Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
1.
ESMO Open ; 7(6): 100635, 2022 12.
Article in English | MEDLINE | ID: mdl-36410086

ABSTRACT

BACKGROUND: Immune checkpoint inhibitor (ICI) therapy is an emerging option for advanced endometrial cancer (EC). Mismatch repair (MMR) status is widely regarded as a biomarker predictive of response to ICIs. The predictive value of MMR based on small, single-arm trials, however, is conflicting. In this meta-analysis, we aimed to assess the activity of single-agent ICI in advanced EC, and compared the magnitude of treatment benefit in MMR deficient (dMMR) and MMR proficient (pMMR) EC. METHODS: We carried out an electronic search to identify prospective trials of single-agent ICI in advanced EC. Data on objective response rate (ORR) and progression-free survival (PFS) were extracted and pooled. ORR was estimated using the inverse variance method and subgroup difference by MMR status was examined. PFS difference according to MMR status was summarized using the Kaplan-Meier approach. RESULTS: From eight trials with 492 women, the pooled ORR was 19% [95% confidence interval (CI) 16% to 22%]. ORR was significantly greater in dMMR (n = 281) than pMMR EC (n = 211) (dMMR: 46%, pMMR: 8%; risk ratio 5.74, 95% CI 3.58-9.21; interaction P < 0.001). Complete response was 11% and 0.05% and median PFS was 8.3 and 2.1 months in dMMR and pMMR EC, respectively (hazard ratio PFS 0.58, 95% CI 0.38-0.89; P = 0.01). The 12-month PFS rates were 42.0% and 20.7%, respectively. CONCLUSION: Single-agent ICI is associated with a 5.74 times greater objective response and 42% reduction in risk of disease progression or death in dMMR compared with pMMR EC. MMR status should be determined prospectively and be used as a stratification factor in future trials of advanced EC. Further translational analysis is urgently required to identify the cause of dMMR and allow subclassification of EC into different dMMR molecular subtypes.


Subject(s)
Endometrial Neoplasms , Programmed Cell Death 1 Receptor , Humans , Female , Prospective Studies , B7-H1 Antigen , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/genetics
2.
Ann Oncol ; 31(12): 1606-1622, 2020 12.
Article in English | MEDLINE | ID: mdl-33004253

ABSTRACT

BACKGROUND: Homologous recombination repair deficiency (HRD) is a frequent feature of high-grade serous ovarian, fallopian tube and peritoneal carcinoma (HGSC) and is associated with sensitivity to PARP inhibitor (PARPi) therapy. HRD testing provides an opportunity to optimise PARPi use in HGSC but methodologies are diverse and clinical application remains controversial. MATERIALS AND METHODS: To define best practice for HRD testing in HGSC the ESMO Translational Research and Precision Medicine Working Group launched a collaborative project that incorporated a systematic review approach. The main aims were to (i) define the term 'HRD test'; (ii) provide an overview of the biological rationale and the level of evidence supporting currently available HRD tests; (iii) provide recommendations on the clinical utility of HRD tests in clinical management of HGSC. RESULTS: A broad range of repair genes, genomic scars, mutational signatures and functional assays are associated with a history of HRD. Currently, the clinical validity of HRD tests in ovarian cancer is best assessed, not in terms of biological HRD status per se, but in terms of PARPi benefit. Clinical trials evidence supports the use of BRCA mutation testing and two commercially available assays that also incorporate genomic instability for identifying subgroups of HGSCs that derive different magnitudes of benefit from PARPi therapy, albeit with some variation by clinical scenario. These tests can be used to inform treatment selection and scheduling but their use is limited by a failure to consistently identify a subgroup of patients who derive no benefit from PARPis in most studies. Existing tests lack negative predictive value and inadequately address the complex and dynamic nature of the HRD phenotype. CONCLUSIONS: Currently available HRD tests are useful for predicting likely magnitude of benefit from PARPis but better biomarkers are urgently needed to better identify current homologous recombination proficiency status and stratify HGSC management.


Subject(s)
Ovarian Neoplasms , Poly(ADP-ribose) Polymerase Inhibitors , Biomarkers , Carcinoma, Ovarian Epithelial , Female , Homologous Recombination , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
3.
Ginebra; European Society for Medical Oncology; 2020; Sept. 28, 2020. 17 p.
Monography in English | BIGG - GRADE guidelines | ID: biblio-1127783

ABSTRACT

The European Society for Medical Oncology (ESMO) Translational Research and Precision Medicine Working Group identified that there is currently uncertainty within the oncology community surrounding the different methods for HRD testing in HGSC. To address this, a collaborative project was launched with a number of clinicians and scientists with expertise in the fields of PARPi clinical trials, cancer genomics and DNA repair. The group defined three main aims for the project: (i) Define the term 'HRD test' and recommend how an HRD test's clinical validity is currently best assessed in the context of HGSC, (ii) provide an overview of the biological rationale and the level of evidence supporting currently available HRD tests, and (iii) provide recommendations on the clinical utility of HRD tests in clinical management of HGSC.


Subject(s)
Humans , Female , Biomarkers , Genital Neoplasms, Female/diagnosis , Germ-Line Mutation/genetics , Homologous Recombination/genetics , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use
4.
Gynecol Oncol ; 158(2): 431-439, 2020 08.
Article in English | MEDLINE | ID: mdl-32451123

ABSTRACT

BACKGROUND: BRCA1/2 mutation status has increasing relevance for ovarian cancer treatments, making traditional coordination of genetic testing by genetic services unsustainable. Consequently alternative models of genetic testing have been developed to improve testing at the initial diagnosis for all eligible women. METHODS: A training module to enable mainstreamed genetic testing by oncology healthcare professionals was developed by genetic health professionals. Oncology healthcare professionals completed questionnaires before and 12 months post-training to assess perceived skills, competence and barriers to their coordinating genetic testing for women with high-grade non-mucinous epithelial ovarian cancer. Genetic health professionals were surveyed 12 months post-training to assess perceived barriers to implementation of mainstreaming. RESULTS: 185 oncology healthcare professionals were trained in 42 workshops at 35 Australasian hospitals. Of the 273 tests ordered by oncology healthcare professionals post-training, 241 (93.1%) met national testing guidelines. The number of tests ordered by genetic health professionals reduced significantly (z = 45.0, p = 0.008). Oncology healthcare professionals' perceived barriers to mainstreamed testing decreased from baseline to follow-up (t = 2.39, p = 0.023), particularly perceived skills, knowledge and attitudes. However, only 58% reported either 'always' or 'nearly always' having ordered BRCA testing for eligible patients at 12 months, suggesting oncology healthcare professionals' perceived barriers were not systematically addressed through training. CONCLUSIONS: Oncology healthcare professionals have demonstrated a willingness to be involved in the provision of genetic testing in a mainstreaming model. If oncology services are to hold responsibility for coordinating genetic testing, their readiness will require understanding of barriers not addressed by training alone to inform future intervention design.


Subject(s)
Carcinoma, Ovarian Epithelial/genetics , Genetic Testing/methods , Genetics/education , Medical Oncology/education , Ovarian Neoplasms/genetics , Adolescent , Adult , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Education, Medical, Continuing , Female , Health Personnel/education , Humans , Male , Middle Aged , Young Adult
5.
Nat Commun ; 8(1): 620, 2017 09 20.
Article in English | MEDLINE | ID: mdl-28931816

ABSTRACT

CD103+CD11b+ dendritic cells (DCs) are unique to the intestine, but the factors governing their differentiation are unclear. Here we show that transforming growth factor receptor 1 (TGFßR1) has an indispensable, cell intrinsic role in the development of these cells. Deletion of Tgfbr1 results in markedly fewer intestinal CD103+CD11b+ DCs and a reciprocal increase in the CD103-CD11b+ dendritic cell subset. Transcriptional profiling identifies markers that define the CD103+CD11b+ DC lineage, including CD101, TREM1 and Siglec-F, and shows that the absence of CD103+CD11b+ DCs in CD11c-Cre.Tgfbr1 fl/fl mice reflects defective differentiation from CD103-CD11b+ intermediaries, rather than an isolated loss of CD103 expression. The defect in CD103+CD11b+ DCs is accompanied by reduced generation of antigen-specific, inducible FoxP3+ regulatory T cells in vitro and in vivo, and by reduced numbers of endogenous Th17 cells in the intestinal mucosa. Thus, TGFßR1-mediated signalling may explain the tissue-specific development of these unique DCs.Developmental cues for the different dendritic cell (DC) subsets in the intestine are yet to be defined. Here the authors show that TGFßR1 signalling is needed for development of CD103+CD11b+ intestinal DCs from CD103-CD11b+ cells and that they contribute to the generation of Th17 and regulatory T cells.


Subject(s)
Cell Differentiation/genetics , Dendritic Cells/immunology , Intestinal Mucosa/immunology , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Animals , Antigens, CD/immunology , CD11b Antigen/immunology , Cell Lineage , Colitis/immunology , Dendritic Cells/cytology , Immunity, Mucosal , Integrin alpha Chains/immunology , Intestinal Mucosa/cytology , Intestines/cytology , Intestines/immunology , Lymphopoiesis/genetics , Mice , Mice, Knockout , Receptor, Transforming Growth Factor-beta Type I , T-Lymphocytes, Regulatory/cytology , Th17 Cells/cytology
6.
Mucosal Immunol ; 10(4): 831-844, 2017 07.
Article in English | MEDLINE | ID: mdl-28198365

ABSTRACT

Our understanding of conventional dendritic cell (cDC) development and the functional specializations of distinct subsets in the peripheral tissues has increased greatly in recent years. Here, we review cDC development from the distinct progenitors in the bone marrow through to the distinct cDC subsets found in barrier tissues, providing an overview of the different subsets described in each location. In addition, we detail the transcription factors and local signals that have been proposed to control this developmental process. Importantly, despite these significant advances, numerous questions remain to be answered regarding cDC development. For example, it remains unclear whether the different subsets described, such as the CD103+CD11b+ and CD103-CD11b+ cDCs in the intestines, truly represent different populations or rather distinct developmental or activation stages. Furthermore, whether distinct progenitors exist for these cDC subsets remains to be determined. Thus in the last part of this review we discuss what we believe will be the main questions facing the field for the coming years.


Subject(s)
Bone Marrow Cells/physiology , Dendritic Cells/physiology , Animals , Antigens, CD/metabolism , CD11b Antigen/metabolism , Cell Differentiation , Humans , Integrin alpha Chains/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
7.
Cell Death Differ ; 23(2): 303-12, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26184912

ABSTRACT

Genetic recombination during B-cell development regularly results in the generation of autoreactive, potentially pathogenic B-cell receptors (BCRs). Consequently, multiple mechanisms link inappropriate BCR specificity to clonal deletion. Similar pathways remain in malignant B cells, offering the potential for targeting BCR signaling. Recently, small molecule inhibitors have realized this potential and, therefore, a deeper understanding of BCR-induced signaling networks in malignant cells is vital. The BH3-only protein Bim has a key role in BCR-induced apoptosis, but it has long been proposed that additional BH3-only proteins also contribute, although conclusive proof has been lacking. Here, we comprehensively characterized the mechanism of BCR-induced apoptosis in Eµ-Myc murine lymphoma cells. We demonstrate the upregulation of Bim, Bik, and Noxa during BCR signaling in vitro and that intrinsic apoptosis has a prominent role in anti-BCR antibody therapy in vivo. Furthermore, lymphomas deficient in these individual BH3-only proteins display significant protection from BCR-induced cell death, whereas combined loss of Noxa and Bim offers enhanced protection in comparison with loss of Bim alone. Some but not all of these effects were reversed upon inhibition of Syk or MEK. These observations indicate that BCR signaling elicits maximal cell death through upregulation of multiple BH3-only proteins; namely Bim, Bik, and Noxa.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Apoptosis Regulatory Proteins/metabolism , Lymphoma, B-Cell/metabolism , Membrane Proteins/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcr/metabolism , Proto-Oncogene Proteins/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Apoptosis , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Cell Line, Tumor , Lymphoma, B-Cell/pathology , Membrane Proteins/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Mitochondrial Proteins/genetics , Neoplasm Transplantation , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Signal Transduction
8.
Biochim Biophys Acta ; 1855(2): 223-34, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25783201

ABSTRACT

Effective systemic treatment of cancer relies on the delivery of agents with optimal therapeutic potential. The molecular age of medicine has provided genomic tools that can identify a large number of potential therapeutic targets in individual patients, heralding the promise of personalized treatment. However, determining which potential targets actually drive tumor growth and should be prioritized for therapy is challenging. Indeed, reliable molecular matches of target and therapeutic agent have been stringently validated in the clinic for only a small number of targets. Patient-derived xenografts (PDXs) are tumor models developed in immunocompromised mice using tumor procured directly from the patient. As patient surrogates, PDX models represent a powerful tool for addressing individualized therapy. Challenges include humanizing the immune system of PDX models and ensuring high quality molecular annotation, in order to maximize insights for the clinic. Importantly, PDX can be sampled repeatedly and in parallel, to reveal clonal evolution, which may predict mechanisms of drug resistance and inform therapeutic strategy design.


Subject(s)
Drug Resistance, Neoplasm/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Xenograft Model Antitumor Assays , Animals , Disease Models, Animal , Drug Resistance, Neoplasm/immunology , Humans , Mice , Neoplasms/pathology , Precision Medicine
9.
Mucosal Immunol ; 8(1): 38-48, 2015 Jan.
Article in English | MEDLINE | ID: mdl-24850430

ABSTRACT

Cross-presentation of cellular antigens is crucial for priming CD8(+) T cells, and generating immunity to intracellular pathogens--particularly viruses. It is unclear which intestinal phagocytes perform this function in vivo. To address this, we examined dendritic cells (DCs) from the intestinal lymph of IFABP-tOVA 232-4 mice, which express ovalbumin in small intestinal epithelial cells (IECs). Among lymph DCs (LDCs) only CD103(+) CD11b(-) CD8α(+) DCs cross-present IEC-derived ovalbumin to CD8(+) OT-I T cells. Similarly, in the mesenteric lymph nodes (MLNs), cross-presentation of IEC-ovalbumin was limited to the CD11c(+) MHCII(hi) CD8α(+) migratory DCs, but absent from all other subsets, including the resident CD8α(hi) DCs. Crucially, delivery of purified CD8α(+) LDCs, but not other LDC subsets, into the MLN subcapsular lymphatic sinus induced proliferation of ovalbumin-specific, gut-tropic CD8(+) T cells in vivo. Finally, in 232-4 mice treated with R848, CD8α(+) LDCs were uniquely able to cross-prime interferon γ-producing CD8(+) T cells and drive their migration to the intestine. Our results clearly demonstrate that migrating CD8α(+) intestinal DCs are indispensable for cross-presentation of cellular antigens and, in conditions of inflammation, for the initial differentiation of effector CD8(+) T cells. They may therefore represent an important target for the development of antiviral vaccinations.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Ovalbumin/metabolism , Animals , Antigens/immunology , CD8 Antigens/metabolism , Cell Movement/drug effects , Cell Movement/genetics , Cell Proliferation/drug effects , Cells, Cultured , Cross-Priming/drug effects , Cross-Priming/genetics , Imidazoles/administration & dosage , Imidazoles/pharmacology , Interferon-gamma/metabolism , Intestinal Mucosa/immunology , Lymph/immunology , Lymphocyte Activation/drug effects , Lymphocyte Activation/genetics , Membrane Glycoproteins/agonists , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Ovalbumin/genetics , Ovalbumin/immunology , Toll-Like Receptor 7/agonists
10.
Mucosal Immunol ; 8(2): 327-39, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25138666

ABSTRACT

The identification of intestinal macrophages (mφs) and dendritic cells (DCs) is a matter of intense debate. Although CD103(+) mononuclear phagocytes (MPs) appear to be genuine DCs, the nature and origins of CD103(-) MPs remain controversial. We show here that intestinal CD103(-)CD11b(+) MPs can be separated clearly into DCs and mφs based on phenotype, gene profile, and kinetics. CD64(-)CD103(-)CD11b(+) MPs are classical DCs, being derived from Flt3 ligand-dependent, DC-committed precursors, not Ly6C(hi) monocytes. Surprisingly, a significant proportion of these CD103(-)CD11b(+) DCs express CCR2 and there is a selective decrease in CD103(-)CD11b(+) DCs in mice lacking this chemokine receptor. CCR2(+)CD103(-) DCs are present in both the murine and human intestine, drive interleukin (IL)-17a production by T cells in vitro, and show constitutive expression of IL-12/IL-23p40. These data highlight the heterogeneity of intestinal DCs and reveal a bona fide population of CCR2(+) DCs that is involved in priming mucosal T helper type 17 (Th17) responses.


Subject(s)
Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/immunology , Intestinal Mucosa/cytology , Intestinal Mucosa/immunology , T-Lymphocyte Subsets/metabolism , Th17 Cells/metabolism , Animals , Antigens, CD/metabolism , Dendritic Cells/metabolism , Humans , Immunophenotyping , Integrin alpha Chains/metabolism , Interferon Regulatory Factors/metabolism , Interleukin-12/metabolism , Interleukin-17/biosynthesis , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Monocytes/immunology , Monocytes/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Phenotype , Receptors, CCR2/metabolism , T-Lymphocyte Subsets/immunology , Th17 Cells/immunology
11.
Mucosal Immunol ; 7(1): 101-13, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23653115

ABSTRACT

Interleukin-22 (IL-22) is mainly produced at barrier surfaces by T cells and innate lymphoid cells and is crucial to maintain epithelial integrity. However, dysregulated IL-22 action leads to deleterious inflammation and is involved in diseases such as psoriasis, intestinal inflammation, and cancer. IL-22 binding protein (IL-22BP) is a soluble inhibitory IL-22 receptor and may represent a crucial regulator of IL-22. We show both in rats and mice that, in the steady state, the main source of IL-22BP is constituted by a subset of conventional dendritic cells (DCs) in lymphoid and non-lymphoid tissues. In mouse intestine, IL-22BP was specifically expressed in lamina propria CD103(+)CD11b(+) DC. In humans, IL-22BP was expressed in immature monocyte-derived DC and strongly induced by retinoic acid but dramatically reduced upon maturation. Our data suggest that a subset of immature DCs may actively participate in the regulation of IL-22 activity in the gut by producing high levels of IL-22BP.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Gene Expression Regulation/drug effects , Receptors, Interleukin/genetics , Tretinoin/pharmacology , Animals , CD4 Antigens/metabolism , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/drug effects , Gene Expression Profiling , Humans , Intestinal Mucosa/metabolism , Intestines/immunology , Male , Monocytes/cytology , Monocytes/immunology , Monocytes/metabolism , Organ Specificity/genetics , Protein Isoforms , Rats , Receptors, Interleukin/metabolism , Spleen/cytology , Spleen/immunology , Spleen/metabolism
13.
Mucosal Immunol ; 6(3): 498-510, 2013 May.
Article in English | MEDLINE | ID: mdl-22990622

ABSTRACT

Macrophages (mφ) are essential for intestinal homeostasis and the pathology of inflammatory bowel disease (IBD), but it is unclear whether discrete mφ populations carry out these distinct functions or if resident mφ change during inflammation. We show here that most resident mφ in resting mouse colon express very high levels of CX3CR1, are avidly phagocytic and MHCII(hi), but are resistant to Toll-like receptor (TLR) stimulation, produce interleukin 10 constitutively, and express CD163 and CD206. A smaller population of CX3CR1(int) cells is present in resting colon and it expands during experimental colitis. Ly6C(hi)CCR2(+) monocytes can give rise to all mφ subsets in both healthy and inflamed colon and we show that the CX3CR1(int) pool represents a continuum in which newly arrived, recently divided monocytes develop into resident CX3CR1(hi) mφ. This process is arrested during experimental colitis, resulting in the accumulation of TLR-responsive pro-inflammatory mφ. Phenotypic analysis of human intestinal mφ indicates that analogous processes occur in the normal and Crohn's disease ileum. These studies show for the first time that resident and inflammatory mφ in the intestine represent alternative differentiation outcomes of the same precursor and targeting these events could offer routes for therapeutic intervention in IBD.


Subject(s)
Colitis/immunology , Colon/immunology , Inflammatory Bowel Diseases/immunology , Macrophages/immunology , Monocytes/immunology , Animals , Antigens, Ly/metabolism , CX3C Chemokine Receptor 1 , Cell Differentiation , Cell Movement , Cell Proliferation , Cells, Cultured , Colitis/chemically induced , Histocompatibility Antigens Class II/metabolism , Humans , Inflammation/pathology , Interleukin-10/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, Chemokine/genetics , Receptors, Chemokine/metabolism
14.
Mucosal Immunol ; 6(1): 104-13, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22718260

ABSTRACT

Intestinal dendritic cells (DCs) continuously migrate through lymphatics to mesenteric lymph nodes where they initiate immunity or tolerance. Recent research has focused on populations of intestinal DCs expressing CD103. Here we demonstrate, for the first time, the presence of two distinct CD103(-) DC subsets in intestinal lymph. Similar to CD103(+) DCs, these intestine-derived CD103(-) DCs are responsive to Flt3 and they efficiently prime and confer a gut-homing phenotype to naive T cells. However, uniquely among intestinal DCs, CD103(-) CD11b(+) CX(3)CR1(int) lymph DCs induce the differentiation of both interferon-γ and interleukin-17-producing effector T cells, even in the absence of overt stimulation. Priming by CD103(-) CD11b(+) DCs represents a novel mechanism for the rapid generation of effector T-cell responses in the gut. Therefore, these cells may prove to be valuable targets for the treatment of intestinal inflammation or in the development of effective oral vaccines.


Subject(s)
Antigens, CD/metabolism , Cell Movement/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Integrin alpha Chains/metabolism , Intestinal Mucosa/immunology , Lymph/immunology , T-Lymphocyte Subsets/immunology , Aldehyde Dehydrogenase/metabolism , Animals , CD11b Antigen/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Immunophenotyping , Interferon-gamma/biosynthesis , Interleukin-17/biosynthesis , Intestinal Mucosa/metabolism , Lymph/cytology , Lymphocyte Activation , Male , Membrane Proteins/metabolism , Mice , Receptors, CCR/metabolism
15.
Cell Death Dis ; 3: e306, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22573037

ABSTRACT

The pro-apoptotic BH3-only protein, BIK, is widely expressed and although many critical functions in developmental or stress-induced death have been ascribed to this protein, mice lacking Bik display no overt abnormalities. It has been postulated that Bik can serve as a tumour suppressor, on the basis that its deficiency and loss of apoptotic function have been reported in many human cancers, including lymphoid malignancies. Evasion of apoptosis is a major factor contributing to c-Myc-induced tumour development, but despite this, we found that Bik deficiency did not accelerate Eµ-Myc-induced lymphomagenesis. Co-operation between BIK and NOXA, another BH3-only protein, has been previously described, and was attributed to their complementary binding specificities to distinct subsets of pro-survival BCL-2 family proteins. Nevertheless, combined deficiency of Bik and Noxa did not alter the onset of Eµ-Myc transgene induced lymphoma development. Moreover, although p53-mediated induction of Bik has been reported, neither Eµ-Myc/Bik(-/-) nor Eµ-Myc/Bik(-/-)Noxa(-/-) lymphomas were more resistant than control Eµ-Myc lymphomas to killing by DNA damaging drugs, either in vitro or in vivo. These results suggest that Bik, even in combination with Noxa, is not a potent suppressor of c-Myc-driven tumourigenesis or critical for chemotherapeutic drug-induced killing of Myc-driven tumours.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Antineoplastic Agents/pharmacology , DNA Damage/drug effects , Lymphoma/pathology , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Antineoplastic Agents/therapeutic use , Apoptosis , Apoptosis Regulatory Proteins , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Etoposide/pharmacology , Etoposide/therapeutic use , Female , Lymphoma/drug therapy , Lymphoma/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Proteins/genetics , Protein Binding , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/metabolism
16.
Reproduction ; 143(4): 469-76, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22301887

ABSTRACT

Reports indicate that germ-line stem cells present in adult mice can rapidly generate new oocytes and contribute to the primordial follicle reserve following conditions of ovotoxic stress. We further investigated the hypothesis that adult mice have the capacity to generate new oocytes by monitoring primordial follicle numbers throughout postnatal life and following depletion of the primordial follicle reserve by exposure to doxorubicin (DXR), trichostatin A (TSA), or whole-body γ-irradiation. We show that primordial follicle number remains stable in adult C57BL/6 mice between the ages of 25 and 100 days. However, within 2 days of treatment with DXR or TSA, primordial follicle numbers had declined to 65 and 51% respectively (P<0.05-0.01 when compared to untreated controls), with no restoration of follicle numbers evident after 7 days for either treatment. Furthermore, ovaries from mice subjected to sterilizing doses of γ-irradiation (0.45 or 4.5 Gy) revealed complete ablation of all primordial follicles 5 days after treatment, with no indication of follicular renewal. We conclude that neo-folliculogenesis does not occur following chemical or γ-irradiation mediated depletion of the primordial follicle reserve.


Subject(s)
Ovarian Follicle/radiation effects , Animals , Antibiotics, Antineoplastic , Doxorubicin , Female , Gamma Rays , Hydroxamic Acids , Mice , Mice, Inbred C57BL , Ovarian Follicle/drug effects , Protein Synthesis Inhibitors
17.
Aust Dent J ; 56(2): 221-6, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21623817

ABSTRACT

Avoidance of dental care and neglect of oral health may occur in patients with inherited bleeding disorders because of concerns about perioperative and postoperative bleeding, but this is likely to result in the need for crisis care, and more complex and high-risk procedures. Most routine dental care in this special needs group can be safely managed in the general dental setting following consultation with the patient's haematologist and adherence to simple protocols. Many of the current protocols for dental treatment of patients with inherited bleeding disorders were devised many years ago and now need revision. There is increasing evidence that the amount of factor cover previously recommended for dental procedures can now be safely reduced or may no longer be required in many cases. There is still a need for close cooperation and discussion between the patient's haematologist and dental surgeon before any invasive treatment is performed. A group of hospital based dentists from centres where patients with inherited bleeding disorders are treated met and, after discussions, a management protocol for dental treatment was formulated.


Subject(s)
Blood Coagulation Disorders, Inherited , Dental Care for Chronically Ill , Dental Service, Hospital , Anesthesia, Dental , Anesthesia, Local , Blood Coagulation Factors/therapeutic use , Dental Implants , Dental Prosthesis, Implant-Supported , General Practice, Dental , Hematology , Humans , Interprofessional Relations , Oral Surgical Procedures , Orthodontics, Corrective , Periodontal Diseases/therapy , Root Canal Therapy , Tooth Extraction
18.
Am J Physiol Lung Cell Mol Physiol ; 299(4): L455-71, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20622121

ABSTRACT

Lung development requires coordinated signaling between airway and vascular growth, but the link between these processes remains unclear. Mammalian target of rapamycin complex-1 (mTORC1) can amplify hypoxia-inducible factor-1α (HIF-1α) vasculogenic activity through an NH(2)-terminal mTOR binding (TOS) motif. We hypothesized that this mechanism coordinates vasculogenesis with the fibroblast growth factor (FGF)-10/FGF-receptor2b/Spry2 regulator of airway branching. First, we tested if the HIF-1α TOS motif participated in epithelial-mesenchymal vascular signaling. mTORC1 activation by insulin significantly amplified HIF-1α activity at fetal Po(2) (23 mmHg) in human bronchial epithelium (16HBE14o-) and induced vascular traits (Flk1, sprouting) in cocultured human embryonic lung mesenchyme (HEL-12469). This enhanced activation of HIF-1α by mTORC1 was abolished on expression of a HIF-1α (F99A) TOS-mutant and also suppressed vascular differentiation of HEL-12469 cocultures. Next, we determined if vasculogenesis in fetal lung involved regulation of mTORC1 by the FGF-10/FGFR2b/Spry2 pathway. Fetal airway epithelium displayed distinct mTORC1 activity in situ, and its hyperactivation by TSC1(-/-) knockout induced widespread VEGF expression and disaggregation of Tie2-positive vascular bundles. FGF-10-coated beads grafted into fetal lung explants from Tie2-LacZ transgenic mice induced localized vascular differentiation in the peripheral mesenchyme. In rat fetal distal lung epithelial (FDLE) cells cultured at fetal Po(2), FGF-10 induced mTORC1 and amplified HIF-1α activity and VEGF secretion without induction of ERK1/2. This was accompanied by the formation of a complex between Spry2, the cCBL ubiquitin ligase, and the mTOR repressor, TSC2, which abolished GTPase activity directed against Rheb, the G protein inducer of mTORC1. Thus, mTORC1 links HIF-1α-driven vasculogenesis with the FGF-10/FGFR2b/Spry2 airway branching periodicity regulator.


Subject(s)
Biological Clocks , Fibroblast Growth Factor 10/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/metabolism , Neovascularization, Physiologic , Nerve Tissue Proteins/metabolism , Receptor, Fibroblast Growth Factor, Type 2/metabolism , Transcription Factors/metabolism , Animals , Blotting, Western , Bronchi/cytology , Bronchi/metabolism , Cells, Cultured , Circadian Rhythm , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Enzyme-Linked Immunosorbent Assay , Epithelial Cells/metabolism , Fetus/cytology , Fetus/metabolism , Fibroblast Growth Factor 10/genetics , Fibroblasts/cytology , Fibroblasts/metabolism , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Immunoenzyme Techniques , Immunoprecipitation , Lung/cytology , Mesoderm/cytology , Mesoderm/metabolism , Mice , Nerve Tissue Proteins/genetics , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/genetics , Tuberous Sclerosis Complex 1 Protein , Tumor Suppressor Proteins/physiology
19.
Cell Death Differ ; 16(5): 684-96, 2009 May.
Article in English | MEDLINE | ID: mdl-19148184

ABSTRACT

Evasion of apoptosis contributes importantly to c-Myc-induced tumorigenesis. The BH3-only Bcl-2 family members Puma and Noxa are critical pro-apoptotic transcriptional targets of p53, a major mediator of Myc-induced apoptosis and suppressor of Myc-induced tumorigenesis. Hence, we have explored the impact of their individual or combined loss on myc-driven lymphomagenesis. Notably, Puma deficiency both increased B-lineage cells and accelerated the development of B lymphoma, accompanied by leukaemia, but not of pre-B lymphoma. Noxa deficiency alone also increased B-lineage cells but did not accelerate lymphomagenesis. However, its deficiency combined with loss of one puma allele produced more rapid onset of both pre-B and B lymphomas than did loss of a single puma allele alone. Nevertheless, the acceleration evoked by loss of both genes was not as marked as that caused by p53 heterozygosity. These results show that Puma imposes a significant, and Noxa a minor barrier to c-Myc-driven lymphomagenesis. They also indicate that additional BH3-only proteins probably also drive Myc-induced apoptosis and that non-apoptotic functions of p53 may contribute substantially to its tumour suppressor role.


Subject(s)
Apoptosis , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Proteins/genetics , Alleles , Animals , Apoptosis Regulatory Proteins , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Lymphoma, B-Cell/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Precursor Cells, B-Lymphoid/metabolism , Precursor Cells, B-Lymphoid/pathology , Proto-Oncogene Proteins c-bcl-2/deficiency , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/deficiency
20.
Proc Natl Acad Sci U S A ; 104(19): 8071-6, 2007 May 08.
Article in English | MEDLINE | ID: mdl-17470784

ABSTRACT

Histone deacetylase inhibitors (HDACi) can elicit a range of biological responses that affect tumor growth and survival, including inhibition of cell cycle progression, induction of tumor cell-selective apoptosis, suppression of angiogenesis, and modulation of immune responses, and show promising activity against hematological malignancies in clinical trials. Using the Emu-myc model of B cell lymphoma, we screened tumors with defined genetic alterations in apoptotic pathways for therapeutic responsiveness to the HDACi vorinostat. We demonstrated a direct correlation between induction of tumor cell apoptosis in vivo and therapeutic efficacy. Vorinostat did not require p53 activity or a functional death receptor pathway to kill Emu-myc lymphomas and mediate a therapeutic response but depended on activation of the intrinsic apoptotic pathway with the proapoptotic BH3-only proteins Bid and Bim playing an important role. Our studies provide important information regarding the mechanisms of action of HDACi that have broad implications regarding stratification of patients receiving HDACi therapy alone or in combination with other anticancer agents.


Subject(s)
Apoptosis/drug effects , Enzyme Inhibitors/therapeutic use , Histone Deacetylase Inhibitors , Hydroxamic Acids/therapeutic use , Lymphoma, B-Cell/drug therapy , Animals , Apoptosis Regulatory Proteins/physiology , BH3 Interacting Domain Death Agonist Protein/physiology , Bcl-2-Like Protein 11 , Genes, myc , Membrane Proteins/physiology , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-bcl-2/physiology , TNF-Related Apoptosis-Inducing Ligand/physiology , Tumor Suppressor Protein p53/physiology , Vorinostat , bcl-X Protein/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...