Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Sci Signal ; 15(756): eabj3490, 2022 10 18.
Article in English | MEDLINE | ID: mdl-36256706

ABSTRACT

Mutations in guanosine triphosphatase KRAS are common in lung, colorectal, and pancreatic cancers. The constitutive activity of mutant KRAS and its downstream signaling pathways induces metabolic rewiring in tumor cells that can promote resistance to existing therapeutics. In this review, we discuss the metabolic pathways that are altered in response to treatment and those that can, in turn, alter treatment efficacy, as well as the role of metabolism in the tumor microenvironment (TME) in dictating the therapeutic response in KRAS-driven cancers. We highlight metabolic targets that may provide clinical opportunities to overcome therapeutic resistance and improve survival in patients with these aggressive cancers.


Subject(s)
Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction , Guanosine , Cell Line, Tumor , Tumor Microenvironment/genetics
2.
Mol Cancer Ther ; 21(4): 594-606, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35086954

ABSTRACT

Multivalent second-generation TRAIL-R2 agonists are currently in late preclinical development and early clinical trials. Herein, we use a representative second-generation agent, MEDI3039, to address two major clinical challenges facing these agents: lack of predictive biomarkers to enable patient selection and emergence of resistance. Genome-wide CRISPR knockout screens were notable for the lack of resistance mechanisms beyond the canonical TRAIL-R2 pathway (caspase-8, FADD, BID) as well as p53 and BAX in TP53 wild-type models, whereas a CRISPR activatory screen identified cell death inhibitors MCL-1 and BCL-XL as mechanisms to suppress MEDI3039-induced cell death. High-throughput drug screening failed to identify genomic alterations associated with response to MEDI3039; however, transcriptomics analysis revealed striking association between MEDI3039 sensitivity and expression of core components of the extrinsic apoptotic pathway, most notably its main apoptotic effector caspase-8 in solid tumor cell lines. Further analyses of colorectal cell lines and patient-derived xenografts identified caspase-8 expression ratio to its endogenous regulator FLIP(L) as predictive of sensitivity to MEDI3039 in several major solid tumor types and a further subset indicated by caspase-8:MCL-1 ratio. Subsequent MEDI3039 combination screening of TRAIL-R2, caspase-8, FADD, and BID knockout models with 60 compounds with varying mechanisms of action identified two inhibitor of apoptosis proteins (IAP) that exhibited strong synergy with MEDI3039 that could reverse resistance only in BID-deleted models. In summary, we identify the ratios of caspase-8:FLIP(L) and caspase-8:MCL-1 as potential predictive biomarkers for second-generation TRAIL-R2 agonists and loss of key effectors such as FADD and caspase-8 as likely drivers of clinical resistance in solid tumors.


Subject(s)
Proto-Oncogene Proteins c-bcl-2 , TNF-Related Apoptosis-Inducing Ligand , Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Caspase 8/genetics , Cell Line, Tumor , Genomics , Humans , Proto-Oncogene Proteins c-bcl-2/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/pharmacology
3.
Mol Cancer Ther ; 20(9): 1627-1639, 2021 09.
Article in English | MEDLINE | ID: mdl-34389694

ABSTRACT

Inhibitors of apoptosis proteins (IAPs) are intracellular proteins, with important roles in regulating cell death, inflammation, and immunity. Here, we examined the clinical and therapeutic relevance of IAPs in colorectal cancer. We found that elevated expression of cIAP1 and cIAP2 (but not XIAP) significantly correlated with poor prognosis in patients with microsatellite stable (MSS) stage III colorectal cancer treated with 5-fluorouracil (5FU)-based adjuvant chemotherapy, suggesting their involvement in promoting chemoresistance. A novel IAP antagonist tolinapant (ASTX660) potently and rapidly downregulated cIAP1 in colorectal cancer models, demonstrating its robust on-target efficacy. In cells co-cultured with TNFα to mimic an inflammatory tumor microenvironment, tolinapant induced caspase-8-dependent apoptosis in colorectal cancer cell line models; however, the extent of apoptosis was limited because of inhibition by the caspase-8 paralogs FLIP and, unexpectedly, caspase-10. Importantly, tolinapant-induced apoptosis was augmented by FOLFOX in human colorectal cancer and murine organoid models in vitro and in vivo, due (at least in part) to FOLFOX-induced downregulation of class I histone deacetylases (HDAC), leading to acetylation of the FLIP-binding partner Ku70 and downregulation of FLIP. Moreover, the effects of FOLFOX could be phenocopied using the clinically relevant class I HDAC inhibitor, entinostat, which also induced acetylation of Ku70 and FLIP downregulation. Further analyses revealed that caspase-8 knockout RIPK3-positive colorectal cancer models were sensitive to tolinapant-induced necroptosis, an effect that could be exploited in caspase-8-proficient models using the clinically relevant caspase inhibitor emricasan. Our study provides evidence for immediate clinical exploration of tolinapant in combination with FOLFOX in poor prognosis MSS colorectal cancer with elevated cIAP1/2 expression.


Subject(s)
Baculoviral IAP Repeat-Containing 3 Protein/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Colorectal Neoplasms/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Morpholines/pharmacology , Piperazines/pharmacology , Pyrroles/pharmacology , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Cancers (Basel) ; 13(13)2021 Jul 03.
Article in English | MEDLINE | ID: mdl-34283054

ABSTRACT

Drug resistance is a major cause of cancer treatment failure, effectively driven by processes that promote escape from therapy-induced cell death. The mechanisms driving evasion of apoptosis have been widely studied across multiple cancer types, and have facilitated new and exciting therapeutic discoveries with the potential to improve cancer patient care. However, an increasing understanding of the crosstalk between cancer hallmarks has highlighted the complexity of the mechanisms of drug resistance, co-opting pathways outside of the canonical "cell death" machinery to facilitate cell survival in the face of cytotoxic stress. Rewiring of cellular metabolism is vital to drive and support increased proliferative demands in cancer cells, and recent discoveries in the field of cancer metabolism have uncovered a novel role for these programs in facilitating drug resistance. As a key organelle in both metabolic and apoptotic homeostasis, the mitochondria are at the forefront of these mechanisms of resistance, coordinating crosstalk in the event of cellular stress, and promoting cellular survival. Importantly, the appreciation of this role metabolism plays in the cytotoxic response to therapy, and the ability to profile metabolic adaptions in response to treatment, has encouraged new avenues of investigation into the potential of exploiting metabolic addictions to improve therapeutic efficacy and overcome drug resistance in cancer. Here, we review the role cancer metabolism can play in mediating drug resistance, and the exciting opportunities presented by imposed metabolic vulnerabilities.

5.
FEBS J ; 288(18): 5374-5388, 2021 09.
Article in English | MEDLINE | ID: mdl-33660894

ABSTRACT

Resistance to chemotherapy-induced cell death is a major barrier to effective treatment of solid tumours such as colorectal cancer, CRC. Herein, we present a study aimed at developing a proteomics-based predictor of response to standard-of-care (SoC) chemotherapy in combination with antagonists of IAPs (inhibitors of apoptosis proteins), which have been implicated as mediators of drug resistance in CRC. We quantified the absolute expression of 19 key apoptotic proteins at baseline in a panel of 12 CRC cell lines representative of the genetic diversity seen in this disease to identify which proteins promote resistance or sensitivity to a model IAP antagonist [birinapant (Bir)] alone and in combination with SoC chemotherapy (5FU plus oxaliplatin). Quantitative western blotting demonstrated heterogeneous expression of IAP interactome proteins across the CRC cell line panel, and cell death analyses revealed a widely varied response to Bir/chemotherapy combinations. Baseline protein expression of cIAP1, caspase-8 and RIPK1 expression robustly correlated with response to Bir/chemotherapy combinations. Classifying cell lines into 'responsive', 'intermediate' and 'resistant' groups and using linear discriminant analysis (LDA) enabled the identification of a 12-protein signature that separated responders to Bir/chemotherapy combinations in the CRC cell line panel with 100% accuracy. Moreover, the LDA model was able to predict response accurately when cells were cocultured with Tumour necrosis factor-alpha to mimic a pro-inflammatory tumour microenvironment. Thus, our study provides the starting point for a proteomics-based companion diagnostic that predicts response to IAP antagonist/SoC chemotherapy combinations in CRC.


Subject(s)
Alkaline Phosphatase/genetics , Caspase 8/genetics , Colorectal Neoplasms/drug therapy , Inhibitor of Apoptosis Proteins/genetics , Receptor-Interacting Protein Serine-Threonine Kinases/genetics , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Dipeptides/pharmacology , Drug Resistance, Neoplasm/genetics , Fluorouracil/pharmacology , GPI-Linked Proteins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Indoles/pharmacology , Neoplasm Proteins/genetics , Oxaliplatin/pharmacology , Proteomics/standards , Transcriptome/drug effects , Tumor Microenvironment/drug effects
6.
Cell Death Dis ; 11(11): 1020, 2020 11 30.
Article in English | MEDLINE | ID: mdl-33257690

ABSTRACT

Colorectal cancer is a molecularly heterogeneous disease. Responses to genotoxic chemotherapy in the adjuvant or palliative setting vary greatly between patients, and colorectal cancer cells often resist chemotherapy by evading apoptosis. Antagonists of an inhibitor of apoptosis proteins (IAPs) can restore defective apoptosis signaling by degrading cIAP1 and cIAP2 proteins and by inhibition of XIAP. Due to the multiple molecular mechanisms-of-action of these targets, responses to IAP antagonist may differ between molecularly distinct colon cancer cells. In this study, responses to the IAP antagonist Birinapant and oxaliplatin/5-fluorouracil (5-FU) were investigated in 14 colon cancer cell lines, representing the consensus molecular subtypes (CMS). Treatment with Birinapant alone did not result in a substantial increase in apoptotic cells in this cell line panel. Annexin-V/PI assays quantified by flow cytometry and high-content screening showed that Birinapant increased responses of CMS1 and partially CMS3 cell lines to oxaliplatin/5-FU, whereas CMS2 cells were not effectively sensitized. FRET-based imaging of caspase-8 and -3 activation validated these differences at the single-cell level, with CMS1 cells displaying sustained activation of caspase-8-like activity during Birinapant and oxaliplatin/5-FU co-treatment, ultimately activating the intrinsic mitochondrial apoptosis pathway. In CMS2 cell lines, Birinapant exhibited synergistic effects in combination with TNFα, suggesting that Birinapant can restore extrinsic apoptosis signaling in the context of inflammatory signals in this subtype. To explore this further, we co-cultured CMS2 and CMS1 colon cancer cells with peripheral blood mononuclear cells. We observed increased cell death during Birinapant single treatment in these co-cultures, which was abrogated by anti-TNFα-neutralizing antibodies. Collectively, our study demonstrates that IAP inhibition is a promising modulator of response to oxaliplatin/5-FU in colorectal cancers of the CMS1 subtype, and may show promise as in the CMS2 subtype, suggesting that molecular subtyping may aid as a patient stratification tool for IAP antagonists in this disease.


Subject(s)
Colonic Neoplasms/drug therapy , Dipeptides/therapeutic use , Indoles/therapeutic use , Apoptosis , Dipeptides/pharmacology , Humans , Indoles/pharmacology
7.
Proc Natl Acad Sci U S A ; 117(30): 17808-17819, 2020 07 28.
Article in English | MEDLINE | ID: mdl-32661168

ABSTRACT

p53 is the most frequently mutated, well-studied tumor-suppressor gene, yet the molecular basis of the switch from p53-induced cell-cycle arrest to apoptosis remains poorly understood. Using a combination of transcriptomics and functional genomics, we unexpectedly identified a nodal role for the caspase-8 paralog and only human pseudo-caspase, FLIP(L), in regulating this switch. Moreover, we identify FLIP(L) as a direct p53 transcriptional target gene that is rapidly up-regulated in response to Nutlin-3A, an MDM2 inhibitor that potently activates p53. Genetically or pharmacologically inhibiting expression of FLIP(L) using siRNA or entinostat (a clinically relevant class-I HDAC inhibitor) efficiently promoted apoptosis in colorectal cancer cells in response to Nutlin-3A, which otherwise predominantly induced cell-cycle arrest. Enhanced apoptosis was also observed when entinostat was combined with clinically relevant, p53-activating chemotherapy in vitro, and this translated into enhanced in vivo efficacy. Mechanistically, FLIP(L) inhibited p53-induced apoptosis by blocking activation of caspase-8 by the TRAIL-R2/DR5 death receptor; notably, this activation was not dependent on receptor engagement by its ligand, TRAIL. In the absence of caspase-8, another of its paralogs, caspase-10 (also transcriptionally up-regulated by p53), induced apoptosis in Nutlin-3A-treated, FLIP(L)-depleted cells, albeit to a lesser extent than in caspase-8-proficient cells. FLIP(L) depletion also modulated transcription of canonical p53 target genes, suppressing p53-induced expression of the cell-cycle regulator p21 and enhancing p53-induced up-regulation of proapoptotic PUMA. Thus, even in the absence of caspase-8/10, FLIP(L) silencing promoted p53-induced apoptosis by enhancing PUMA expression. Thus, we report unexpected, therapeutically relevant roles for FLIP(L) in determining cell fate following p53 activation.


Subject(s)
CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Acetylation , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/genetics , Benzamides/pharmacology , Caspase 8/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Drug Synergism , Gene Expression Regulation , Humans , Imidazoles/metabolism , Models, Biological , Piperazines/metabolism , Protein Binding , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-mdm2/metabolism , Pyridines/pharmacology , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tumor Suppressor Protein p53/genetics
8.
EMBO Rep ; 21(3): e49254, 2020 03 04.
Article in English | MEDLINE | ID: mdl-32009295

ABSTRACT

The long FLIP splice form FLIP(L) can act as both an inhibitor and promoter of caspase-8 at death-inducing signalling complexes (DISCs) formed by death receptors such as TRAIL-R2 and related intracellular complexes such as the ripoptosome. Herein, we describe a revised DISC assembly model that explains how FLIP(L) can have these opposite effects by defining the stoichiometry (with respect to caspase-8) at which it converts from being anti- to pro-apoptotic at the DISC. We also show that in the complete absence of FLIP(L), procaspase-8 activation at the TRAIL-R2 DISC has significantly slower kinetics, although ultimately the extent of apoptosis is significantly greater. This revised model of DISC assembly also explains why FLIP's recruitment to the TRAIL-R2 DISC is impaired in the absence of caspase-8 despite showing that it can interact with the DISC adaptor protein FADD and why the short FLIP splice form FLIP(S) is the more potent inhibitor of DISC-mediated apoptosis.


Subject(s)
Apoptosis , CASP8 and FADD-Like Apoptosis Regulating Protein , Apoptosis/genetics , CASP8 and FADD-Like Apoptosis Regulating Protein/genetics , Caspase 8/genetics , Caspase 8/metabolism , Humans , Receptors, TNF-Related Apoptosis-Inducing Ligand , Signal Transduction , TNF-Related Apoptosis-Inducing Ligand/genetics
9.
PLoS Comput Biol ; 15(9): e1007374, 2019 09.
Article in English | MEDLINE | ID: mdl-31553717

ABSTRACT

Ligand binding to death receptors activates apoptosis in cancer cells. Stimulation of death receptors results in the formation of intracellular multiprotein platforms that either activate the apoptotic initiator Caspase-8 to trigger cell death, or signal through kinases to initiate inflammatory and cell survival signalling. Two of these platforms, the Death-Inducing Signalling Complex (DISC) and the RIPoptosome, also initiate necroptosis by building filamentous scaffolds that lead to the activation of mixed lineage kinase domain-like pseudokinase. To explain cell decision making downstream of death receptor activation, we developed a semi-stochastic model of DISC/RIPoptosome formation. The model is a hybrid of a direct Gillespie stochastic simulation algorithm for slow assembly of the RIPoptosome and a deterministic model of downstream caspase activation. The model explains how alterations in the level of death receptor-ligand complexes, their clustering properties and intrinsic molecular fluctuations in RIPoptosome assembly drive heterogeneous dynamics of Caspase-8 activation. The model highlights how kinetic proofreading leads to heterogeneous cell responses and results in fractional cell killing at low levels of receptor stimulation. It reveals that the noise in Caspase-8 activation-exclusively caused by the stochastic molecular assembly of the DISC/RIPoptosome platform-has a key function in extrinsic apoptotic stimuli recognition.


Subject(s)
Apoptosis/physiology , Caspase 8 , Models, Biological , Receptors, Death Domain , Caspase 8/chemistry , Caspase 8/metabolism , Cell Survival/physiology , Computational Biology , Humans , Neoplasms/metabolism , Receptors, Death Domain/chemistry , Receptors, Death Domain/metabolism
10.
Cell Death Dis ; 9(11): 1081, 2018 10 22.
Article in English | MEDLINE | ID: mdl-30349042

ABSTRACT

Expression of tumor necrosis factor-α (TNFα) in the serum of prostate cancer patients is associated with poorer outcome and progression to castrate-resistant (CRPC) disease. TNFα promotes the activity of NFκB, which regulates a number of anti-apoptotic and proinflammatory genes, including those encoding the inhibitor of apoptosis proteins (IAPs); however, in the presence of IAP antagonists, TNFα can induce cell death. In the presence of recombinant or macrophage-derived TNFα, we found that IAP antagonists triggered degradation of cIAP1 and induced formation of Complex-IIb, consisting of caspase-8, FADD and RIPK1 in CRPC models; however, no, or modest levels of apoptosis were induced. This resistance was found to be mediated by both the long (L) and short (S) splice forms of the caspase-8 inhibitor, FLIP, another NFκB-regulated protein frequently overexpressed in CRPC. By decreasing FLIP expression at the post-transcriptional level in PC3 and DU145 cells (but not VCaP), the Class-I histone deacetylase (HDAC) inhibitor Entinostat promoted IAP antagonist-induced cell death in these models in a manner dependent on RIPK1, FADD and Caspase-8. Of note, Entinostat primarily targeted the nuclear rather than cytoplasmic pool of FLIP(L). While the cytoplasmic pool of FLIP(L) was highly stable, the nuclear pool was more labile and regulated by the Class-I HDAC target Ku70, which we have previously shown regulates FLIP stability. The efficacy of IAP antagonist (TL32711) and Entinostat combination and their effects on cIAP1 and FLIP respectively were confirmed in vivo, highlighting the therapeutic potential for targeting IAPs and FLIP in proinflammatory CRPC.


Subject(s)
Apoptosis/drug effects , CASP8 and FADD-Like Apoptosis Regulating Protein/metabolism , Cell Nucleus/drug effects , Cytoplasm/drug effects , Drug Resistance, Neoplasm/drug effects , Histone Deacetylase Inhibitors/pharmacology , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Caspase 8/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cytoplasm/metabolism , Histone Deacetylases/metabolism , Humans , Male , Mice , Mice, Inbred BALB C , Mice, SCID , NF-kappa B/metabolism , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/metabolism , THP-1 Cells , Tumor Necrosis Factor-alpha/metabolism
11.
Diagn Progn Res ; 2: 18, 2018.
Article in English | MEDLINE | ID: mdl-31093566

ABSTRACT

BACKGROUND: In recent years, there has been increasing focus on the earlier detection of deterioration in the clinical condition of hospital patients with the aim of instigating earlier treatment to reverse this deterioration and prevent adverse outcomes. This is especially important in the ED, a dynamic environment with large volumes of undifferentiated patients, which carries inherent patient risk. SNAP40 is an innovative medical-grade device that can be worn on the upper arm that continuously monitors patients' vital signs including relative changes in systolic blood pressure, respiratory rate, heart rate, movement, blood oxygen saturation and temperature. It uses automated risk analysis to potentially allow clinical staff to easily and quickly identify high-risk patients. The aim of this study is to investigate whether the SNAP40 device is able to identify deterioration in the vital sign physiology of an ED patient earlier than current standard monitoring and observation charting techniques. METHODS/DESIGN: Single centre, teaching hospital ED open label, prospective, observational cohort study recruiting 250 high acuity participants aged 16 years or over presenting to the ED. Participants will be approached and enrolled in the ED and after consent will have the SNAP40 wearable monitoring device attached which will be used alongside standard care monitoring. Participants will be observed throughout their time in the ED. Any SNAP40 device alarm, standard monitoring alarms or standard practice vital sign observations indicating a deterioration in a patient's vital sign physiology (defined as an increase in NEWS score) will be recorded. Primary outcome is time to detection of deterioration. Secondary outcomes include staff time spent performing observations and responding to standard monitoring alarms, clinical escalation of care when deterioration is detected and participants and staff rating of experience of both SNAP40 and current monitoring. DISCUSSION: The SNAP40-ED study aims to recruit 250 patients. It will be the first study to compare the ability of a novel ambulatory monitoring device to detect deterioration compared to standard care in the ED. It may allow the earlier detection of deterioration in the clinical condition of ED patients and therefore earlier treatment to reverse this deterioration and prevent adverse outcomes. TRIAL REGISTRATION: NCT03179267 ClinicalTrials.gov. Registered on June 17, 2017.

12.
Oncotarget ; 7(7): 7885-98, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26799286

ABSTRACT

PTEN loss is prognostic for patient relapse post-radiotherapy in prostate cancer (CaP). Infiltration of tumor-associated macrophages (TAMs) is associated with reduced disease-free survival following radical prostatectomy. However, the association between PTEN loss, TAM infiltration and radiotherapy response of CaP cells remains to be evaluated. Immunohistochemical and molecular analysis of surgically-resected Gleason 7 tumors confirmed that PTEN loss correlated with increased CXCL8 expression and macrophage infiltration. However PTEN status had no discernable correlation with expression of other inflammatory markers by CaP cells, including TNF-α. In vitro, exposure to conditioned media harvested from irradiated PTEN null CaP cells induced chemotaxis of macrophage-like THP-1 cells, a response partially attenuated by CXCL8 inhibition. Co-culture with THP-1 cells resulted in a modest reduction in the radio-sensitivity of DU145 cells. Cytokine profiling revealed constitutive secretion of TNF-α from CaP cells irrespective of PTEN status and IR-induced TNF-α secretion from THP-1 cells. THP-1-derived TNF-α increased NFκB pro-survival activity and elevated expression of anti-apoptotic proteins including cellular inhibitor of apoptosis protein-1 (cIAP-1) in CaP cells, which could be attenuated by pre-treatment with a TNF-α neutralizing antibody. Treatment with a novel IAP antagonist, AT-IAP, decreased basal and TNF-α-induced cIAP-1 expression in CaP cells, switched TNF-α signaling from pro-survival to pro-apoptotic and increased radiation sensitivity of CaP cells in co-culture with THP-1 cells. We conclude that targeting cIAP-1 can overcome apoptosis resistance of CaP cells and is an ideal approach to exploit high TNF-α signals within the TAM-rich microenvironment of PTEN-deficient CaP cells to enhance response to radiotherapy.


Subject(s)
Chemoradiotherapy , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Macrophages/pathology , PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/radiotherapy , Radiation-Sensitizing Agents/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Blotting, Western , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cells, Cultured , Chemotaxis/drug effects , Chemotaxis/radiation effects , DNA Methylation/drug effects , DNA Methylation/radiation effects , Flow Cytometry , Humans , Immunoenzyme Techniques , Inhibitor of Apoptosis Proteins/drug effects , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-8/metabolism , Macrophages/drug effects , Macrophages/radiation effects , Male , Neoplasm Grading , Prognosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , X-Rays
13.
Clin Obstet Gynecol ; 56(1): 25-34, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23337842

ABSTRACT

Cervical cancer and human papillomavirus-related diseases continue to cause significant morbidity and mortality in the United States and worldwide. As we begin to understand the natural course of human papillomavirus infection, and the consequences of both its detection and treatment, changes have been made to our clinical approaches. The purpose of this review is to outline the management guidelines for the management of abnormal cytology. Successful triage of abnormal cytology in 2012 will allow for continued detection of precancerous lesions reducing the incidence of cervical cancer and increasing the detection of early stage disease.


Subject(s)
Cervix Uteri/pathology , Uterine Cervical Dysplasia/pathology , Uterine Cervical Neoplasms/pathology , Adolescent , Alphapapillomavirus , Colposcopy , Early Detection of Cancer , Female , Humans , Papillomavirus Infections/complications , Postmenopause , Practice Guidelines as Topic , Pregnancy , Uterine Cervical Neoplasms/prevention & control , Uterine Cervical Neoplasms/virology , Vaginal Smears , Uterine Cervical Dysplasia/prevention & control , Uterine Cervical Dysplasia/virology
14.
Palliat Support Care ; 11(4): 289-93, 2013 Aug.
Article in English | MEDLINE | ID: mdl-22854052

ABSTRACT

Dying is an act of creativity, and we each die as cultural beings. Culture helps us create the meaning death requests of us. However, the dominant culture of the healthcare system views death as a failure of modern medicine, an event of unspeakable terror and taboo. Palliative clinicians must honor each dying person's cultural identity (as well as the person's family), not subject it to the dominant discourse of Western medicine. This article offers practical guidelines for palliative clinicians to do so, as well as a case vignette.


Subject(s)
Alzheimer Disease/nursing , Attitude to Death , Cultural Characteristics , Empathy , Nurse-Patient Relations , Palliative Care/psychology , Terminal Care/psychology , Aged , Attitude of Health Personnel , Female , Hispanic or Latino/psychology , Humans , Practice Guidelines as Topic
15.
PLoS One ; 6(11): e28077, 2011.
Article in English | MEDLINE | ID: mdl-22140510

ABSTRACT

BACKGROUND: Recent evidence links aberrant activation of Hedgehog (Hh) signaling with the pathogenesis of several cancers including medulloblastoma, basal cell, small cell lung, pancreatic, prostate and ovarian. This investigation was designed to determine if inhibition of this pathway could inhibit serous ovarian cancer growth. METHODOLOGY: We utilized an in vivo pre-clinical model of serous ovarian cancer to characterize the anti-tumor activity of Hh pathway inhibitors cyclopamine and a clinically applicable derivative, IPI-926. Primary human serous ovarian tumor tissue was used to generate tumor xenografts in mice that were subsequently treated with cyclopamine or IPI-926. PRINCIPAL FINDINGS: Both compounds demonstrated significant anti-tumor activity as single agents. When IPI-926 was used in combination with paclitaxel and carboplatinum (T/C), no synergistic effect was observed, though sustained treatment with IPI-926 after cessation of T/C continued to suppress tumor growth. Hh pathway activity was analyzed by RT-PCR to assess changes in Gli1 transcript levels. A single dose of IPI-926 inhibited mouse stromal Gli1 transcript levels at 24 hours with unchanged human intra-tumor Gli1 levels. Chronic IPI-926 therapy for 21 days, however, inhibited Hh signaling in both mouse stromal and human tumor cells. Expression data from the micro-dissected stroma in human serous ovarian tumors confirmed the presence of Gli1 transcript and a significant association between elevated Gli1 transcript levels and worsened survival. CONCLUSIONS/SIGNIFICANCE: IPI-926 treatment inhibits serous tumor growth suggesting the Hh signaling pathway contributes to the pathogenesis of ovarian cancer and may hold promise as a novel therapeutic target, especially in the maintenance setting.


Subject(s)
Hedgehog Proteins/metabolism , Ovarian Neoplasms/pathology , Signal Transduction , Xenograft Model Antitumor Assays , Animals , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Hedgehog Proteins/genetics , Humans , Maintenance Chemotherapy , Mice , Neoplasms, Cystic, Mucinous, and Serous/drug therapy , Neoplasms, Cystic, Mucinous, and Serous/genetics , Neoplasms, Cystic, Mucinous, and Serous/pathology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction/drug effects , Stromal Cells/drug effects , Stromal Cells/metabolism , Stromal Cells/pathology , Survival Analysis , Transcription Factors/genetics , Transcription Factors/metabolism , Veratrum Alkaloids/pharmacology , Veratrum Alkaloids/therapeutic use , Zinc Finger Protein GLI1
16.
Am J Obstet Gynecol ; 205(6): 565.e1-6, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21855843

ABSTRACT

OBJECTIVE: We sought to examine the evolution of surgical care for early-stage endometrial cancers and factors affecting use of laparoscopy. STUDY DESIGN: Women with surgically managed early-stage endometrial cancer were divided into 2 groups corresponding to before and after addition of faculty with formal fellowship training in laparoscopic staging and access to a robotic surgery platform. RESULTS: In all, 502 women were identified. Laparoscopic management increased from 24-69% between time periods (P < .0001). Performance of comprehensive surgical staging, and lymph node counts, increased (P < .0001) despite an increase in median body mass index (P = .001). A traditional "straight stick" technique was performed in 72% of laparoscopic cases during the later period. Laparoscopy patients had lower estimated blood losses and shorter hospital stays (each P < .0001) compared to laparotomy patients. CONCLUSION: Addition of faculty with formal fellowship training in laparoscopic staging and access to a robotic surgery platform shifted management of early-stage endometrial cancer toward laparoscopy.


Subject(s)
Endometrial Neoplasms/pathology , Endometrial Neoplasms/surgery , Laparoscopy , Neoplasm Staging/methods , Preoperative Care/methods , Adult , Aged , Aged, 80 and over , Blood Loss, Surgical , Body Mass Index , Early Diagnosis , Endometrial Neoplasms/epidemiology , Fellowships and Scholarships , Female , Humans , Laparoscopy/education , Laparoscopy/methods , Laparoscopy/statistics & numerical data , Laparotomy/education , Laparotomy/methods , Laparotomy/statistics & numerical data , Length of Stay , Lymph Node Excision/education , Lymph Node Excision/methods , Lymph Nodes/pathology , Medical Staff, Hospital/education , Middle Aged , Neoplasm Staging/statistics & numerical data , Neoplasm Staging/trends , Preoperative Care/statistics & numerical data , Preoperative Care/trends , Retrospective Studies , Robotics/education , Robotics/methods , Robotics/statistics & numerical data
17.
Ann Surg Oncol ; 18(10): 2912-8, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21424880

ABSTRACT

PURPOSE: We sought to examine how splenectomy as part of up-front cytoreductive surgery in ovarian cancer influences the postoperative course and affects survival. METHODS: We reviewed cases of ovarian cancer diagnosed at Massachusetts General Hospital from 1994 to 2008 and found 44 patients who had a splenectomy as part of their up-front cytoreductive surgery. These were compared to 171 patients who did not undergo splenectomy. We evaluated age at diagnosis, estimated blood loss, percentage of patients whose disease was optimally cytoreduced (<1 cm), reason for splenectomy (oncologic vs. surgical), length of stay, time to first chemotherapy treatment, and survival. RESULTS: In the splenectomy cohort, the mean age at diagnosis was 64 (44-83) years. A total of 37 of 44 (84%) patients were optimally cytoreduced. Mean estimated blood loss was 1326 ml. The purpose of splenectomy was to accomplish an optimal cytoreduction (oncologic) in 82% of cases. Median length of stay was 13 (6-76) days. Median time to first chemotherapy was 13.5 (5-54) days. The median disease-free interval and overall survival of the splenectomy cohort were 8 and 30 months, respectively. The median overall survival for patients whose disease was optimally cytoreduced in the splenectomy cohort compared to the no-splenectomy group was 30 and 45 months (P < 0.045), respectively. CONCLUSIONS: The addition of splenectomy to up-front cytoreductive surgery was feasible and safe. However, it appears to carry with it a shortened survival that is unrelated to postoperative morbidity. Our data raise the questions that splenectomy is needed for optimal cytoreduction in more biologically aggressive disease and that splenectomy may be an independent prognostic factor related to depressed immune function.


Subject(s)
Adenocarcinoma, Clear Cell/surgery , Cystadenocarcinoma, Serous/surgery , Ovarian Neoplasms/surgery , Splenectomy/mortality , Adenocarcinoma, Clear Cell/mortality , Adenocarcinoma, Clear Cell/pathology , Adult , Aged , Aged, 80 and over , Cystadenocarcinoma, Serous/mortality , Cystadenocarcinoma, Serous/pathology , Female , Follow-Up Studies , Humans , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/mortality , Ovarian Neoplasms/pathology , Prognosis , Survival Rate
18.
Rev Obstet Gynecol ; 4(3-4): 117-22, 2011.
Article in English | MEDLINE | ID: mdl-22229064

ABSTRACT

Since its advent in the early 1990s, laparoscopic surgical staging for early ovarian cancer has been explored as an option with the potential to offer women equivalent cancer control and survival as provided by laparotomy but with the clear benefits of minimally invasive surgery. A limited but expanding body of literature suggests aggressive surgical staging can be performed with equivalent tissue assessment compared with laparotomy. Given the lack of randomized, controlled trials, the risks and benefits of such a procedure remain ambiguous. This review summarizes the current body of literature regarding the role of laparoscopy in upfront surgical staging of ovarian cancer. This review presents the history, rationale, and established benefits and risks of utilizing this approach in women who present with malignancy that appears confined to the ovary. Although retrospective data confirm the feasibility, safety, and efficacy of laparoscopic staging of early ovarian cancer, more prospective data will be required to confirm equivalent survival in a patient population that has the potential to be cured.

19.
Gynecol Oncol ; 119(2): 299-304, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20691465

ABSTRACT

OBJECTIVE: The aim of this study was to evaluate the survival impact of cytoreductive surgery and other prognostic determinants in patients with stage IIIC and IV uterine papillary serous carcinoma (UPSC). METHODS: All patients with FIGO stage IIIC and IV UPSC who underwent surgical staging at the two participating institutions, between January 1, 1995 and December 31, 2007, were identified from the tumor registry database. The Kaplan-Meier method was used to generate overall survival (OS) data. Factors predictive of outcome were compared using the log-rank test and Cox regression analysis. RESULTS: Analysis of 79 patients with stage IIIC-IV disease was performed. Optimal cytoreduction was associated with a median survival of 36 months, compared with 12 months for patients who underwent a suboptimal surgical effort (p=0.001), and a disease-free survival (DFS) of 21 months vs. 10 months (p=0.001), respectively. Regression analysis identified stage (HR=2.4, p=0.03), absence of visible residual disease (HR=0.5, p=0.03), and chemotherapy (HR=0.1, p<0.001) as independent predictors of OS. CONCLUSIONS: Cytoreduction to no gross residual disease and the use of platinum therapy are associated with a significant survival benefit for patients with stage IIIC-IV UPSC. Recommended management for this group of patients should consist of maximal surgical cytoreduction followed by platinum-based chemotherapy, preferably in combination with paclitaxel. Adjuvant radiation therapy should also be considered.


Subject(s)
Carcinoma, Papillary/pathology , Cystadenocarcinoma, Serous/pathology , Uterine Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Carcinoma, Papillary/drug therapy , Carcinoma, Papillary/radiotherapy , Carcinoma, Papillary/surgery , Chemotherapy, Adjuvant , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/radiotherapy , Cystadenocarcinoma, Serous/surgery , Disease-Free Survival , Female , Humans , Middle Aged , Neoplasm Staging , Radiotherapy, Adjuvant , Survival Rate , Uterine Neoplasms/drug therapy , Uterine Neoplasms/radiotherapy , Uterine Neoplasms/surgery
20.
Front Biosci (Elite Ed) ; 2(3): 882-905, 2010 06 01.
Article in English | MEDLINE | ID: mdl-20515761

ABSTRACT

Uterine tumors, whether benign or malignant, are diagnosed in a significant portion of women and are associated with a number of co-morbidities that negatively impact quality of life. Uterine tumors can be derived from the epithelial (endometrial hyperplasia or carcinoma) and mesenchymal (leiomyoma, sarcoma) layers of the uterus. The exact etiologies of the various tumor types are yet to be defined. Collectively their development and progression often results from aberrant steroid hormone exposure or dysregulation of related growth factor signaling and apoptotic pathways, reflecting the role of steroid hormone-dependent signaling and survival pathways in the cycles of cell growth and involution that characterize normal uterine physiology. While molecular analyses of human tumors can identify candidate genetic and epigenetic lesions contributing to uterine tumor initiation and progression, in vivo genetic models are needed to establish the functional significance of such lesions and their contribution to tumorigenesis. For this purpose, genetically-engineered mouse models have proven valuable. Here we review genetically-modified mouse models that develop uterine tumors and compare their pathology, utility/feasibility, and discuss their clinical relevance.


Subject(s)
Disease Models, Animal , Uterine Neoplasms/pathology , Animals , Female , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...