Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
2.
Clin Lung Cancer ; 24(6): e205-e213, 2023 09.
Article in English | MEDLINE | ID: mdl-37188597

ABSTRACT

BACKGROUND: Often, patients with NSCLC experience recurrent/refractory (R/R) disease within 2 years of chemoradiation (CRT) and consolidative durvalumab. Despite prior immune checkpoint inhibitor exposure, immunotherapy with or without chemotherapy is typically initiated if a driver-oncogene is absent. However, there remains a paucity of data regarding the efficacy of immunotherapy in this patient population. Here, we present survival outcomes associated with pembrolizumab for R/R NSCLC. MATERIALS AND METHODS: We retrospectively assessed adults with NSCLC who received pembrolizumab for R/R disease between January 2016 to January 2023. Primary objective was to estimate OS and PFS in this cohort compared to historical outcomes. Secondary objective was to compare OS and PFS among subgroups. RESULTS: Fifty patients were evaluated. Median follow-up time was 11.3 months (2.9-38.2). OS was 10.6 months (95% CI, 8.8-19.2); 1-year OS rate 49% (95% CI, 36 - 67%). PFS was 6.1 months (95% CI, 4.7-9.0); 1-year PFS rate 25% (95% CI, 15%-42%). Current smokers had significantly better median OS/PFS as compared to former smokers (NA vs. 10.5 and 9.9 vs. 6.0 months, respectively). The addition of chemotherapy demonstrated an OS benefit (median OS 12.9 vs. 6.0 months) but was not statistically significant. CONCLUSION: Patients with R/R NSCLC represent a distinct cohort with inferior survival outcomes when compared to those with de novo stage IV disease treated with pembrolizumab-based regimens. Based on our findings, we recommend oncologists exercise caution when considering checkpoint inhibitor monotherapy in the front-line setting for R/R NSCLC, regardless of PD-L1 expression.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Adult , Humans , Retrospective Studies , Lung Neoplasms/drug therapy , Antibodies, Monoclonal/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy
3.
J Natl Compr Canc Netw ; 19(8): 915-921, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33878726

ABSTRACT

BACKGROUND: Bone metastases and skeletal-related events (SREs) are a frequent cause of morbidity in patients with metastatic non-small cell lung cancer (mNSCLC). Data are limited on bone metastases and SREs in patients with mNSCLC treated using immune checkpoint inhibitors (ICIs), and on the efficacy of bone-modifying agents (BMAs) in this setting. Here we report the incidence, impact on survival, risk factors for bone metastases and SREs, and impact of BMAs in patients with mNSCLC treated with ICIs in a multi-institutional cohort. PATIENTS AND METHODS: We conducted a retrospective study of patients with mNSCLC treated with ICIs at 2 tertiary care centers from 2014 through 2017. Overall survival (OS) was compared between patients with and without baseline bone metastases using a log-rank test. A Cox regression model was used to evaluate the association between OS and the presence of bone metastases at ICI initiation, controlling for other confounding factors. RESULTS: We identified a cohort of 330 patients who had received ICIs for metastatic disease. Median patient age was 63 years, most patients were treated in the second line or beyond (n=259; 78%), and nivolumab was the most common ICI (n=211; 64%). Median OS was 10 months (95% CI, 8.4-12.0). In our cohort, 124 patients (38%) had baseline bone metastases, and 43 (13%) developed SREs during or after ICI treatment. Patients with bone metastases had a higher hazard of death after controlling for performance status, histology, line of therapy, and disease burden (hazard ratio, 1.57; 95% CI, 1.19-2.08; P=.001). Use of BMAs was not associated with OS or a decreased risk of SREs. CONCLUSIONS: Presence of bone metastases at baseline was associated with a worse prognosis for patients with mNSCLC treated with ICI after controlling for multiple clinical characteristics. Use of BMAs was not associated with reduced SREs or a difference in survival.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/pathology , Middle Aged , Nivolumab/therapeutic use , Retrospective Studies
4.
Cancers (Basel) ; 12(1)2020 Jan 03.
Article in English | MEDLINE | ID: mdl-31947893

ABSTRACT

(1) Background: Circulating tumor cell (CTC) clusters are emerging as clinically significant harbingers of metastases in solid organ cancers. Prior to engaging these CTC clusters in animal models of metastases, it is imperative for technology to identify them with high sensitivity. These clusters often present heterogeneous surface markers and current methods for isolation of clusters may fall short. (2) Methods: We applied an inertial microfluidic Labyrinth device for high-throughput, biomarker-independent, size-based isolation of CTCs/CTC clusters from patients with metastatic non-small-cell lung cancer (NSCLC). (3) Results: Using Labyrinth, CTCs (PanCK+/DAPI+/CD45-) were isolated from patients (n = 25). Heterogeneous CTC populations, including CTCs expressing epithelial (EpCAM), mesenchymal (Vimentin) or both markers were detected. CTCs were isolated from 100% of patients (417 ± 1023 CTCs/mL). EpCAM- CTCs were significantly greater than EpCAM+ CTCs. Cell clusters of ≥2 CTCs were observed in 96% of patients-of which, 75% were EpCAM-. CTCs revealed identical genetic aberrations as the primary tumor for RET, ROS1, and ALK genes using fluorescence in situ hybridization (FISH) analysis. (4) Conclusions: The Labyrinth device recovered heterogeneous CTCs in 100% and CTC clusters in 96% of patients with metastatic NSCLC. The majority of recovered CTCs/clusters were EpCAM-, suggesting that these would have been missed using traditional antibody-based capture methods.

5.
Int J Radiat Oncol Biol Phys ; 108(1): 170-177, 2020 09 01.
Article in English | MEDLINE | ID: mdl-31756415

ABSTRACT

PURPOSE: Preclinical data and subset analyses from immunotherapy clinical trials indicate that prior radiation therapy was associated with better progression-free survival and overall survival when combined with immune checkpoint inhibitors in patients with non-small cell lung cancer. We present a prospective study of hypofractionated image guided radiation therapy (HIGRT) to a single site of metastatic disease concurrently with atezolizumab in patients with metastatic non-small cell lung cancer. METHODS AND MATERIALS: Patients meeting eligibility criteria received 1200 mg of atezolizumab intravenously every 3 weeks with concurrent 3- or 5-fraction HIGRT starting no later than the second cycle. The 3-fraction regimen employed a minimum of 8 Gy per fraction compared with 6 Gy for the 5-fraction regimen. Imaging was obtained every 12 weeks to assess response. RESULTS: From October 2015 to February 2017, 12 patients were enrolled in the study (median age 64; range, 55-77 years). The best response by the Response Evaluation in Solid Tumors criteria was partial response in 3 and stable disease in 3, for a disease control rate of 50%. Five patients had a grade 3 immune-related adverse event, including choreoretinitis (n = 1), pneumonitis (n = 1), transaminitis (n = 1), fatigue (n = 1), and peripheral neuropathy (n = 1). The median progression-free survival was 2.3 months, and the median overall survival was 6.9 months (range, 0.4-not reached). There was no clear association between peripheral blood T cell repertoire characteristics at baseline, PD-L1, or tumor mutations and response or outcome. One long-term survivor exhibited oligoclonal T cell populations in a baseline tumor biopsy that were consistently detected in peripheral blood over the entire course of the study. CONCLUSIONS: HIGRT plus atezolizumab resulted in an overall response rate of 25% and disease control rate of 50% in this pilot study. The incidence of grade 3 adverse events was similar to that of atezolizumab alone. Alhough it was a pilot study with limited sample size, the results generated hypotheses worthy of further investigation.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Radiation Dose Hypofractionation , Radiotherapy, Image-Guided , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/metabolism , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/pathology , Combined Modality Therapy , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/pathology , Male , Middle Aged , Pilot Projects , Safety , Treatment Outcome
6.
Oncogene ; 38(28): 5749, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31197211

ABSTRACT

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Nat Commun ; 10(1): 1430, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926774

ABSTRACT

Although clinically apparent metastasis is associated with late stages of cancer development, micro-metastatic dissemination may be an early event. However, the fate of these early disseminated tumor cells (DTC) remains elusive. We show that despite their capacity to disseminate into secondary organs, 4T1 tumor models develop overt metastasis while EMT6-tumor bearing mice clear DTCs shed from primary tumors as well as those introduced by intravenous (IV) injection. Following the surgical resection of primary EMT6 tumors, mice do not develop detectable metastasis and reject IV-injected tumor cells. In contrast, these cells readily grow and metastasize in immuno-deficient athymic or Rag2-/- mice, an effect mimicked by CD8+ T-cell depletion in immunocompetent mice. Furthermore, recombinant G-CSF or adoptive transfer of granulocytic-MDSCs isolated from 4T1 tumor-bearing mice, induce metastasis by suppressing CD8+ T-cells in EMT6-primed mice. Our studies support the concept of immune surveillance providing molecular insights into the immune mechanisms during tumor progression.


Subject(s)
Immunity , Neoplasms/immunology , Neoplasms/pathology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Humans , Lymphocyte Subsets/immunology , Mice , Models, Biological , Neoplasm Invasiveness , Neoplasm Metastasis , Survival Analysis , Tail/blood supply , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/immunology , Triple Negative Breast Neoplasms/surgery , Veins/pathology
9.
Oncogene ; 38(4): 469-482, 2019 01.
Article in English | MEDLINE | ID: mdl-30166590

ABSTRACT

TNFα is a pleiotropic cytokine which fuels tumor cell growth, invasion, and metastasis in some malignancies, while in others it induces cytotoxic cell death. However, the molecular mechanism by which TNFα exerts its diverse effects on breast cancer subtypes remains elusive. Using in vitro assays and mouse xenografts, we show here that TNFα contributes to the aggressive properties of triple negative breast cancer (TNBC) cell lines via upregulation of TNFAIP3(A20). In a striking contrast, TNFα induces a potent cytotoxic cell death in luminal (ER+) breast cancer cell lines which fail to upregulate A20 expression. Overexpression of A20 not only protects luminal breast cancer cell lines from TNFα-induced cell death via inducing HSP70-mediated anti-apoptotic pathway but also promotes a robust EMT/CSC phenotype by activating the pStat3-mediated inflammatory signaling. Furthermore, A20 overexpression in luminal breast cancer cells induces aggressive metastatic properties in mouse xenografts via generating a permissive inflammatory microenvironment constituted by granulocytic-MDSCs. Collectively, our results reveal a mechanism by which A20 mediates pleiotropic effects of TNFα playing role in aggressive behaviors of TNBC subtype while its deficiency results in TNFα-induced apoptotic cell death in luminal breast cancer subtype.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Genetic Pleiotropy , Neoplasm Proteins/physiology , Tumor Necrosis Factor alpha-Induced Protein 3/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , Apoptosis/physiology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Female , HSP72 Heat-Shock Proteins/antagonists & inhibitors , HSP72 Heat-Shock Proteins/physiology , Heterografts , Humans , Inflammation , Lung Neoplasms/secondary , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Invasiveness/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Purine Nucleosides/pharmacology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , STAT3 Transcription Factor/physiology , Signal Transduction , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Necrosis Factor alpha-Induced Protein 3/biosynthesis , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor-alpha/genetics
10.
Cancer Res ; 78(24): 6728-6735, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30333118

ABSTRACT

: Almost all patients with EGFR-driven lung cancer who are treated with EGFR tyrosine kinase inhibitors (TKI) develop resistance to treatment. A single base (c.2369C>T) transition mutation, EGFR T790M, is the most frequent resistance event after first-generation exposure to EGFR TKIs. Whether T790M mutation is acquired or is selected from a preexisting clone has been a matter of significant debate. In this study, we show that treatment with EGFR TKIs leads to activation of the NFκB pathway, which in turn induces expression of activation-induced cytidine deaminase (AICDA). In turn, AICDA causes deamination of 5-methylcytosine to thymine at position c.2369 to generate the T790M mutation. Pharmacologic inhibition of the NFκB pathway or knockout of AICDA decreased the frequency or prevented the development of T790M mutation, respectively. In addition, patients treated with first-line EGFR TKI displayed increased expression of AICDA and detection of the T790M mutation upon progression. These results identify the mechanism of T790M acquisition and present an opportunity to target the process to delay or prevent it. SIGNIFICANCE: These findings identify the mechanism behind acquisition of a common resistance mutation to TKI treatment in lung cancer.


Subject(s)
5-Methylcytosine/chemistry , Cytidine Deaminase/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Aged , Cell Line, Tumor , Deamination , Disease Progression , ErbB Receptors/genetics , Female , Humans , Hydrolysis , Male , Mass Spectrometry , Methylation , Middle Aged , Mutation , NF-kappa B/metabolism , Polymerase Chain Reaction
11.
J Thorac Dis ; 10(2): 554-556, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29607110
12.
JAMA Oncol ; 3(10): 1358-1365, 2017 Oct 01.
Article in English | MEDLINE | ID: mdl-28570742

ABSTRACT

IMPORTANCE: Our previous studies demonstrated that tumors significantly decrease in size and metabolic activity after delivery of 45 Gy of fractionated radiatiotherapy (RT), and that metabolic shrinkage is greater than anatomic shrinkage. This study aimed to determine whether 18F-fludeoxyglucose-positron emission tomography/computed tomography (FDG-PET/CT) acquired during the course of treatment provides an opportunity to deliver higher-dose radiation to the more aggressive areas of the tumor to improve local tumor control without increasing RT-induced lung toxicity (RILT), and possibly improve survival. OBJECTIVE: To determine whether adaptive RT can target high-dose radiation to the FDG-avid tumor on midtreatment FDG-PET to improve local tumor control of locally advanced non-small-cell lung cancer (NSCLC). DESIGN, SETTING, AND PARTICIPANTS: A phase 2 clinical trial conducted at 2 academic medical centers with 42 patients who had inoperable or unresectable stage II to stage III NSCLC enrolled from November 2008, to May 2012. Patients with poor performance, more than 10% weight loss, poor lung function, and/or oxygen dependence were included, providing that the patients could tolerate the procedures of PET scanning and RT. INTERVENTION: Conformal RT was individualized to a fixed risk of RILT (grade >2) and adaptively escalated to the residual tumor defined on midtreatment FDG-PET up to a total dose of 86 Gy in 30 daily fractions. Medically fit patients received concurrent weekly carboplatin plus paclitaxel followed by 3 cycles of consolidation. MAIN OUTCOMES AND MEASURES: The primary end point was local tumor control. The trial was designed to achieve a 20% improvement in 2-year control from 34% of our prior clinical trial experience with 63 to 69 Gy in a similar patient population. RESULTS: The trial reached its accrual goal of 42 patients: median age, 63 years (range, 45-83 years); male, 28 (67%); smoker or former smoker, 39 (93%); stage III, 38 (90%). Median tumor dose delivered was 83 Gy (range, 63-86 Gy) in 30 daily fractions. Median follow-up for surviving patients was 47 months. The 2-year rates of infield and overall local regional tumor controls (ie, including isolated nodal failure) were 82% (95% CI, 62%-92%) and 62% (95% CI, 43%-77%), respectively. Median overall survival was 25 months (95% CI, 12-32 months). The 2-year and 5-year overall survival rates were 52% (95% CI, 36%-66%) and 30% (95% CI, 16%-45%), respectively. CONCLUSIONS AND RELEVANCE: Adapting RT-escalated radiation dose to the FDG-avid tumor detected by midtreatment PET provided a favorable local-regional tumor control. The RTOG 1106 trial is an ongoing clinical trial to validate this finding in a randomized fashion. TRIAL REGISTRATION: clinicaltrials.gov Identifier: NCT01190527.


Subject(s)
Carboplatin/administration & dosage , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/therapy , Paclitaxel/administration & dosage , Positron Emission Tomography Computed Tomography/methods , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Carboplatin/therapeutic use , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/metabolism , Chemoradiotherapy , Dose Fractionation, Radiation , Female , Fluorodeoxyglucose F18/metabolism , Humans , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/metabolism , Male , Middle Aged , Paclitaxel/therapeutic use , Precision Medicine , Radiotherapy, Conformal , Survival Analysis , Treatment Outcome
13.
Nat Commun ; 8: 14979, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28382931

ABSTRACT

It is widely accepted that dynamic and reversible tumour cell plasticity is required for metastasis, however, in vivo steps and molecular mechanisms are poorly elucidated. We demonstrate here that monocytic (mMDSC) and granulocytic (gMDSC) subsets of myeloid-derived suppressor cells infiltrate in the primary tumour and distant organs with different time kinetics and regulate spatiotemporal tumour plasticity. Using co-culture experiments and mouse transcriptome analyses in syngeneic mouse models, we provide evidence that tumour-infiltrated mMDSCs facilitate tumour cell dissemination from the primary site by inducing EMT/CSC phenotype. In contrast, pulmonary gMDSC infiltrates support the metastatic growth by reverting EMT/CSC phenotype and promoting tumour cell proliferation. Furthermore, lung-derived gMDSCs isolated from tumour-bearing animals enhance metastatic growth of already disseminated tumour cells. MDSC-induced 'metastatic gene signature' derived from murine syngeneic model predicts poor patient survival in the majority of human solid tumours. Thus spatiotemporal MDSC infiltration may have clinical implications in tumour progression.


Subject(s)
Gene Expression Profiling/methods , Granulocytes/metabolism , Monocytes/metabolism , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Proliferation/genetics , Cells, Cultured , Female , Humans , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms/metabolism , Neoplasms/pathology
14.
Mol Cancer Ther ; 14(3): 779-787, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25673823

ABSTRACT

Developmental pathways such as Notch play a pivotal role in tissue-specific stem cell self-renewal as well as in tumor development. However, the role of Notch signaling in breast cancer stem cells (CSC) remains to be determined. We utilized a lentiviral Notch reporter system to identify a subset of cells with a higher Notch activity (Notch(+)) or reduced activity (Notch(-)) in multiple breast cancer cell lines. Using in vitro and mouse xenotransplantation assays, we investigated the role of the Notch pathway in breast CSC regulation. Breast cancer cells with increased Notch activity displayed increased sphere formation as well as expression of breast CSC markers. Interestingly Notch(+) cells displayed higher Notch4 expression in both basal and luminal breast cancer cell lines. Moreover, Notch(+) cells demonstrated tumor initiation capacity at serial dilutions in mouse xenografts, whereas Notch(-) cells failed to generate tumors. γ-Secretase inhibitor (GSI), a Notch blocker but not a chemotherapeutic agent, effectively targets these Notch(+) cells in vitro and in mouse xenografts. Furthermore, elevated Notch4 and Hey1 expression in primary patient samples correlated with poor patient survival. Our study revealed a molecular mechanism for the role of Notch-mediated regulation of breast CSCs and provided a compelling rationale for CSC-targeted therapeutics.


Subject(s)
Breast Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , Receptors, Notch/metabolism , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Animals , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Heterografts , Humans , MCF-7 Cells , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Neoplastic Stem Cells/drug effects , Repressor Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
15.
Pediatr Infect Dis J ; 33 Suppl 1: S62-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24343617

ABSTRACT

BACKGROUND: Before the introduction of rotavirus vaccine in Egypt, information on the burden of disease and the circulating rotavirus genotypes is critical to monitor vaccine effectiveness. METHODS: A cohort of 348 Egyptian children was followed from birth to 2 years of age with twice-weekly home visits to detect diarrheal illness. VP7 and VP4 genes were genotyped by reverse-transcription polymerase chain reaction and DNA sequencing. RESULTS: Forty percentage of children had rotavirus-associated diarrhea at least once by their second birthday. One hundred and twelve children experienced a single rotavirus diarrheal episodes (RDE) at a median age of 9 months; while 27 infants had their second RDE at a median age of 15 months and 1 infant had 3 RDE at the age of 2, 16 and 22 months. Of the 169 RDE, 82% could be assigned a G-type, while 58% had been identified a P-type. The most prevalent genotype was G2 (32%), followed by G1 (24%) and G9 (19%). G2P[4] rotavirus episodes were significantly associated with fever (P = 0.03) and vomiting (P = 0.06) when compared with other genotypes. G2 strains were the predominant genotype causing 50% of the second RDE while G9 represented 25% of the second RDE. CONCLUSIONS: Genotypes identified are similar to those detected globally except for absence of G4. Our finding that 75% of the second RDE were due to G2 and G9 indicates a possible reduction in natural protection afforded by these types compared with G1, where 90% of G1 cases did not experience a second xposure, indicating greater protection against recurrent symptomatic infection.


Subject(s)
Diarrhea/virology , Rotavirus Infections/virology , Rotavirus/genetics , Cohort Studies , Diarrhea/epidemiology , Egypt/epidemiology , Feces/virology , Humans , Infant , Rotavirus Infections/epidemiology , Seasons
16.
J Thorac Oncol ; 9(1): 7-17, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24346089

ABSTRACT

Cancer research has received a fresh impetus from the concept of cancer stem cell (CSC) which postulates the existence of a tumor cell population uniquely endowed with self-renewal capacity and therapy resistance. Despite recent progresses including targeted therapy, lung cancer treatment remains a challenge owing largely to disease recurrence. Providing a conceptual model of tumor resistance and disease relapse, the lung CSC has received extensive attention, leading to a flourishing literature and several ongoing clinical trials. In this study, we will discuss the data suggesting the existence of CSC in lung tumors and the potential clinical utility of CSCs as prognostic markers or cellular targets of new therapeutic strategies. We will also touch on the new fundamental developments of the CSC concept that ought to be considered if the integration of the CSC concept into clinical practice is to be successful and impact on lung cancer treatment.


Subject(s)
Lung Neoplasms/pathology , Neoplastic Stem Cells/drug effects , AC133 Antigen , Antigens, CD/analysis , Antineoplastic Agents/pharmacology , Biomarkers, Tumor , Drug Resistance, Neoplasm , Glycoproteins/analysis , Humans , Lung Neoplasms/drug therapy , Peptides/analysis
17.
Clin Cancer Res ; 19(8): 1972-80, 2013 Apr 15.
Article in English | MEDLINE | ID: mdl-23444212

ABSTRACT

PURPOSE: The cancer stem cell theory postulates that tumors contain a subset of cells with stem cell properties of self-renewal, differentiation, and tumor initiation. The purpose of this study is to determine the role of Notch activity in identifying lung cancer stem cells. EXPERIMENTAL DESIGN: We investigated the role of Notch activity in lung adenocarcinoma using a Notch GFP reporter construct and a γ-secretase inhibitor (GSI), which inhibits Notch pathway activity. RESULTS: Transduction of lung cancer cells with Notch GFP reporter construct identified a subset of cells with high Notch activity (GFP-bright). GFP-bright cells had the ability to form more tumor spheres in serum-free media and were able to generate both GFP-bright and GFP-dim (lower Notch activity) cell populations. GFP-bright cells were resistant to chemotherapy and were tumorigenic in serial xenotransplantation assays. Tumor xenografts of mice treated with GSI had decreased expression of downstream effectors of Notch pathway and failed to regenerate tumors upon reimplantation in NOD/SCID mice. Using multivariate analysis, we detected a statistically significant correlation between poor clinical outcome and Notch activity (reflected in increased Notch ligand expression or decreased expression of the negative modulators), in a group of 443 patients with lung adenocarcinoma. This correlation was further confirmed in an independent group of 89 patients with adenocarcinoma in which Hes-1 overexpression correlated with poor overall survival. CONCLUSIONS: Notch activity can identify lung cancer stem cell-like population and its inhibition may be an appropriate target for treating lung adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Lung Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Receptors, Notch/genetics , Signal Transduction/genetics , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Animals , Apoptosis/drug effects , Apoptosis/genetics , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Line, Tumor , Cyclic S-Oxides/pharmacology , Flow Cytometry , Gene Expression Regulation, Neoplastic , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Mice, Inbred NOD , Mice, Nude , Mice, SCID , Multivariate Analysis , Neoplastic Stem Cells/drug effects , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , Receptors, Notch/antagonists & inhibitors , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Thiadiazoles/pharmacology , Transcription Factor HES-1 , Xenograft Model Antitumor Assays
18.
Mol Cell ; 47(4): 570-84, 2012 Aug 24.
Article in English | MEDLINE | ID: mdl-22819326

ABSTRACT

Although inactivation of the PTEN gene has been implicated in the development of resistance to the HER2 targeting antibody trastuzumab, the mechanisms mediating this resistance remain elusive. We generated trastuzumab resistant cells by knocking down PTEN expression in HER2 overexpressing breast cancer cell lines and demonstrate that development of trastuzumab resistance in these cells is mediated by activation of an IL6 inflammatory feedback loop leading to expansion of the cancer stem cell (CSC) population. Long term trastuzumab treatment generates highly enriched CSCs which display an EMT phenotype secreting over 100-fold more IL6 than parental cells. An IL6 receptor antibody interrupted this inflammatory feedback loop reducing the cancer stem cell population resulting in decreased tumor growth and metastasis in mouse xenographs. These studies demonstrate that trastuzumab resistance may be mediated by an IL6 inflammatory loop and suggest that blocking this loop may provide alternative strategy to overcome trastuzumab resistance.


Subject(s)
Antibodies, Monoclonal, Humanized/pharmacology , Breast Neoplasms/metabolism , Inflammation/metabolism , Interleukin-6/metabolism , Neoplastic Stem Cells/metabolism , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Down-Regulation/genetics , Drug Resistance, Neoplasm , Female , Humans , Inflammation/genetics , Inflammation/pathology , Interleukin-6/genetics , Interleukin-8/genetics , Interleukin-8/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Neoplastic Stem Cells/pathology , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Receptor, ErbB-2/genetics , Trastuzumab
19.
Clin Cancer Res ; 15(20): 6386-90, 2009 Oct 15.
Article in English | MEDLINE | ID: mdl-19808871

ABSTRACT

PURPOSE: An embryonic stem cell (ESC) profile correlates with poorly differentiated breast, bladder, and glioma cancers. In this article, we assess the correlation between the ESC profile and clinical variables in lung cancer. EXPERIMENTAL DESIGN: Microarray gene expression analysis was done using Affymetrix Human Genome U133A on 443 samples of human lung adenocarcinoma and 130 samples of squamous cell carcinoma (SCC). To identify gene set enrichment patterns, we used the Genomica software. RESULTS: Our analysis showed that an increased expression of the ESC gene set and a decreased expression of the Polycomb target gene set identified poorly differentiated lung adenocarcinoma. In addition, this gene expression signature was associated with markers of poor prognosis and worse overall survival in lung adenocarcinoma. However, there was no correlation between this ESC gene signature and any histologic or clinical variable assessed in lung SCC. CONCLUSIONS: This work suggests that not all poorly differentiated non-small cell lung cancers exhibit a gene expression profile similar to that of ESC, and that other characteristics may play a more important role in the determination of differentiation and survival in SCC of the lung.


Subject(s)
Adenocarcinoma/genetics , Carcinoma, Squamous Cell/genetics , Lung Neoplasms/genetics , Adenocarcinoma/pathology , Carcinoma, Squamous Cell/pathology , Embryonic Stem Cells/pathology , Gene Expression Profiling , Humans , Lung Neoplasms/pathology , Prognosis
20.
J Neuroophthalmol ; 28(1): 27-30, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18347455

ABSTRACT

Paraneoplastic opsoclonus-myoclonus syndrome (OMS) is associated with small cell lung cancer (SCLC) in adults. Without appropriate treatment for SCLC, all reported patients with SCLC and OMS have died of complications of OMS within 3 months of diagnosis. With appropriate treatment, about half of reported patients have had improvement in neurologic function, and several have become long-term survivors (6-84 months). We report a patient with SCLC who presented with OMS and was refractory to immunosuppressive therapy but responded rapidly to antineoplastic therapy and remains alive with no sign of SCLC recurrence and minimal residual neurologic deficits 30 months after diagnosis. In patients presenting with OMS, early recognition and treatment of the underlying malignancy probably improve the chances for recovery from the OMS with minimal deficit and ultimate survival.


Subject(s)
Carcinoma, Small Cell/complications , Lung Neoplasms/complications , Lung/pathology , Opsoclonus-Myoclonus Syndrome/therapy , Paraneoplastic Syndromes, Nervous System/therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain/drug effects , Brain/immunology , Brain/physiopathology , Carcinoma, Small Cell/physiopathology , Carcinoma, Small Cell/therapy , Disease Progression , Female , Humans , Immunoglobulins, Intravenous/therapeutic use , Immunosuppressive Agents/therapeutic use , Lung/diagnostic imaging , Lung Neoplasms/physiopathology , Lung Neoplasms/therapy , Middle Aged , Opsoclonus-Myoclonus Syndrome/diagnosis , Opsoclonus-Myoclonus Syndrome/physiopathology , Paraneoplastic Syndromes, Nervous System/diagnosis , Paraneoplastic Syndromes, Nervous System/physiopathology , Radiotherapy , Steroids/therapeutic use , Survivors , Tomography, X-Ray Computed , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...