Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Cancer Res ; 81(21): 5451-5463, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34462275

ABSTRACT

Ionizing radiation (IR) and chemotherapy are mainstays of treatment for patients with rhabdomyosarcoma, yet the molecular mechanisms that underlie the success or failure of radiotherapy remain unclear. The transcriptional repressor SNAI2 was previously identified as a key regulator of IR sensitivity in normal and malignant stem cells through its repression of the proapoptotic BH3-only gene PUMA/BBC3. Here, we demonstrate a clear correlation between SNAI2 expression levels and radiosensitivity across multiple rhabdomyosarcoma cell lines. Modulating SNAI2 levels in rhabdomyosarcoma cells through its overexpression or knockdown altered radiosensitivity in vitro and in vivo. SNAI2 expression reliably promoted overall cell growth and inhibited mitochondrial apoptosis following exposure to IR, with either variable or minimal effects on differentiation and senescence, respectively. Importantly, SNAI2 knockdown increased expression of the proapoptotic BH3-only gene BIM, and chromatin immunoprecipitation sequencing experiments established that SNAI2 is a direct repressor of BIM/BCL2L11. Because the p53 pathway is nonfunctional in the rhabdomyosarcoma cells used in this study, we have identified a new, p53-independent SNAI2/BIM signaling axis that could potentially predict clinical responses to IR treatment and be exploited to improve rhabdomyosarcoma therapy. SIGNIFICANCE: SNAI2 is identified as a major regulator of radiation-induced apoptosis in rhabdomyosarcoma through previously unknown mechanisms independent of p53.


Subject(s)
Bcl-2-Like Protein 11/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Gene Expression Regulation, Neoplastic/radiation effects , Radiation, Ionizing , Rhabdomyosarcoma/prevention & control , Snail Family Transcription Factors/metabolism , Animals , Apoptosis , Bcl-2-Like Protein 11/genetics , Bcl-2-Like Protein 11/metabolism , Biomarkers, Tumor/genetics , Cell Cycle , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, SCID , RNA-Seq , Rhabdomyosarcoma/etiology , Rhabdomyosarcoma/pathology , Snail Family Transcription Factors/genetics , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
2.
Neumol. pediátr. (En línea) ; 12(3): 122-124, jul. 2017. tab
Article in Spanish | LILACS | ID: biblio-999090

ABSTRACT

Cystic adenomatoid malformation is the most frequent congenital pulmonary malformation. The usual treatment is surgical resection. However there is controversy over management in asymptomatic patients. The possible malignization would justify surgery of cystic lesions. Relation with pleuropulmonary blastoma has been described, however it is not clear whether this is a primary tumor or cyst malignization. Cystic adenomatoid malformation also has association with adenocarcinoma and rhabdomyosarcoma. Currently available evidence suggests surgical resection, despite the natural course of congenital lung cystic lesions is uncertain


La malformación adenomatoidea quística (MAQ) es la anomalía del desarrollo pulmonar más frecuente. El tratamiento habitual es la resección quirúrgica, no obstante existe controversia sobre el manejo en pacientes asintomáticos. La posible malignización de las lesiones quísticas es uno de los argumentos que justifican la cirugía en estos pacientes. Se ha descrito relación con blastoma pleuropulmonar, sin embargo no está claro si se trataría de una lesión quística que se maligniza o es una entidad diferente. También hay asociación con adenocarcinoma y rabdomiosarcoma . Actualmente se sugiere la resección quirúrgica como el tratamiento más adecuado, sin embargo la evolución natural de las lesiones quísticas pulmonares congénitas es incierta


Subject(s)
Humans , Cystic Adenomatoid Malformation of Lung, Congenital/surgery , Cystic Adenomatoid Malformation of Lung, Congenital/complications , Pulmonary Blastoma/etiology , Lung Neoplasms/etiology , Rhabdomyosarcoma/etiology , Rhabdomyosarcoma/prevention & control , Adenocarcinoma/etiology , Adenocarcinoma/prevention & control , Lung Neoplasms/prevention & control
3.
Pediatr Blood Cancer ; 63(9): 1557-62, 2016 09.
Article in English | MEDLINE | ID: mdl-27198935

ABSTRACT

BACKGROUND: Rhabdomyosarcoma (RMS) is a rare, highly malignant tumor arising from primitive mesenchymal cells that differentiate into skeletal muscle. Relatively little is known about RMS susceptibility. Based on growing evidence regarding the role of early immunologic challenges on RMS development, we evaluated the role of infections and immunizations on this clinically significant pediatric malignancy. PROCEDURE: RMS cases (n = 322) were enrolled from the third trial coordinated by the Intergroup Rhabdomyosarcoma Study Group. Population-based controls (n = 322) were pair matched to cases on race, sex, and age. The following immunizations were assessed: diphtheria, pertussis, and tetanus (DPT); measles, mumps, and rubella; and oral polio vaccine. We also evaluated if immunizations were complete versus incomplete. We examined selected infections including chickenpox, mumps, pneumonia, scarlet fever, rubella, rubeola, pertussis, mononucleosis, and lung infections. Conditional logistic regression models were used to calculate an odds ratio (OR) and 95% confidence interval (CI) for each exposure, adjusted for maternal education and total annual income. RESULTS: Incomplete immunization schedules (OR = 5.30, 95% CI: 2.47-11.33) and incomplete DPT immunization (OR = 1.56, 95% CI: 1.06-2.29) were positively associated with childhood RMS. However, infections did not appear to be associated with childhood RMS. CONCLUSIONS: This is the largest study of RMS to date demonstrating a possible protective effect of immunizations against the development of childhood RMS. Further studies are needed to validate our findings. Our findings add to the growing body of literature, suggesting a protective role of routine vaccinations in childhood cancer and specifically in childhood RMS.


Subject(s)
Infections/complications , Rhabdomyosarcoma/prevention & control , Vaccination , Adolescent , Child , Child, Preschool , Female , Humans , Male , Rhabdomyosarcoma/etiology
4.
Oncotarget ; 7(8): 8797-808, 2016 Feb 23.
Article in English | MEDLINE | ID: mdl-26848777

ABSTRACT

There is no effective drug to treat EV71 infection yet. Traditional Chinese herbs are great resources for novel antiviral compounds. Here we showed that Oblongifolin M (OM), an active compound isolated from Garcinia oblongifolia, potently inhibited EV71 infection in a dose dependent manner. To identify its potential effectors in the host cells, we successfully identified 18 proteins from 52 differentially expressed spots by comparative proteomics studies. Further studies showed that knockdown of ERp57 inhibited viral replication through downregulating viral IRES (internal ribosome entry site) activities, whereas ectopic expression of ERp57 increased IRES activity and partly rescued the inhibitory effects of OM on viral replication. We demonstrated that OM is an effective antiviral agent; and that ERp57 is one of its cellular effectors against EV71 infection.


Subject(s)
Enterovirus Infections/prevention & control , Garcinia/chemistry , Plant Extracts/pharmacology , Protein Disulfide-Isomerases/metabolism , Rhabdomyosarcoma/prevention & control , Terpenes/pharmacology , Virus Replication/drug effects , Antiviral Agents/pharmacology , Blotting, Western , Electrophoresis, Gel, Two-Dimensional , Enterovirus A, Human/pathogenicity , Enterovirus Infections/metabolism , Enterovirus Infections/pathology , Enterovirus Infections/virology , Genome, Viral , Humans , Protein Disulfide-Isomerases/antagonists & inhibitors , Protein Disulfide-Isomerases/genetics , Proteomics , RNA, Messenger/genetics , RNA, Small Interfering/genetics , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/metabolism , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/virology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tumor Cells, Cultured , Viral Proteins/metabolism
5.
Vaccine ; 32(26): 3223-31, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24731809

ABSTRACT

Recent studies have suggested that pan inhibitors of dipeptidyl peptidase-4 activity and/or structure homologs (DASH), including ARI-4175, can mediate tumor regression by immune-mediated mechanisms. This study assessed the potential of combining ARI-4175 with cancer vaccines. We evaluated ARI-4175's effect on immunogenic modulation, ability to sensitize tumor cells to antigen-specific CTL killing, effect on immune-cell subsets and function, and antitumor activity in 2 tumor models, both as a monotherapy and in combination with a recombinant viral or dendritic cell (DC)-based tumor-cell vaccine. ARI-4175's effects on the growth, surface phenotype, and antigen-specific CTL-mediated lysis of murine and human carcinoma cell lines were assessed in vitro. In vivo, C57BL-6 mice were treated orally with ARI-4175, after which splenocytes were assessed by flow cytometry and functional assays. Antitumor studies were performed in murine models of colon carcinoma (MC38-CEA(+) in CEA-transgenic C57BL-6 mice) and rhabdomyosarcoma (M3-9-M in C57BL-6 mice). Mice received oral ARI-4175 alone or in combination with a vaccine consisting of recombinant vaccinia/fowlpox CEA-TRICOM (colon model) or a DC-based tumor-cell vaccine (rhabdomyosarcoma model). Exposure to ARI-4175 had no effect on the proliferation or viability of carcinoma cells in vitro; however, it did alter tumor phenotype, making murine and human tumor cells more sensitive to antigen-specific CTL killing. Assessment of immune-cell subsets and function indicated that ARI-4175 increased levels of natural killer cells and DCs. Detrimental immune effects, including reduced T effector cells and increased immunosuppressive cells (Tregs, MDSCs), were normalized when treatment stopped, suggesting that scheduling is critical when combining this agent with vaccine. As a monotherapy, ARI-4175 had potent antitumor activity in both tumor models, and had even greater effects when combined with a vaccine (either DC-based or poxviral vector based). These findings provide the rationale for the combined use of cancer immunotherapy with DASH enzyme inhibitors such as ARI-4175.


Subject(s)
Adjuvants, Immunologic/pharmacology , Boron Compounds/pharmacology , Cancer Vaccines/immunology , Dipeptides/pharmacology , T-Lymphocytes, Cytotoxic/immunology , Administration, Oral , Animals , Cell Line, Tumor , Colonic Neoplasms/prevention & control , Dipeptidyl-Peptidase IV Inhibitors/pharmacology , Female , Humans , Lymphocyte Subsets/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Transplantation , Rhabdomyosarcoma/prevention & control
6.
Int J Cancer ; 134(2): 431-6, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-23824786

ABSTRACT

Rhabdomyosarcoma (RMS) is a highly malignant tumor of developing muscle that can occur anywhere in the body. Due to its rarity, relatively little is known about the epidemiology of RMS. Atopic disease is hypothesized to be protective against several malignancies; however, to our knowledge, there have been no assessments of atopy and childhood RMS. Therefore, we explored this association in a case-control study of 322 childhood RMS cases and 322 pair-matched controls. Cases were enrolled in a trial run by the Intergroup Rhabdomyosarcoma Study Group. Controls were matched to cases on race, sex and age. The following atopic conditions were assessed: allergies, asthma, eczema and hives; in addition, we examined other immune-related factors: birth order, day-care attendance and breastfeeding. Conditional logistic-regression models were used to calculate an odds ratio (OR) and 95% confidence interval (CI) for each exposure, adjusted for age, race, sex, household income and parental education. As the two most common histologic types of RMS are embryonal (n=215) and alveolar (n=66), we evaluated effect heterogeneity of these exposures. Allergies (OR=0.60, 95% CI: 0.41-0.87), hives (OR = 0.61, 95% CI: 0.38-0.97), day-care attendance (OR=0.48, 95% CI: 0.32-0.71) and breastfeeding for ≥ 12 months (OR=0.36, 95% CI: 0.18-0.70) were inversely associated with childhood RMS. These exposures did not display significant effect heterogeneity between histologic types (p>0.52 for all exposures). This is the first study indicating that atopic exposures may be protective against childhood RMS, suggesting additional studies are needed to evaluate the immune system's role in the development of this tumor.


Subject(s)
Asthma/complications , Eczema/complications , Hypersensitivity/complications , Rhabdomyosarcoma/etiology , Rhabdomyosarcoma/prevention & control , Urticaria/complications , Adolescent , Adult , Asthma/immunology , Case-Control Studies , Child , Child, Preschool , Eczema/immunology , Female , Follow-Up Studies , Humans , Hypersensitivity/immunology , Infant , Infant, Newborn , Male , Prognosis , Risk Factors , Urticaria/immunology , Young Adult
7.
Am J Pathol ; 182(6): 2121-31, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23562272

ABSTRACT

Cellular immunotherapy may provide a strategy to overcome the poor prognosis of metastatic and recurrent rhabdomyosarcoma (RMS) under the current regimen of polychemotherapy. Because little is known about resistance mechanisms of RMS to cytotoxic T cells, we investigated RMS cell lines and biopsy specimens for expression and function of immune costimulatory receptors and anti-apoptotic molecules by RT-PCR, Western blot analysis, IHC, and cytotoxicity assays using siRNA or transfection-modified RMS cell lines, together with engineered RMS-directed cytotoxic T cells specific for the fetal acetylcholine receptor. We found that costimulatory CD80 and CD86 were consistently absent from all RMSs tested, whereas inducible T-cell co-stimulator ligand (ICOS-L; alias B7H2) was expressed by a subset of RMSs and was inducible by tumor necrosis factor α in two of five RMS cell lines. Anti-apoptotic survivin, along with other inhibitor of apoptosis (IAP) family members (cIAP1, cIAP2, and X-linked inhibitor of apoptosis protein), was overexpressed by RMS cell lines and biopsy specimens. Down-regulation of survivin by siRNA or pharmacologically in RMS cells increased their susceptibility toward a T-cell attack, whereas induction of ICOS-L did not. Treatment of RMS-bearing Rag(-/-) mice with fetal acetylcholine receptor-specific chimeric T cells delayed xenograft growth; however, this happened without definitive tumor eradication. Combined blockade of survivin and application of chimeric T cells in vivo suppressed tumor proliferation during survivin inhibition. In conclusion, survivin blockade provides a strategy to sensitize RMS cells for T-cell-based therapy.


Subject(s)
Inhibitor of Apoptosis Proteins/immunology , Receptors, Cholinergic/immunology , Rhabdomyosarcoma/immunology , T-Lymphocytes, Cytotoxic/immunology , Animals , B7-1 Antigen/biosynthesis , B7-2 Antigen/biosynthesis , Biopsy , CD28 Antigens/immunology , Child, Preschool , Costimulatory and Inhibitory T-Cell Receptors/metabolism , Cytotoxicity, Immunologic/immunology , Female , Gene Knockdown Techniques , Humans , Immunotherapy, Adoptive/methods , Inducible T-Cell Co-Stimulator Ligand/biosynthesis , Inducible T-Cell Co-Stimulator Ligand/immunology , Infant , Inhibitor of Apoptosis Proteins/genetics , Ligands , Male , Mice , Mice, Knockout , Neoplasm Transplantation , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/prevention & control , Signal Transduction/immunology , Survivin , T-Lymphocytes/transplantation , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Necrosis Factor-alpha/immunology
8.
Ann N Y Acad Sci ; 1176: 197-204, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19796248

ABSTRACT

Pluripotent stem cells represent an almost unlimited source of most somatic cell types, providing them with great potential for cell-based therapies. The earliest methods used for generating human pluripotent stem cells as embryonic stem cells from human embryos suffered from ethical and technical drawbacks. These problems have been solved in part through the efficient induction of pluripotency in somatic cells using forced expression of a tetrad of factors. Here, we describe the formation of rhabdomyosarcomas originating from factor-induced pluripotent stem (iPS) cells derived from mouse neural stem cells. This underscores the commonly accepted notion that the use of retroviral delivery methods for inducing pluripotency will not be suited for clinical applications. However, the iPS cell field is developing rapidly. Safer protocols are now available for producing pluripotent stem cells. Here the current state-of-the-art in this field will be discussed.


Subject(s)
Pluripotent Stem Cells/pathology , Pluripotent Stem Cells/physiology , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/prevention & control , Stem Cell Transplantation/adverse effects , Animals , Cell Culture Techniques , Gene Transfer Techniques , Humans , Lentivirus/genetics , Mice , Neurons/physiology , Rhabdomyosarcoma/virology
9.
J Clin Invest ; 119(8): 2366-78, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19620785

ABSTRACT

Many microRNAs (miRNAs), posttranscriptional regulators of numerous cellular processes and developmental events, are downregulated in tumors. However, their role in tumorigenesis remains largely unknown. In this work, we examined the role of the muscle-specific miRNAs miR-1 and miR-206 in human rhabdomyosarcoma (RMS), a soft tissue sarcoma thought to arise from skeletal muscle progenitors. We have shown that miR-1 was barely detectable in primary RMS of both the embryonal and alveolar subtypes and that both miR-1 and miR-206 failed to be induced in RMS cell lines upon serum deprivation. Moreover, reexpression of miR-206 in RMS cells promoted myogenic differentiation and blocked tumor growth in xenografted mice by switching the global mRNA expression profile to one that resembled mature muscle. Finally, we showed that the product of the MET proto-oncogene, the Met tyrosine-kinase receptor, which is overexpressed in RMS and has been implicated in RMS pathogenesis, was downregulated in murine satellite cells by miR-206 at the onset of normal myogenesis. Thus, failure of posttranscriptional modulation may underlie Met overexpression in RMS and other types of cancer. We propose that tissue-specific miRNAs such as miR-1 and miR-206, given their ability to modulate hundreds of transcripts and to act as nontoxic differentiating agents, may override the genomic heterogeneity of solid tumors and ultimately hold greater therapeutic potential than single gene-directed drugs.


Subject(s)
MicroRNAs/physiology , Muscle Development/physiology , Rhabdomyosarcoma/prevention & control , Animals , Cell Cycle , Cell Differentiation , Cell Line, Tumor , Humans , Mice , MicroRNAs/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-met , Receptors, Growth Factor/antagonists & inhibitors , Receptors, Growth Factor/physiology , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Xenograft Model Antitumor Assays
10.
Int J Palliat Nurs ; 15(10): 510-4, 2009 Oct.
Article in English | MEDLINE | ID: mdl-20081723

ABSTRACT

To be truly comprehensive, palliative care for children must address more than pain control and symptom management. Holistic care also encompasses attention to the child's relationships, hopes, fears and wishes. Parents and caregivers of dying children are generally the primary decision-makers in the child's care and can find the transition from active, to palliative care, particularly difficult. Nurses who understand the parents' perspective can better support them. Children reveal their hopes and fears through play. By being attuned to symbols and themes in play, nurses can better interpret the dying child's journey. Nurses can facilitate communication and connection between parents and child and thereby promote healing during the dying process.


Subject(s)
Communication , Nurse's Role/psychology , Palliative Care , Parents/psychology , Pediatric Nursing/methods , Play Therapy/methods , Attitude to Health , Child, Preschool , Empathy , Fear , Female , Holistic Health , Humans , Mouth Neoplasms/prevention & control , Mouth Neoplasms/psychology , Nurse-Patient Relations , Palliative Care/methods , Palliative Care/psychology , Parent-Child Relations , Parents/education , Professional-Family Relations , Psychology, Child , Rhabdomyosarcoma/prevention & control , Rhabdomyosarcoma/psychology , Social Support
11.
Cancer Cell ; 14(5): 369-81, 2008 Nov 04.
Article in English | MEDLINE | ID: mdl-18977326

ABSTRACT

Studies support the importance of microRNAs in physiological and pathological processes. Here we describe the regulation and function of miR-29 in myogenesis and rhabdomyosarcoma (RMS). Results demonstrate that in myoblasts, miR-29 is repressed by NF-kappaB acting through YY1 and the Polycomb group. During myogenesis, NF-kappaB and YY1 downregulation causes derepression of miR-29, which in turn accelerates differentiation by targeting its repressor YY1. However, in RMS cells and primary tumors that possess impaired differentiation, miR-29 is epigenetically silenced by an activated NF-kappaB-YY1 pathway. Reconstitution of miR-29 in RMS in mice inhibits tumor growth and stimulates differentiation, suggesting that miR-29 acts as a tumor suppressor through its promyogenic function. Together, these results identify a NF-kappaB-YY1-miR-29 regulatory circuit whose disruption may contribute to RMS.


Subject(s)
Gene Expression Regulation, Developmental , MicroRNAs/metabolism , Muscle Development/physiology , Myoblasts, Skeletal/cytology , NF-kappa B/metabolism , Rhabdomyosarcoma/metabolism , YY1 Transcription Factor/metabolism , Animals , Blotting, Western , Cell Cycle/physiology , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Chromatin Immunoprecipitation , Computational Biology , Down-Regulation , Feedback, Physiological , Fibroblasts , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Myoblasts, Skeletal/metabolism , NF-kappa B/genetics , Nucleic Acid Conformation , Promoter Regions, Genetic , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/prevention & control , Signal Transduction , YY1 Transcription Factor/genetics
12.
Int J Cancer ; 94(5): 645-51, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11745458

ABSTRACT

Rhabdomyosarcoma is the most common soft-tissue sarcoma of childhood. Rhabdomyosarcoma cell lines overexpress insulin-like growth factor-II (IGF-II), an autocrine growth factor that is inhibited by insulin-like growth factor binding protein-6 (IGFBP-6). IGFBP-6 is associated with myoblast quiescence, and expression in rhabdomyosarcoma cells is low. The effect of IGFBP-6 on 2 rhabdomyosarcoma cell lines, RD and Rh30, was studied. IGFBP-6 inhibited anchorage-dependent growth of RD and Rh30 cells in a dose-dependent manner (p < 0.0001). IGFBP-6 also inhibited anchorage-independent growth of RD cells in soft agar in a dose-dependent manner (p < 0.01). Anchorage-independent growth of RD cells on polyhydroxyethylmethacrylate-coated plates was decreased to a minimum of 48% of control after treatment with IGFBP-6 (p < 0.001). In this system, IGFBP-6 increased apoptosis 4-fold (p < 0.001). IGF-II partially reversed the IGFBP-6-induced decrease in growth and increase in apoptosis. Rh30 cells were stably transfected with an IGFBP-6 cDNA and subcutaneous xenografts established in BALB/c nude mice. After 18 days, sizes of 2 independent clones of IGFBP-6-overexpressing Rh30 cells were reduced to 12% and 26% of vector control-transfected tumors (p = 0.0006 and 0.002, respectively). IGFBP-6 therefore inhibits proliferation and promotes apoptosis of rhabdomyosarcoma in vitro and dramatically inhibits xenograft growth in vivo, at least in part by inhibiting IGF-II. Low expression of IGFBP-6 may therefore contribute to rhabdomyosarcoma growth and metastasis.


Subject(s)
Insulin-Like Growth Factor Binding Protein 6/physiology , Rhabdomyosarcoma/prevention & control , Animals , Apoptosis , Cell Division , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Rhabdomyosarcoma/pathology , Tumor Cells, Cultured
13.
Pediatr Hematol Oncol ; 17(7): 541-50, 2000.
Article in English | MEDLINE | ID: mdl-11033729

ABSTRACT

Data from a case-control study in Lower Saxony, Germany, were used to assess whether the risk for childhood cancer may be reduced by bacille Calmette-Guérin (BCG) vaccination in the neonatal period. There were 420 newly diagnosed childhood cancer cases from the German cancer registry and 613 controls eligible for this study. A mailed questionnaire was completed during a telephone interview with parents. Details on the perinatal history were abstracted from the birth charts by nurses blinded to the children's case-control status. Complete information was available for 259 cases and for 323 controls. A total of 85% of the controls had been BCG vaccinated in the newborn period. The adjusted odds ratios for BCG vaccination were 0.90 (95% confidence interval; 0.51-1.61) for leukemia and 0.61 (95% confidence interval; 0.25-1.50) for other cancers. Based on these data the probability of a 50% or more reduction of more reduction of the cancer risk by BCG vaccination in the newborn period is small. The statistical power of this study, however, was not high enough to rule out a smaller, still relevant reduction in cancer risk.


Subject(s)
BCG Vaccine/therapeutic use , Leukemia/prevention & control , Acute Disease , Adolescent , Brain Neoplasms/epidemiology , Brain Neoplasms/prevention & control , Case-Control Studies , Child , Child, Preschool , Germany/epidemiology , Humans , Infant , Infant, Newborn , Leukemia/epidemiology , Neuroblastoma/epidemiology , Neuroblastoma/prevention & control , Precursor Cell Lymphoblastic Leukemia-Lymphoma/epidemiology , Precursor Cell Lymphoblastic Leukemia-Lymphoma/prevention & control , Rhabdomyosarcoma/epidemiology , Rhabdomyosarcoma/prevention & control , Risk Factors , Wilms Tumor/epidemiology , Wilms Tumor/prevention & control
14.
Clin Cancer Res ; 6(3): 998-1007, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10741727

ABSTRACT

The antitumor activity of the methylating agent temozolomide has been evaluated against a panel of 17 xenografts derived from pediatric solid tumors. Temozolomide was administered p.o. daily for five consecutive days at a dose level of 66 mg/kg. Courses of treatment were repeated every 21 days for three cycles. Tumor lines were classified as having high, intermediate, or low sensitivity, determined by complete responses, partial responses, or stable disease, respectively. Overall, temozolomide induced complete responses in five lines and partial responses in three additional tumor lines, giving objective regressions in 47% of xenograft lines. Analysis of temozolomide plasma systemic exposure indicated that this dose level was relevant to exposure achieved in patients. Tumors were analyzed by immunoblotting for levels of O6-methylguanine-DNA methyltransferase (MGMT) and two mismatch repair proteins, MLH-1 and MSH-2. Tumors classified as having high or intermediate sensitivity had low or undetectable MGMT and expressed detectable MLH-1 and MSH-2 proteins. Tumors classified as having low sensitivity had either (a) high MGMT or (b) low or undetectable MGMT but were deficient in MLH-1. The relationship between p53 and response to temozolomide was also examined. In vitro temozolomide did not induce p21cip1 in p53-competent NB-1643 neuroblastoma cells. Suppression of p53 function in NB1643 clones through stable expression of a trans dominant negative p53 (NB1643p53TDN) did not confer temozolomide resistance. Similarly, tumor sensitivity to temozolomide did not segregate with p53 genotype or p53 functional status. These results indicate that MGMT is the primary mechanism for temozolomide resistance, but in the absence of MGMT, proficient mismatch repair determines sensitivity to this agent.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , DNA-Binding Proteins , Dacarbazine/analogs & derivatives , Neoplasms, Experimental/prevention & control , Adaptor Proteins, Signal Transducing , Animals , Antineoplastic Agents, Alkylating/pharmacokinetics , Base Pair Mismatch , Brain Neoplasms/pathology , Brain Neoplasms/prevention & control , Carrier Proteins , Cell Division/drug effects , Child , DNA Repair , Dacarbazine/blood , Dacarbazine/pharmacokinetics , Dacarbazine/therapeutic use , Female , Humans , Mice , Mice, Inbred CBA , MutL Protein Homolog 1 , MutS Homolog 2 Protein , Neoplasm Proteins/metabolism , Neoplasm Transplantation , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Neuroblastoma/pathology , Neuroblastoma/prevention & control , Nuclear Proteins , O(6)-Methylguanine-DNA Methyltransferase/metabolism , Phenotype , Proto-Oncogene Proteins/metabolism , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/prevention & control , Temozolomide , Transplantation, Heterologous , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/physiology
15.
Vopr Onkol ; 45(6): 650-4, 1999.
Article in Russian | MEDLINE | ID: mdl-10703515

ABSTRACT

Tumor growth and proliferative activity of tumor cells were suppressed and the number of pulmonary metastases in C57B16 mice decreased 3.3-fold following seven injections of cycloferon (100 mg/kg body) to induce interferon production. Injections were carried out 1-16 days after subcutaneous transplantation of Lewis lung carcinoma. After mice were immunized with ovine red blood cells, cycloferon administration raised thymus-dependent humoral immune response. After eight injections of cycloferon (50 mg/kg body) into rats, from day of intravenous transplantation of rhabdomyosarcoma RA-23 until day 20, no significant effect on metastasizing into the lung was recorded. However, single injection of cyclophosphamide 50 mg/kg inhibited metastasis formation. The highest suppressor effect was registered with combination cycloferon-cyclophophamide treatment: mean weight of metastasis decreased by half, as compared with treatment with cyclophosphamide alone. Both drugs caused karyotypical abnormalities to occur in metastatic cells. Tumor growth and spreading suppression after cycloferon should be attributed to cytotoxic antitumor action, cell proliferation inhibition and immunomodulating effect.


Subject(s)
Acridines/therapeutic use , Antineoplastic Agents/therapeutic use , Carcinoma, Lewis Lung/drug therapy , Carcinoma, Lewis Lung/prevention & control , Interferon Inducers/therapeutic use , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/prevention & control , Acridines/pharmacology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents, Alkylating/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/secondary , Cell Division/drug effects , Cyclophosphamide/therapeutic use , Injections , Interferon Inducers/pharmacology , Mice , Mice, Inbred C57BL , Rats , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/secondary
16.
Exp Lung Res ; 24(4): 629-39, 1998.
Article in English | MEDLINE | ID: mdl-9659588

ABSTRACT

Both green and black tea have been shown to inhibit lung tumorigenesis in laboratory animal experiments. Green tea inhibited N-nitrosodiethylamine-induced lung tumor incidence and multiplicity in female A/J mice when tea was given either during the carcinogen treatment period or during the post-carcinogen treatment period. In a separate tumorigenesis model, both decaffeinated black tea and decaffeinated green tea inhibited 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-induced lung tumor formation. Studies in which tea was administered during different time periods in relation to the NNK suggest that tea can inhibit lung tumorigenesis at both the initiation and promotion stages. The antiproliferative effects of tea may be responsible for these anti-carcinogenic actions. Black tea polyphenol preparations decreased NNK-induced hyperproliferation. Black tea also inhibited the progression of pulmonary adenomas to adenocarcinomas and the formation of spontaneous lung tumors in A/J mice. Growth inhibition by various tea polyphenols has been demonstrated in human lung H661 and H1299 cells. Although inhibition of cell growth and signal transduction pathways by tea components have been demonstrated, the concentrations required to produce the effect are higher than achievable in tissues in vivo. More research is necessary to translate these laboratory results to applications in human chemoprevention.


Subject(s)
Adenoma/prevention & control , Anticarcinogenic Agents/pharmacology , Cell Division/drug effects , Flavonoids , Lung Neoplasms/prevention & control , Phenols/pharmacology , Polymers/pharmacology , Tea , Adenoma/chemically induced , Adenoma/pathology , Animals , Catechin/analysis , Diethylnitrosamine/toxicity , Disease Progression , Female , Lung Neoplasms/chemically induced , Lung Neoplasms/pathology , Mice , Nitrosamines/toxicity , Rhabdomyosarcoma/prevention & control , Tumor Cells, Cultured
17.
Carcinogenesis ; 19(3): 501-7, 1998 Mar.
Article in English | MEDLINE | ID: mdl-9525286

ABSTRACT

We investigated the effects of black tea (BT) and green tea (GT) infusion on the spontaneous formation of lung tumors and rhabdomyosarcomas in A/J mice. Female A/J mice, 6 weeks of age, were allocated into five groups (50 per group) and were given the following as the sole source of drinking fluid: (i) deionized water (control group), (ii) 0.5% BT, (iii) 1% BT, (iv) 2% BT and (v) 1% GT. After 60 weeks, the mice were killed by decapitation. Lung tumor incidence, multiplicity and volume were significantly lower in the 2% BT group as compared with the controls (27 versus 52%, 0.33 versus 0.72 tumors/mouse and 4.27 versus 38.3 mm3, respectively). The 1% GT group had significantly lower lung tumor multiplicity (0.41/mouse), while the 1% BT group had significantly decreased tumor volume (7.17 mm3). Rhabdomyosarcomas were found in 34% of the mice in the control group, and both the 1 and 2% BT groups had significantly lower incidences at 13 and 14%, respectively. The mice in the 2% BT group weighed 16% less than those in the control group, although they consumed more food than the control group. The other tea-consuming groups also weighed less than the control group (7.8-11%) while consuming more food and fluid. In a separate experiment, similar carcinogenesis inhibition was also observed in female A/J mice that were given 0.6% and then 0.3% instant black tea for 52 weeks. These results demonstrate the inhibitory activity of BT against the spontaneous formation of lung tumors and rhabdomyosarcomas in mice.


Subject(s)
Lung Neoplasms/prevention & control , Rhabdomyosarcoma/prevention & control , Tea , Animals , Body Weight , Drinking Behavior , Feeding Behavior , Female , Lung Neoplasms/pathology , Mice , Rhabdomyosarcoma/pathology
18.
Br J Cancer ; 70(5): 1000-3, 1994 Nov.
Article in English | MEDLINE | ID: mdl-7947075

ABSTRACT

Eleven survivors of pelvic rhabdomyosarcoma underwent bladder function studies and upper urinary tract evaluation at a mean of 6.6 years after completion of therapy, which included a conservative, bladder-sparing surgical policy. Primary tumour sites were: bladder base/prostate, 6; bladder dome, 1; vagina, 2; and pelvic side wall, 2. Seven children (five bladder base/prostate, one vagina and one pelvic side wall tumours) had received irradiation to the pelvis with external beam alone, brachytherapy or both. All seven of these patients had markedly reduced functional bladder capacity (11-48% of mean expected value for age) and abnormal voiding patterns, though bladder compliance was not reduced and bladder emptying was almost complete in five cases. Four of these children also had upper tract dilatation and two required reconstructive bladder surgery because of severe bilateral hydronephrosis. By contrast, each of four children treated without radiotherapy had a normal functional bladder capacity and a normal voiding pattern. all survivors of pelvic rhabdomyosarcoma, especially those who have received radiotherapy, should be carefully monitored for dysfunction of both lower and upper urinary tracts. The frequency-volume voiding chart is a sensitive and easily accomplished method of assessing bladder function in these patients.


Subject(s)
Kidney/physiology , Pelvic Neoplasms/prevention & control , Rhabdomyosarcoma/prevention & control , Urinary Bladder/physiology , Child , Child, Preschool , Combined Modality Therapy , Female , Humans , Infant , Kidney/radiation effects , Male , Pelvic Neoplasms/radiotherapy , Pelvic Neoplasms/surgery , Rhabdomyosarcoma/radiotherapy , Rhabdomyosarcoma/surgery , Time Factors , Urinary Bladder/radiation effects , Urinary Tract/radiation effects , Urinary Tract Physiological Phenomena , Urodynamics
19.
J Clin Oncol ; 8(11): 1854-7, 1990 Nov.
Article in English | MEDLINE | ID: mdl-2230872

ABSTRACT

A series of 15 consecutive children with head and neck nonorbital rhabdomyosarcoma (RMSA) with meningeal extension were prospectively treated with chemotherapy consisting of Adriamycin (doxorubicin; Adria Laboratory, Columbus, OH) (ADM), vincristine (VCR), cyclophosphamide (CPM), and dactinomycin (DACT) followed by radiotherapy (60 Gy) to the primary tumor volume, along with intrathecal methotrexate (IT MTX). Thirteen of 15 responded to preradiation chemotherapy. Four of 13 relapsed. Relapse occurred at the level of the primary tumor in three of four. The 3-year progression-free survival (PFS) was 59%, similar to that achieved in a previous series treated with a comparable therapeutic approach that also included whole-brain radiotherapy as a prophylaxis of possible occult meningeal seeding. It is concluded that CNS prophylaxis with radiotherapy is questionable in the management of childhood RMSA with meningeal extension.


Subject(s)
Central Nervous System Diseases/prevention & control , Head and Neck Neoplasms/radiotherapy , Meningeal Neoplasms/radiotherapy , Nervous System Neoplasms/prevention & control , Rhabdomyosarcoma/radiotherapy , Adolescent , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Child , Child, Preschool , Combined Modality Therapy , Female , Follow-Up Studies , Head and Neck Neoplasms/drug therapy , Humans , Male , Meningeal Neoplasms/drug therapy , Meningeal Neoplasms/prevention & control , Meningeal Neoplasms/secondary , Prospective Studies , Rhabdomyosarcoma/drug therapy , Rhabdomyosarcoma/prevention & control , Rhabdomyosarcoma/secondary , Survival Rate
20.
Cancer Immunol Immunother ; 25(2): 111-8, 1987.
Article in English | MEDLINE | ID: mdl-3117366

ABSTRACT

Nocardia delipidated cell mitogen (NDCM), a particulate fraction prepared from Nocardia opaca, injected i.p. in an oil/water emulsion to F6 rhabdomyosarcoma-bearing rats, inhibited the development of pulmonary metastases; 6 out of 10 rats were protected. Repeated i.p. administration of emulsified NDCM and of two other compounds, a Nocardia water soluble mitogen (NWSM a hydrosoluble fraction) and purified cell walls (CW, an insoluble macromolecular fraction) in Lewis lung carcinoma (LLC)-bearing mice resulted in a significant reduction of lung metastases. The efficiency of these fractions was enhanced by association with monokines. A combination regimen of NDCM, NWSM, and CW (100 micrograms/0.1 ml) and monokines (0.1 ml), injected i.p. in LLC-bearing mice, yielded a greater antimetastatic effect than either therapy alone. Peritoneal macrophages from mice which had been injected i.p. with NWSM or CW, when triggered either by TPA (tetradecanoyl phorbol acetate) or by zymosan, released large quantities of hydrogen peroxide and had a high rate of glucose consumption. These macrophages were activated as judged by their cytostatic activity against syngeneic P815 mastocytoma growth; they expressed biochemical markers which have been reported to characterize the activated state. Incubation of thioglycollate-elicited peritoneal macrophages with NWSM, and monokines for 72 h resulted in a cytotoxic activity against labeled LLC cells; addition of macrophage activating factor significantly increased the cytotoxic capacity of these macrophages. In view of this we postulate that the antimetastatic effect of soluble and insoluble N. opaca fractions and monokines might be mediated by activated peritoneal macrophages.


Subject(s)
Adjuvants, Immunologic/therapeutic use , Carcinoma/secondary , Cell Wall/immunology , Lung Neoplasms/secondary , Macrophage Activation/drug effects , Nocardia/immunology , Rhabdomyosarcoma/secondary , Adjuvants, Immunologic/immunology , Adjuvants, Immunologic/isolation & purification , Animals , Carcinoma/immunology , Carcinoma/pathology , Carcinoma/prevention & control , Drug Synergism , Female , Lung Neoplasms/immunology , Lung Neoplasms/prevention & control , Macrophages/drug effects , Macrophages/pathology , Male , Mast-Cell Sarcoma , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Monokines , Nocardia/analysis , Peritoneal Cavity/pathology , Proteins/pharmacology , Rats , Rats, Inbred Strains , Rhabdomyosarcoma/immunology , Rhabdomyosarcoma/pathology , Rhabdomyosarcoma/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL
...