Neovascularization, Pathologic/prevention & control , Vaccines, DNA/administration & dosage , Administration, Oral , Animals , CD8-Positive T-Lymphocytes/immunology , Chemokine CCL21/immunology , Gene Transfer Techniques , Immunologic Memory , Inhibitor of Apoptosis Proteins , Interleukin-18/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation , Mice , Microtubule-Associated Proteins/immunology , Neoplasm Invasiveness , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/immunology , Neovascularization, Pathologic/immunology , Neovascularization, Pathologic/pathology , Proto-Oncogene Proteins c-fos/immunology , Repressor Proteins , Salmonella typhimurium , Self Tolerance/immunology , Survivin , Vaccines, DNA/immunology , Vaccines, DNA/therapeutic use , Vascular Endothelial Growth Factor Receptor-2/immunology
8.
Arterioscler Thromb Vasc Biol ; 27(9): 2050-7, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17600223

ABSTRACT

OBJECTIVE: Vascular endothelial growth factor receptor 2 (VEGFR2)-overexpressing cells may form an interesting target for the treatment of atherosclerosis because of their involvement in processes that contribute to this disease, such as angiogenesis. METHODS AND RESULTS: We vaccinated mice against VEGFR2 by an orally administered DNA vaccine, comprising a plasmid, encoding murine VEGFR2, carried by live attenuated Salmonella typhimurium. This vaccine induces cellular immunity against cells that overexpress VEGFR2. Vaccination of hypercholesterolemic mice against VEGFR2 resulted in a marked induction of CD8+ cytotoxic T cells specific for VEGFR2 and led to an inhibition of angiogenesis in a hindlimb ischemia model. Interestingly, VEGFR2 vaccination attenuated the progression of preexisting advanced atherosclerotic lesions in the brachiocephalic artery of apoE-/- mice. Furthermore, VEGFR2 vaccination strongly reduced the initiation of collar-induced atherosclerosis in the carotid arteries of LDLr-/- mice. In addition, denudation of the carotid artery, as a model for postinterventional lesion formation, resulted in delayed endothelial replacement and significantly increased neointima formation on VEGFR2 vaccination. CONCLUSIONS: These data indicate the prominent role of VEGFR2+ cells in cardiovascular diseases and show that induction of cellular immunity against atherosclerosis-associated cells by means of DNA vaccination may contribute to the development of novel therapies against atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Hypercholesterolemia/therapy , Immunotherapy, Active/methods , Neovascularization, Pathologic/prevention & control , Vascular Endothelial Growth Factor Receptor-2/immunology , Administration, Oral , Animals , Atherosclerosis/pathology , CD8-Positive T-Lymphocytes/immunology , Coculture Techniques , Endothelial Cells/immunology , Endothelial Cells/pathology , Female , Hindlimb , Histocytochemistry , Immunity, Cellular/immunology , Ischemia/therapy , Mice , Vaccines, DNA/administration & dosage
9.
Int J Radiat Oncol Biol Phys ; 66(4): 1143-51, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-16979835

ABSTRACT

BACKGROUND: We analyzed the long-term results of patients with locally advanced rectal cancer using a multimodal approach consisting of total mesorectal excision (TME), intraoperative electron-beam radiation therapy (IOERT), and pre- or postoperative chemoradiation (CRT). PATIENTS AND METHODS: Between 1991 and 2003, 210 patients with locally advanced rectal cancer (65 International Union Against Cancer [UICC] Stage II, 116 UICC Stage III, and 29 UICC Stage IV cancers) were treated with TME, IOERT, and preoperative or postoperative CHT. A total of 122 patients were treated postoperatively; 88 patients preoperatively. Preoperative or postoperative fluoropyrimidine-based CRT was applied in 93% of these patients. RESULTS: Median age was 61 years (range, 26-81). Median follow-up was 61 months. The 5-year actuarial overall survival (OS), disease-free survival (DFS), local control rate (LC), and distant relapse free survival (DRS) of all patients was 69%, 66%, 93%, and 67%, respectively. Multivariate analysis revealed that UICC stage and resection status were the most important independent prognostic factors for OS, DFS, and DRS. The resection status was the only significant factor for local control. T-stage, tumor localization, type of resection, and type of chemotherapy had no significant impact on OS, DFS, DRS, and LC. Acute and late complications > or =Grade 3 were seen in 17% and 13% of patients, respectively. CONCLUSION: Multimodality treatment with TME and IOERT boost in combination with moderate dose pre- or postoperative CRT is feasible and results in excellent long-term local control rates in patients with intermediate to high-risk locally advanced rectal cancer.


Subject(s)
Dose Fractionation, Radiation , Drug Therapy/mortality , Radiotherapy, High-Energy/mortality , Rectal Neoplasms/mortality , Rectal Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Digestive System Surgical Procedures , Female , Germany/epidemiology , Humans , Intraoperative Care/statistics & numerical data , Longitudinal Studies , Male , Middle Aged , Sacrum , Survival Analysis , Survival Rate , Treatment Outcome
10.
J Clin Invest ; 116(7): 1955-62, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16794736

ABSTRACT

Tumor-associated fibroblasts are key regulators of tumorigenesis. In contrast to tumor cells, which are genetically unstable and mutate frequently, the presence of genetically more stable fibroblasts in the tumor-stromal compartment makes them an optimal target for cancer immunotherapy. These cells are also the primary source of collagen type I, which contributes to decreased chemotherapeutic drug uptake in tumors and plays a significant role in regulating tumor sensitivity to a variety of chemotherapies. To specifically kill tumor-associated fibroblasts, we constructed an oral DNA vaccine targeting fibroblast activation protein (FAP), which is specifically overexpressed by fibroblasts in the tumor stroma. Through CD8+ T cell-mediated killing of tumor-associated fibroblasts, our vaccine successfully suppressed primary tumor cell growth and metastasis of multidrug-resistant murine colon and breast carcinoma. Furthermore, tumor tissue of FAP-vaccinated mice revealed markedly decreased collagen type I expression and up to 70% greater uptake of chemotherapeutic drugs. Most importantly, pFap-vaccinated mice treated with chemotherapy showed a 3-fold prolongation in lifespan and marked suppression of tumor growth, with 50% of the animals completely rejecting a tumor cell challenge. This strategy opens a new venue for the combination of immuno- and chemotherapies.


Subject(s)
Cancer Vaccines , Fibroblasts/metabolism , Gelatinases/metabolism , Membrane Proteins/metabolism , Neoplasms , Serine Endopeptidases/metabolism , Vaccines, DNA , Animals , CD8-Positive T-Lymphocytes/immunology , Collagen Type I/genetics , Collagen Type I/metabolism , Endopeptidases , Female , Gelatinases/genetics , Gelatinases/therapeutic use , Membrane Proteins/genetics , Membrane Proteins/therapeutic use , Mice , Mice, Inbred BALB C , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/therapy , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/therapeutic use , Serine Endopeptidases/genetics , Serine Endopeptidases/therapeutic use , Survival Rate
11.
Int J Radiat Oncol Biol Phys ; 64(5): 1416-23, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16413697

ABSTRACT

PURPOSE: To analyze long-term prognosis and morbidity after limb-sparing treatment of patients with extremity soft-tissue sarcoma, with intraoperative electron boost radiotherapy (IOERT) followed by a moderate dose of external beam radiotherapy (EBRT). METHODS AND MATERIALS: A total of 153 patients who were treated in a single center from 1991 to 2004 were evaluated. Median IOERT dose was 15 Gy, mean EBRT dose 43 Gy (range, 40-50.4 Gy) in conventional fractionation (1.8-2 Gy). Median duration of follow-up was 33 months. Acute toxicity was assessed with Common Toxicity Criteria; late toxic effects were scored according to European Organization for Research and Treatment of Cancer/Radiation Therapy Oncology Group criteria. RESULTS: Five-year overall survival and 5-year local control rates were 77% and 78%, respectively. Whereas tumor size, patient age, and EBRT dose did not significantly affect outcome, resection status and grading were significant for survival; resection status and IOERT dose were significant for local control. Extremity salvage until death or time of follow-up was achieved in 90% of our patients, 86% of whom showed excellent limb function without impairment in activities of daily life. Acute toxicity Grade 2-4 was observed in 23% and late toxicity Grade 2-4 in 17% of patients. CONCLUSIONS: Treatment with IOERT combined with moderate doses of external beam irradiation yields high local control and extremity preservation rates in resected extremity soft-tissue sarcoma.


Subject(s)
Extremities , Limb Salvage/methods , Sarcoma/radiotherapy , Sarcoma/surgery , Adolescent , Adult , Aged , Aged, 80 and over , Electrons/therapeutic use , Female , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/surgery , Radiotherapy/methods , Sarcoma/mortality , Survival Rate
12.
Int J Radiat Oncol Biol Phys ; 64(1): 235-41, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16257132

ABSTRACT

PURPOSE: Intraoperative electron-beam radiotherapy (IOERT) has been applied for local dose escalation in over 1,400 patients in Heidelberg since 1991. Among these were 30 children, in 18 of whom IOERT was employed in radiation treatment with external-beam radiotherapy (EBRT) on account of incomplete resection. We address the question whether IOERT is able to compensate for microscopic or macroscopic tumor residue if employed in the overall radiation regimen. METHODS AND MATERIALS: The data of the aforementioned 18 children were analyzed with regard to local recurrence, overall survival, and complication rates. All children suffered from either sarcomas or neuroblastomas. In all children, IOERT was employed for local dose escalation after or before EBRT. RESULTS: After a median follow-up of 60.5 months, 15 of the treated children are alive. One local failure has been observed. Six children show clinically significant late morbidity, including the loss of a treated limb (Radiation Therapy Oncology Group Grade 4 [RTOG 4]), a severe nerve lesion (RTOG 3), an orthopedic complication (RTOG 2), a ureteral stenosis (not clinically significant), and a kidney hypotrophy (not clinically significant). In 1 child a fracture due to radionecrosis (RTOG 4) was diagnosed; however, in the follow-up, local tumor relapse was diagnosed as another possible reason for the fracture. CONCLUSIONS: Regarding the low incidence of local failure, IOERT seems to be able to compensate incomplete tumor resection in childhood sarcoma and neuroblastoma patients. The incidence of late morbidity is low enough to justify the employment of IOERT as part of the radiation treatment regimen for pediatric patients.


Subject(s)
Bone Neoplasms/radiotherapy , Neuroblastoma/radiotherapy , Retroperitoneal Neoplasms/radiotherapy , Sarcoma/radiotherapy , Adolescent , Child , Child, Preschool , Combined Modality Therapy/methods , Female , Follow-Up Studies , Humans , Infant , Intraoperative Period , Male , Neoplasm, Residual , Neuroblastoma/surgery , Retroperitoneal Neoplasms/surgery , Retroperitoneal Space , Sarcoma/surgery
13.
Clin Cancer Res ; 11(17): 6270-9, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-16144931

ABSTRACT

The involvement of alpha(v)beta3 and alpha(v)beta5 integrins in angiogenesis and the use of integrin antagonists as effective antiangiogenic agents are documented. Radiotherapy is an important therapy option for cancer. It has been shown that ionizing radiation exerts primarily antiangiogenic effects in tumors but has also proangiogenic effects as the reaction of the tumor to protect its own vasculature from radiation damage. Here, we show that combined treatment with S247, an Arg-Gly-Glu peptidomimetic antagonist of alpha(v)beta3 integrin, and external beam radiotherapy are beneficial in local tumor therapy. We found that radiation up-regulates alpha(v)beta3 expression in endothelial cells and consecutively phosphorylates Akt, which may provide a tumor escape mechanism from radiation injury mediated by integrin survival signaling. In the presence of S247, the radiation-induced Akt phosphorylation is strongly inhibited. Our studies on endothelial cell proliferation, migration, tube formation, apoptosis, and clonogenic survival show that the radiosensitivity of endothelial cells is enhanced by the concurrent administration of the integrin antagonist. The in vitro data are successfully translated into human glioma (U87), epidermoid (A431), and prostate cancer (PC3) xenograft models growing s.c. on BALB/c-nu/nu mice. In vivo, the combination of S247 treatment and fractionated radiotherapy (5 x 2.5 Gy) leads to enhanced antiangiogenic and antitumor effects compared with either monotherapies. These results underline the importance of alpha(v)beta3 integrin when tumors protect their microvasculature from radiation-induced damage. The data also indicate that the combination of integrin antagonists and radiotherapy represents a rational approach in local cancer therapy.


Subject(s)
Carcinoma, Squamous Cell/radiotherapy , Glioma/radiotherapy , Integrin alphaVbeta3/antagonists & inhibitors , Neovascularization, Pathologic/prevention & control , Organic Chemicals/therapeutic use , Prostatic Neoplasms/radiotherapy , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Squamous Cell/blood supply , Carcinoma, Squamous Cell/metabolism , Cell Movement/drug effects , Cell Movement/radiation effects , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Endothelium, Vascular/metabolism , Endothelium, Vascular/radiation effects , Glioma/blood supply , Glioma/metabolism , Humans , Integrin alphaVbeta3/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation , Prostatic Neoplasms/blood supply , Prostatic Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
14.
FASEB J ; 19(1): 158-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15498893

ABSTRACT

Acquired multidrug resistance (MDR) remains a major challenge in the treatment of cancer with chemotherapeutic drugs. It can be mediated by the up-regulated expression of different proteins within the tumor cell membrane. Here, we used murine multidrug resistance-1 (MDR-1) as a target-antigen for the immunotherapy of cancer. We successfully demonstrated that peripheral T cell tolerance can be broken by oral administration of a DNA vaccine encoding MDR-1 and carried by attenuated Salmonella typhimurium to secondary lymphoid organs. Thus, mice, immunized orally three times at 2-wk intervals and challenged 2 wk thereafter with either MDR-1 expressing CT-26 colon carcinoma cells or MDR-1 expressing Lewis lung carcinoma cells, revealed a significant increase in life span. This was evident, when compared with animals either vaccinated with the empty control vector or challenged with the parental cell lines lacking overexpression of MDR-1. The immune response induced was antigen-specific and CD8+ T cell-mediated. The presence of the target antigen led to up-regulation of activation markers on CD8+ T cells and resulted in a strong cytotoxic T cell response as well as lysis of tumor target cells in vitro. We furthermore established the vaccine to be an effective treatment for established multi-drug-resistant tumor metastases, resulting in a significantly increased life span of experimental animals. Absence of CD8+ T cells due to in vivo depletion led to abrogation of effectiveness. Taken together, our results demonstrate that T cell tolerance against the MDR-1 self-antigen can be broken. It is anticipated that the combination of such an approach with chemotherapy could lead to more effective treatments of cancer.


Subject(s)
Genes, MDR/genetics , Immunotherapy/methods , T-Lymphocytes/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/biosynthesis , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , CD8-Positive T-Lymphocytes/physiology , Cancer Vaccines , Cell Line, Tumor , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Genes, MDR/immunology , Genetic Vectors/biosynthesis , Genetic Vectors/genetics , Immunity/physiology , Immunization/methods , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Subcutaneous Tissue/metabolism , Subcutaneous Tissue/pathology , T-Lymphocytes/pathology , Transduction, Genetic/methods , Vaccination/methods
15.
Immunol Rev ; 199: 181-90, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15233734

ABSTRACT

Four novel oral DNA vaccines provide long-lived protection against melanoma, colon, breast, and non-small cell lung carcinoma in mouse model systems. The vaccines are delivered by attenuated Salmonella typhimurium to secondary lymphoid organs and are directed against targets such as carcinoembryonic antigen, tyrosine-related protein, vascular endothelial growth factor receptor-2 [also called fetal liver kinase-1 (FLK-1)], and transcription factor Fos-related antigen-1 (Fra-1). The FLK-1 and Fra-1 vaccines are effective in suppressing angiogenesis in the tumor vasculature. All four vaccines are capable of inducing potent cell-mediated protective immunity, breaking peripheral T-cell tolerance against these self-antigens resulting in effective suppression of tumor growth and metastasis. It is anticipated that such research efforts will contribute toward the rational design of future DNA vaccines that will be effective for prevention and treatment of human cancer.


Subject(s)
Cancer Vaccines/therapeutic use , Neoplasm Metastasis/therapy , Neoplasms/therapy , Vaccines, DNA/therapeutic use , Administration, Oral , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Carcinoembryonic Antigen/genetics , Carcinoembryonic Antigen/immunology , Mice , Neoplasms/immunology , Neovascularization, Pathologic/therapy , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/immunology , Salmonella typhimurium/metabolism , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/immunology
16.
Blood ; 102(1): 246-53, 2003 Jul 01.
Article in English | MEDLINE | ID: mdl-12623853

ABSTRACT

Effective therapy of high-risk leukemia with established cytotoxic drugs may be limited by poor antitumor efficacy, systemic toxicity, and the induction of drug resistance. Here, we provide the first evidence that hydrolytically activated prodrugs may overcome these problems. For this purpose, VP16 was functionally blocked by hydrolytically cleavable carbonate linkers with unique characteristics to generate 2 novel prodrugs of VP16. First, we established a more than 3-log higher efficacy of the 2 prodrugs compared with VP16 on a panel of naturally drug-resistant tumor cell lines. Second, the prodrugs did overcome VP16-induced multidrug resistance-1 gene (MDR-1)-mediated multidrug resistance in vitro in a newly established VP16-resistant T-cell leukemia cell line MOVP-3 by functionally blocking MDR-1-mediated efflux. Third, in vivo studies showed a maximum tolerated dose of ProVP16-II (> 45mg/kg), which was at least 3-fold higher than that of VP16 (15 mg/kg). Finally, tests of ProVP16-II in a multidrug-resistant xenograft model of T-cell leukemia expressing MDR-1 indicated that only the mice treated with this prodrug revealed a complete and long-lasting regression of established, drug-resistant leukemia. In summary, the hydrolytically activated etoposide prodrugs proved effective against multidrug-resistant T-cell leukemia in vitro and in vivo and provide proof of concept for a highly promising new strategy for the treatment of MDR-1 drug-resistant malignancies.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Etoposide/pharmacology , Leukemia, T-Cell/pathology , Animals , Cell Survival/drug effects , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Etoposide/chemistry , Female , Humans , Hydrolysis , Inhibitory Concentration 50 , Leukemia, T-Cell/drug therapy , Mice , Mice, Inbred Strains , Prodrugs/chemistry , Prodrugs/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
17.
Nat Med ; 8(12): 1369-75, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12415261

ABSTRACT

Tumor cells are elusive targets for immunotherapy due to their heterogeneity and genetic instability. Here we describe a novel, oral DNA vaccine that targets stable, proliferating endothelial cells in the tumor vasculature rather than tumor cells. Targeting occurs through upregulated vascular-endothelial growth factor receptor 2 (FLK-1) of proliferating endothelial cells in the tumor vasculature. This vaccine effectively protected mice from lethal challenges with melanoma, colon carcinoma and lung carcinoma cells and reduced growth of established metastases in a therapeutic setting. CTL-mediated killing of endothelial cells indicated breaking of peripheral immune tolerance against this self antigen, resulting in markedly reduced dissemination of spontaneous and experimental pulmonary metastases. Angiogenesis in the tumor vasculature was suppressed without impairment of fertility, neuromuscular performance or hematopoiesis, albeit with a slight delay in wound healing. Our strategy circumvents problems in targeting of genetically unstable tumor cells. This approach may provide a new strategy for the rational design of cancer therapies.


Subject(s)
Neoplasms, Experimental/blood supply , Neovascularization, Pathologic/prevention & control , Vaccines, DNA/immunology , Vascular Endothelial Growth Factor Receptor-2/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cytotoxicity, Immunologic , Lung Neoplasms/secondary , Mice , Mice, Inbred C57BL , Vaccination , Wound Healing
18.
J Clin Invest ; 110(2): 185-92, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12122110

ABSTRACT

Development of tumor immunotherapies focuses on inducing autoimmune responses against tumor-associated self-antigens primarily encoded by normal, unmutated genes. We hypothesized that such responses could be elicited by T cell homeostatic proliferation in the periphery, involving expansion of T cells recognizing self-MHC/peptide ligands. Herein, we demonstrate that sublethally irradiated lymphopenic mice transfused with autologous or syngeneic T cells showed tumor growth inhibition when challenged with melanoma or colon carcinoma cells. Importantly, the antitumor response depended on homeostatic expansion of a polyclonal T cell population within lymph nodes. This response was effective even for established tumors, was characterized by CD8(+) T cell-mediated tumor-specific cytotoxicity and IFN-gamma production, and was associated with long-term memory. The results indicate that concomitant induction of the physiologic processes of homeostatic T cell proliferation and tumor antigen presentation in lymph nodes triggers a beneficial antitumor autoimmune response.


Subject(s)
Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , T-Lymphocytes/immunology , Animals , Antigen Presentation , Antigens, Neoplasm , Autoantigens , Autoimmunity , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/therapy , Cytotoxicity, Immunologic , Homeostasis , Immunologic Memory , Immunotherapy , Interferon-gamma/biosynthesis , Lymphocyte Activation , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/transplantation , Vitiligo/immunology , Vitiligo/pathology
SELECTION OF CITATIONS
SEARCH DETAIL