Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 21
1.
Eur J Cancer ; 202: 113975, 2024 May.
Article En | MEDLINE | ID: mdl-38442645

The first-line treatment choice of EGFRIs plus doublet chemotherapy vs. bevacizumab plus doublet chemotherapy remains a topic of interest for patients with left-sided RAS WT mCRC. We conducted a systematic literature review and meta-analysis of clinical trial data published between 2015 and 2024. We evaluated the relative efficacy and safety of first-line EGFRIs plus doublet chemotherapy (FOLFIRI or FOLFOX) vs. bevacizumab plus doublet chemotherapy for patients with RAS WT left-sided mCRC, as well as in all- and right-sided tumors. We identified eight trials with 2624 patients. Five trials reported outcomes by tumor sidedness. In the left-sided population, overall survival (OS) (Hazard Ratio (HR) = 0.80, 95% Confidence Interval (CI): 0.71-0.90) and objective response rate (ORR) (Odds ratio [OR]=1.61, 95% CI: 1.30-1.99) favored EGFRI plus chemotherapy, while no statistically significant differences were observed for progression-free survival (PFS) (HR=0.93, 95% CI: 0.84-1.04) or resection rate (RR). Similar results were found in the all-sided population. In the right-sided population, PFS favored bevacizumab plus chemotherapy (HR=1.45, 95% CI: 1.19-1.78), while no statistically significant differences were observed for OS (HR=1.17, 95% CI: 0.95-1.44), ORR (OR=0.99, 95% CI: 0.69-1.41), and RR. Early tumor shrinkage in the all-sided population favored EGFRI plus chemotherapy (OR=1.72; 95% CI: 1.36-2.17); limited data precluded evaluation by sidedness. Safety was available in 6 trials for all-sided tumors and 1 trial for left-sided tumors, each demonstrating typical class-specific adverse events. This most comprehensive meta-analysis indicates a benefit for first-line EGFRI plus chemotherapy over bevacizumab plus chemotherapy in patients with left-sided RAS WT mCRC.


Colorectal Neoplasms , Adult , Humans , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/adverse effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , ErbB Receptors
2.
Oncologist ; 28(11): e981-e994, 2023 Nov 02.
Article En | MEDLINE | ID: mdl-37432264

PURPOSE: A systematic literature review was conducted to estimate the global prevalence of Kirsten rat sarcoma virus gene (KRAS) mutations, with an emphasis on the clinically significant KRAS G12C mutation, and to estimate the prognostic significance of these mutations in patients with colorectal cancer (CRC). DESIGN: Relevant English-language publications in the Embase, MEDLINE, and the Cochrane Library databases (from 2009 to 2021) and congress presentations (from 2016 to 2021) were reviewed. Eligible studies were those that reported the prevalence and clinical outcomes of the KRAS G12C mutation in patients with CRC. RESULTS: A total of 137 studies (interventional [n = 8], post hoc analyses of randomized clinical trials [n = 6], observational [n = 122], and longitudinal [n =1]) were reviewed. Sixty-eight studies reported the prevalence of KRAS mutations (KRASm) in 42 810 patients with CRC. The median global prevalence of KRASm was 38% (range, 13.3%-58.9%) and that of the KRAS G12C mutation (KRAS G12C) 3.1% (range, 0.7%-14%). Available evidence suggests that KRASm are possibly more common in tumors that develop on the right side of the colon. Limited evidence suggests a lower objective response rate and inferior disease-free/relapse-free survival in patients with KRAS G12C compared with patients with KRASwt or other KRASm. CONCLUSION: Our analysis reveals that KRAS G12C is prevalent in 3% of patients with CRC. Available evidence suggests a poor prognosis for patients with KRAS G12C. Right-sided tumors were more likely to harbor KRASm; however, their role in determining clinical outcomes needs to be investigated further.


Colorectal Neoplasms , Lung Neoplasms , Humans , Proto-Oncogene Proteins p21(ras)/genetics , Prevalence , Colorectal Neoplasms/drug therapy , Neoplasm Recurrence, Local , Mutation , Lung Neoplasms/drug therapy
3.
N Engl J Med ; 388(1): 33-43, 2023 01 05.
Article En | MEDLINE | ID: mdl-36546651

BACKGROUND: KRAS p.G12C mutation occurs in approximately 1 to 2% of pancreatic cancers. The safety and efficacy of sotorasib, a KRAS G12C inhibitor, in previously treated patients with KRAS p.G12C-mutated pancreatic cancer are unknown. METHODS: We conducted a single-group, phase 1-2 trial to assess the safety and efficacy of sotorasib treatment in patients with KRAS p.G12C-mutated pancreatic cancer who had received at least one previous systemic therapy. The primary objective of phase 1 was to assess safety and to identify the recommended dose for phase 2. In phase 2, patients received sotorasib at a dose of 960 mg orally once daily. The primary end point for phase 2 was a centrally confirmed objective response (defined as a complete or partial response). Efficacy end points were assessed in the pooled population from both phases and included objective response, duration of response, time to objective response, disease control (defined as an objective response or stable disease), progression-free survival, and overall survival. Safety was also assessed. RESULTS: The pooled population from phases 1 and 2 consisted of 38 patients, all of whom had metastatic disease at enrollment and had previously received chemotherapy. At baseline, patients had received a median of 2 lines (range, 1 to 8) of therapy previously. All 38 patients received sotorasib in the trial. A total of 8 patients had a centrally confirmed objective response (21%; 95% confidence interval [CI], 10 to 37). The median progression-free survival was 4.0 months (95% CI, 2.8 to 5.6), and the median overall survival was 6.9 months (95% CI, 5.0 to 9.1). Treatment-related adverse events of any grade were reported in 16 patients (42%); 6 patients (16%) had grade 3 adverse events. No treatment-related adverse events were fatal or led to treatment discontinuation. CONCLUSIONS: Sotorasib showed anticancer activity and had an acceptable safety profile in patients with KRAS p.G12C-mutated advanced pancreatic cancer who had received previous treatment. (Funded by Amgen and others; CodeBreaK 100 ClinicalTrials.gov number, NCT03600883.).


Immune Checkpoint Inhibitors , Pancreatic Neoplasms , Humans , Lung Neoplasms/drug therapy , Mutation , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/secondary , Proto-Oncogene Proteins p21(ras)/genetics , Pyridines , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/therapeutic use , Administration, Oral , Treatment Outcome
4.
J Clin Oncol ; 41(3): 460-471, 2023 01 20.
Article En | MEDLINE | ID: mdl-36351210

PURPOSE: Acquired resistance to anti-epidermal growth factor receptor (EGFR) inhibitor (EGFRi) therapy in colorectal cancer (CRC) has previously been explained by the model of acquiring new mutations in KRAS/NRAS/EGFR, among other MAPK-pathway members. However, this was primarily on the basis of single-agent EGFRi trials and little is known about the resistance mechanisms of EGFRi combined with effective cytotoxic chemotherapy in previously untreated patients. METHODS: We analyzed paired plasma samples from patients with RAS/BRAF/EGFR wild-type metastatic CRC enrolled in three large randomized trials evaluating EGFRi in the first line in combination with chemotherapy and as a single agent in third line. The mutational signature of the alterations acquired with therapy was evaluated. CRC cell lines with resistance to cetuximab, infusional fluorouracil, leucovorin, and oxaliplatin, and SN38 were developed, and transcriptional changes profiled. RESULTS: Patients whose tumors were treated with and responded to EGFRi alone were more likely to develop acquired mutations (46%) compared with those treated in combination with cytotoxic chemotherapy (9%). Furthermore, contrary to the generally accepted hypothesis of the clonal evolution of acquired resistance, we demonstrate that baseline resistant subclonal mutations rarely expanded to become clonal at progression, and most remained subclonal or disappeared. Consistent with this clinical finding, preclinical models with acquired resistance to either cetuximab or chemotherapy were cross-resistant to the alternate agents, with transcriptomic profiles consistent with epithelial-to-mesenchymal transition. By contrast, commonly acquired resistance alterations in the MAPK pathway do not affect sensitivity to cytotoxic chemotherapy. CONCLUSION: These findings support a model of resistance whereby transcriptomic mechanisms of resistance predominate in the presence of active cytotoxic chemotherapy combined with EGFRi, with a greater predominance of acquired MAPK mutations after single-agent EGFRi. The proposed model has implications for prospective studies evaluating EGFRi rechallenge strategies guided by acquired MAPK mutations, and highlights the need to address transcriptional mechanisms of resistance.


Colorectal Neoplasms , ErbB Receptors , Humans , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cetuximab/pharmacology , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , ErbB Receptors/antagonists & inhibitors , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Mutation , Prospective Studies , Proto-Oncogene Proteins B-raf/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Drug Resistance, Neoplasm
5.
Oncologist ; 27(8): 663-674, 2022 08 05.
Article En | MEDLINE | ID: mdl-35472176

BACKGROUND: The KRAS p.G12C mutation has recently become an actionable drug target. To further understand KRAS p.G12C disease, we describe clinicopathologic characteristics, treatment patterns, overall survival (OS), and real-world progression-free survival (rwPFS) in patients with metastatic colorectal cancer (mCRC), KRAS p.G12C mutations (KRAS G12C), and other KRAS mutations (KRAS non-G12C) using a de-identified database. PATIENTS AND METHODS: Clinical and tumor characteristics, including treatments received, genomic profile, and clinical outcomes were assessed for patients from a US clinical genomic database with mCRC diagnosed between January 1, 2011, and March 31, 2020, with genomic sequencing data available. RESULTS: Of 6477 patients with mCRC (mCRC cohort), 238 (3.7%) had KRAS G12C and 2947 (45.5%) had KRAS non-G12C mutations. Treatment patterns were generally comparable across lines of therapy (LOT) in KRAS G12C versus KRAS non-G12C cohorts. Median (95% CI) OS after the first LOT was 16.1 (13.0-19.0) months for the KRAS G12C cohort versus 18.3 (17.2-19.3) months for the KRAS non-G12C cohort, and 19.2 (18.5-19.8) months for the mCRC overall cohort; median (95% CI) rwPFS was 7.4 (6.3-9.5), 9.0 (8.2-9.7), and 9.2 (8.6-9.7) months, respectively. The different KRAS non-G12C mutations examined did not affect clinical outcomes. Median OS and rwPFS for all cohorts declined with each subsequent LOT. CONCLUSIONS: Patients with KRAS p.G12C-mutant mCRC have poor treatment outcomes, and outcomes appear numerically worse than for those without this mutation, indicating potential prognostic implications for KRAS p.G12C mutations and an unmet medical need in this population.


Colonic Neoplasms , Colorectal Neoplasms , Rectal Neoplasms , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Humans , Mutation , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Treatment Outcome
6.
J Bone Oncol ; 34: 100423, 2022 Jun.
Article En | MEDLINE | ID: mdl-35378840

Background: Clinical practice guidelines recommend the use of bone-targeting agents for preventing skeletal-related events (SREs) among patients with bone metastases from solid tumors. The anti-RANKL monoclonal antibody denosumab is approved for the prevention of SREs in patients with bone metastases from solid tumors. However, real-world data are lacking on the impact of individual risk factors for SREs, specifically in the context of denosumab discontinuation. Purpose: We aim to identify risk factors associated with SRE incidence following denosumab discontinuation using a machine learning approach to help profile patients at a higher risk of developing SREs following discontinuation of denosumab treatment. Methods: Using the Optum PanTher Electronic Health Record repository, patients diagnosed with incident bone metastases from primary solid tumors between January 1, 2007, and September 1, 2019, were evaluated for inclusion in the study. Eligible patients received ≥ 2 consecutive 120 mg denosumab doses on a 4-week (± 14 days) schedule with a minimum follow-up of ≥ 1 year after the last denosumab dose, or an SRE occurring between days 84 and 365 after denosumab discontinuation. Extreme gradient boosting was used to develop an SRE risk prediction model evaluated on a test dataset. Multiple variables associated with patient demographics, comorbidities, laboratory values, treatments, and denosumab exposures were examined as potential factors for SRE risk using Shapley Additive Explanations (SHAP). Univariate analyses on risk factors with the highest importance from pooled and tumor-specific models were also conducted. Results: A total of 1,414 adult cancer patients (breast: 40%, prostate: 30%, lung: 13%, other: 17%) were eligible, of whom 1,133 (80%) were assigned to model training and 281 (20%) to model evaluation. The median age at inclusion was 67 (range, 19-89) years with a median duration of denosumab treatment of 253 (range, 88-2,726) days; 490 (35%) patients experienced ≥ 1 SRE 83 days after denosumab discontinuation. Meaningful model performance was evaluated by an area under the receiver operating curve score of 77% and an F1 score of 62%; model precision was 60%, with 63% sensitivity and 78% specificity. SHAP identified several significant factors for the tumor-agnostic and tumor-specific models that predicted an increased SRE risk following denosumab discontinuation, including prior SREs, shorter denosumab treatment duration, ≥ 4 clinic visits per month with at least one hospitalization (all-cause) event from the baseline period up to discontinuation of denosumab, younger age at bone metastasis, shorter time to denosumab initiation from bone metastasis, and prostate cancer. Conclusion: This analysis showed a higher cumulative number of SREs, prior SREs relative to denosumab initiation, a higher number of hospital visits, and a shorter denosumab treatment duration as significant factors that are associated with an increased SRE risk after discontinuation of denosumab, in both the tumor-agnostic and tumor-specific models. Our machine learning approach to SRE risk factor identification reinforces treatment guidance on the persistent use of denosumab and has the potential to help clinicians better assess a patient's need to continue denosumab treatment and improve patient outcomes.

7.
Support Care Cancer ; 29(11): 6731-6740, 2021 Nov.
Article En | MEDLINE | ID: mdl-33973081

PURPOSE: To understand the extent to which metastatic colorectal cancer (mCRC) patients receive education on the prevention and management associated with skin rash following Vectibix treatment. Furthermore, to investigate how this adverse event affects a patient's quality of life (QoL) and influences their treatment decisions. METHODS: A cross-sectional survey was administered to 200 mCRC patients (100 Vectibix users and 100 Vectibix non-users). After excluding respondents who had used cetuximab, 61 Vectibix users and 56 Vectibix non-users remained. RESULTS: Most Vectibix users (79%) experienced a skin rash in response to treatment of which 65% considered the rash moderate, 27% mild, and 8% severe. Vectibix users generally felt they were adequately informed about the rash (83%), with the most common messages received related to sun protection. However, sunscreen was used by only 42% of patients prior to rash and 60% of patients following the appearance of rash. The use of oral antibiotics was low prior to rash (21%) and following rash (46%). Among patients experiencing a rash within the past week (n=16), 75% reported the rash had a large negative impact on their QoL based on the Dermatology Life Quality Index. CONCLUSION: There was a disconnect between patients feeling they were adequately informed and use of prevention and management strategies such as sun protection. This suggests a gap in patient education and adoption currently exists on management strategies both prior to and following the appearance of rash. Given the negative impact that skin toxicity has on the patient's quality of life, it is essential that patients receive and subsequently utilize all information that can minimize rash severity.


Antineoplastic Agents , Colorectal Neoplasms , Exanthema/chemically induced , Panitumumab , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Cetuximab , Colorectal Neoplasms/drug therapy , Cross-Sectional Studies , ErbB Receptors/antagonists & inhibitors , Humans , Panitumumab/adverse effects , Panitumumab/therapeutic use , Quality of Life , Surveys and Questionnaires , United States
8.
Future Oncol ; 17(13): 1653-1663, 2021 May.
Article En | MEDLINE | ID: mdl-33629919

Background: Data on RAS testing practices prior to metastatic colorectal cancer (mCRC) treatment initiation are lacking in the USA. Materials & methods: Flatiron data were utilized for patients diagnosed with mCRC between 2011 and 2017. Flatiron is a longitudinal, demographically and geographically diverse database representing data from over 1.5 million active US patients treated at 255 community and hospital-affiliated oncology clinics. Results: Among 17,387 mCRC patients 69% were RAS tested and 31% were never tested. Timing of RAS testing was as follows: 23% were tested at the time of their initial CRC diagnosis, 60% following mCRC diagnosis but prior to first line of treatment, 3% prior to third line, the remaining 14% were tested following third line. Conclusion: A third (31%) of patients failed to receive RAS testing, therefore all treatment options were unavailable to them. These data highlight how universal testing has not been achieved.


Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colorectal Neoplasms/drug therapy , Genetic Testing/statistics & numerical data , Liver Neoplasms/drug therapy , Precision Medicine/statistics & numerical data , Adult , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , DNA Mutational Analysis/statistics & numerical data , Databases, Factual/statistics & numerical data , Female , GTP Phosphohydrolases/genetics , Humans , Liver Neoplasms/genetics , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Longitudinal Studies , Male , Membrane Proteins/genetics , Middle Aged , Molecular Targeted Therapy/methods , Neoplasm Staging , Precision Medicine/methods , Progression-Free Survival , Proto-Oncogene Proteins p21(ras)/genetics , Retrospective Studies , United States/epidemiology
9.
J Oral Pathol Med ; 36(1): 35-42, 2007 Jan.
Article En | MEDLINE | ID: mdl-17181740

BACKGROUND: Collagen XVIII is a ubiquitous basement membrane (BM) component and a precursor of endostatin. METHODS: Using immunohistochemistry and in situ hybridization, we studied the expression and localization of collagen XVIII in different stages of normal oral wound healing, epithelial dysplasia and squamous cell carcinoma (SCC). RESULTS: In mild epithelial dysplasias collagen XVIII appeared as a continuous signal in the BM, whereas in severe epithelial dysplasias and in the invasive areas of oral SCCs collagen XVIII was absent. In situ hybridization showed that collagen XVIII mRNA expression did not decrease in severe dysplasia or oral carcinoma samples when compared with the mild dysplasias. CONCLUSIONS: The results indicate that the absence of collagen XVIII protein in severe oral dysplasias is related to the processing of the protein rather than to changes in mRNA expression.


Carcinoma, Squamous Cell/pathology , Collagen Type XVIII/analysis , Mouth Mucosa/pathology , Mouth Neoplasms/pathology , Precancerous Conditions/pathology , Angiogenesis Inhibitors/analysis , Basement Membrane/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/pathology , Endostatins/analysis , Humans , Immunohistochemistry , In Situ Hybridization , Matrix Metalloproteinase 9/analysis , Neoplasm Invasiveness , Protein Modification, Translational , RNA, Messenger/analysis , Wound Healing/physiology
10.
Hum Mol Genet ; 15(19): 2923-35, 2006 Oct 01.
Article En | MEDLINE | ID: mdl-16926187

The neuropeptide S (NPS)-NPS receptor 1 (NPSR1) pathway has recently been implicated in the pathogenesis of asthma. The purpose of this study was to identify downstream gene targets regulated by NPSR1 upon NPS stimulation. A total of 104 genes were found significantly up-regulated and 42 down-regulated by microarray analysis 6 h after NPS administration. By Gene Ontology enrichment analysis, the categories 'cell proliferation', 'morphogenesis' and 'immune response' were among the most altered. A TMM microarray database comparison suggested a common co-regulated pathway, which includes JUN/FOS oncogene homologs, early growth response genes, nuclear receptor subfamily 4 members and dual specificity phosphatases. The expression of four up-regulated genes, matrix metallopeptidase 10 (MMP10), INHBA (activin A), interleukin 8 (IL8) and EPH receptor A2 (EPHA2), exhibited a significant NPS dose-response relationship as confirmed by quantitative reverse-transcriptase-PCR and for MMP10 by immunoassay. Immunohistochemical analyses revealed that MMP10 and TIMP metallopeptidase inhibitor 3 (TIMP3) were both strongly expressed in bronchial epithelium, and macrophages and eosinophils expressed MMP10 in asthmatic sputum samples. Because remodeling of airway epithelium is a feature of chronic asthma, the up-regulation of MMP10 and TIMP3 by NPS-NPSR1 signaling may be of relevance in the pathogenesis of asthma.


Neuropeptides/genetics , Neuropeptides/metabolism , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Apoptosis , Asthma/etiology , Asthma/genetics , Asthma/metabolism , Base Sequence , Bronchi/metabolism , Cell Line , Cell Proliferation , DNA Primers/genetics , Databases, Genetic , Humans , Matrix Metalloproteinase 10 , Metalloendopeptidases/genetics , Metalloendopeptidases/metabolism , Oligonucleotide Array Sequence Analysis , Signal Transduction , Tissue Inhibitor of Metalloproteinase-3/genetics , Tissue Inhibitor of Metalloproteinase-3/metabolism
11.
Ann Med ; 38(5): 357-66, 2006.
Article En | MEDLINE | ID: mdl-16938805

BACKGROUND: Respiratory distress syndrome (RDS) and bronchopulmonary dysplasia (BPD) have some common features with asthma. AIM: To study whether G protein-coupled receptor for asthma susceptibility (GPRA) contributes to RDS or BPD. METHODS: A haplotype association study was performed in a case-control setting of 521 Finnish infants (including 176 preterm neonates with RDS and 37 with BPD). Immunoreactivity of GPRA isoforms A and B was determined in pulmonary samples of fetuses, term infants and preterm infants with RDS or BPD. GPRA mRNA expression was determined by quantitative real-time polymerase chain reaction (PCR) in samples from nasal respiratory epithelium of adults, term infants and preterm infants. RESULTS: In infants with RDS born at 32-35 weeks of gestation, GPRA haplotype H1 was significantly underrepresented in RDS, whereas haplotype H4/H5 was associated with an increased risk. As in asthma, GPRA B isoform was induced in bronchial smooth muscle cells in RDS and BPD. In nasal respiratory epithelium, relative GPRA mRNA expression was strong in adults, weak in preterm and slightly higher in term samples. CONCLUSIONS: The results suggest that near-term RDS and asthma share the same susceptibility and protective GPRA haplotypes. Altered GPRA expression may play a role in the pathogenesis of RDS and BPD in preterm infants.


Asthma/etiology , Receptors, G-Protein-Coupled/genetics , Respiratory Distress Syndrome, Newborn/etiology , Adult , Asthma/genetics , Base Sequence , Bronchopulmonary Dysplasia/etiology , Bronchopulmonary Dysplasia/genetics , Bronchopulmonary Dysplasia/pathology , Case-Control Studies , Haplotypes , Humans , Infant, Newborn , Infant, Premature , Polymorphism, Genetic , Protein Isoforms/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Respiratory Distress Syndrome, Newborn/genetics , Respiratory Distress Syndrome, Newborn/pathology , Risk Factors
12.
Am J Hum Genet ; 79(1): 149-54, 2006 Jul.
Article En | MEDLINE | ID: mdl-16773575

We performed a genomewide scan in six multiplex families with familial idiopathic pulmonary fibrosis (IPF) who originated from southeastern Finland. The majority of the Finnish multiplex families were clustered in the region, and the population history suggested that the clustering might be explained by an ancestor shared among the patients. The genomewide scan identified five loci of interest. The hierarchical fine mapping in an extended data set with 24 families originating from the same geographic region revealed a shared 110 kb to 13 Mb haplotype on chromosome 4q31, which was significantly more frequent among the patients than in population-based controls (odds ratio 6.3; 95% CI 2.5-15.9; P = .0001). The shared haplotype harbored two functionally uncharacterized genes, ELMOD2 and LOC152586, of which only ELMOD2 was expressed in lung and showed significantly decreased messenger-RNA expression in IPF lung (n = 6) when compared with that of healthy lung (n = 7; P = .05). Our results suggest ELMOD2 as a novel candidate gene for susceptibility in familial IPF.


Cytoskeletal Proteins/genetics , Pulmonary Fibrosis/genetics , Chromosomes, Human, Pair 4 , Female , Humans , Male , Pedigree , RNA, Messenger/genetics
13.
Hum Mol Genet ; 15(10): 1667-79, 2006 May 15.
Article En | MEDLINE | ID: mdl-16600990

G protein-coupled receptor 154 (GPR154) is a recently discovered asthma susceptibility gene upregulated in the airways of asthma patients. We previously observed increased pulmonary mRNA expression of the murine ortholog Gpr154 in a mouse model of ovalbumin (OVA)-induced inflammation. However, the expression profile of GPR154 in leukocytes and the cellular functions of the receptor and its endogenous agonist neuropeptide S (NPS) have remained unidentified. Here, we characterized the mRNA expression of NPS and GPR154 by using real-time RT-PCR in fractionated human blood cells and in peripheral blood mononuclear cells (PBMCs) with monocyte or T cell activation. The expression of GPR154 in leukocytes was further confirmed by immunoblotting experiments and immunohistochemical staining of human sputum samples. Additionally, we characterized the expression of GPR154 in the lung tissue samples and in the bronchoalveolar lavage (BAL) fluid of OVA sensitized and challenged BALB/c mice. In human blood and sputum cells, monocyte/macrophages and eosinophils were identified as GPR154-positive cells. In PBMCs, monocyte activation with LPS but not T cell activation with anti-CD3/CD28 antibodies resulted in increased NPS and GPR154 expression. In the lung tissue samples and in the BAL fluid of OVA-challenged mice, GPR154 expression was upregulated in alveolar macrophages in comparison to controls. In the mouse macrophage RAW 264.7 cell line, NPS-stimulated Galphas- and Galphaq-dependent phagocytosis of Escherichia coli. The results show that GPR154 is upregulated in macrophages after antigen challenge and that NPS is capable of inducing phagocytosis of unopsonized bacteria.


Leukocytes, Mononuclear/metabolism , Macrophages/immunology , Neuropeptides/immunology , Receptors, G-Protein-Coupled/immunology , Animals , Bronchitis/immunology , Bronchoalveolar Lavage Fluid/immunology , CD28 Antigens/immunology , CD3 Complex/immunology , Cell Line , Eosinophils/immunology , Escherichia coli/immunology , Escherichia coli/physiology , Female , Humans , In Vitro Techniques , Lipopolysaccharides/toxicity , Lung/metabolism , Lymphocyte Activation , Macrophages/microbiology , Mice , Mice, Inbred BALB C , Phagocytosis/immunology , Sputum/metabolism
14.
Am J Respir Cell Mol Biol ; 33(3): 262-70, 2005 Sep.
Article En | MEDLINE | ID: mdl-15947423

We recently identified a novel positional asthma susceptibility gene, GPRA, which belongs to the G protein-coupled receptor family. In the present studies, we show that isoform specific activation of GPRA-A with its agonist, Neuropeptide S (NPS) resulted in significant inhibition of cell growth. GPRA has several variants due to extensive alternative splicing. We observed that only the full-length variants, GPRA-A and GPRA-B, with 7 transmembrane topology are transported into the plasma membrane, while the truncated proteins retain intracellular compartments. To clarify disease mechanism, we studied co-expression of the variants without finding any indication that truncated variants would inhibit the receptor transport into the plasma membrane. By using in situ hybridization and immunohistochemistry, we detected ubiquitous expression of GPRA-B, and frequent expression of GPRA-A in the epithelia of several organs including bronchi and gastrointestinal tract. Furthermore, we observed aberrant mRNA and protein expression levels of GPRA in the asthmatic bronchi. Finally, we demonstrate that GPRA and NPS are co-expressed in bronchial epithelium. In summary, this study provides evidence that GPRA might have functional relevance in modulating asthma by increased expression levels in the relevant tissues under diseased state and by potential inhibitory effect of GPRA-A activation on cell growth.


Asthma/genetics , Asthma/physiopathology , Receptors, G-Protein-Coupled/genetics , Amino Acid Sequence , Animals , Antibody Specificity , COS Cells , Cell Division/physiology , Cell Line, Tumor , Chlorocebus aethiops , Gene Expression/immunology , Humans , Immunohistochemistry , In Situ Hybridization , Lung Neoplasms , Molecular Sequence Data , Neuropeptides/genetics , RNA Splicing , Receptors, G-Protein-Coupled/immunology , Receptors, G-Protein-Coupled/metabolism , Respiratory Mucosa/cytology , Signal Transduction/physiology , Transfection
15.
Exp Cell Res ; 307(2): 292-304, 2005 Jul 15.
Article En | MEDLINE | ID: mdl-15950618

Endostatin, a potent inhibitor of endothelial cell proliferation, migration, angiogenesis and tumor growth, is proteolytically cleaved from the C-terminal noncollagenous NC1 domain of type XVIII collagen. We investigated the endostatin formation from human collagen XVIII by several MMPs in vitro. The generation of endostatin fragments differing in molecular size (24-30 kDa) and in N-terminal sequences was identified in the cases of MMP-3, -7, -9, -13 and -20. The cleavage sites were located in the protease-sensitive hinge region between the trimerization and endostatin domains of NC1. MMP-1, -2, -8 and -12 did not show any significant activity against the C-terminus of collagen XVIII. The anti-proliferative effect of the 20-kDa endostatin, three longer endostatin-containing fragments generated in vitro by distinct MMPs and the entire NC1 domain, on bFGF-stimulated human umbilical vein endothelial cells was established. The anti-migratory potential of some of these fragments was also studied. In addition, production of endostatin fragments between 24-30 kDa by human hepatoblastoma cells was shown to be due to MMP action on type XVIII collagen. Our results indicate that certain, especially cancer-related, MMP family members can generate biologically active endostatin-containing polypeptides from collagen XVIII and thus, by releasing endostatin fragments, may participate in the inhibition of endothelial cell proliferation, migration and angiogenesis.


Collagen Type XVIII/metabolism , Endostatins/metabolism , Endothelial Cells/drug effects , Matrix Metalloproteinases/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Blotting, Western , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Collagen Type I/metabolism , Collagen Type XVIII/isolation & purification , Collagenases/genetics , Collagenases/metabolism , Culture Media, Conditioned/chemistry , Endostatins/genetics , Endostatins/pharmacology , Endothelial Cells/metabolism , Gene Expression/genetics , Humans , Hydroxamic Acids/chemistry , Matrix Metalloproteinase Inhibitors , Matrix Metalloproteinases/genetics , Peptide Fragments/genetics , Peptide Fragments/pharmacology , Recombinant Proteins , Reverse Transcriptase Polymerase Chain Reaction
16.
Am J Pathol ; 166(1): 221-9, 2005 Jan.
Article En | MEDLINE | ID: mdl-15632014

Endostatin, a proteolytic fragment of type XVIII collagen, has been shown to inhibit angiogenesis, tumor growth, and endothelial cell proliferation and migration. We analyzed its functions in vivo by generating transgenic mice in which it was overexpressed in the skin and lens capsule under the keratin K14 promoter. Opacity of the lens occurred at 4 months of age in the mouse line J4, with the highest level of endostatin expression. The lens epithelial cells appeared to lose contact with the capsule and began to vacuolize. In 1-year-old mice the lens epithelial cell layer had entirely degenerated, and instead, large plaques of spindle-shaped cells had formed in the anterior region of the lens. Moreover, a widening of the epidermal basement membrane (BM) zone of the skin was observed in electron microscopy. The epidermal BM was conspicuously altered in the J4 mice with high transgene expression, including clear broadening and occurrence of pearl-like protrusions in some areas, whereas the BM was more even in appearance but consistently broadened in the mouse line G20 with moderate transgene expression. In both lines the BM was continuous. Measurements indicated that the lamina densa was 78.54 +/- 53.10 nm in line J4, the large variation reflecting the protrusions of the lamina densa, and 44.24 +/- 11.52 nm in line G20, compared with 33.74 +/- 9.96 nm in wild-type adult mice. Immunoelectron microscopy of wild-type mouse skin type XVIII collagen showed a polarized orientation in the BMs, with the C-terminal endostatin region localized in the lamina densa and the N terminus in average approximately 40 nm more on the dermal side. Type XVIII collagen was dispersed in the transgenic skin, suggesting that the transgene-derived endostatin fragment displaces the full-length collagen XVIII. This may impair the anchoring of the lamina densa to the dermis and thereby lead to loosening of the BMs, resembling the previously observed situation in collagen XVIII-null mice.


Basement Membrane/pathology , Cataract/pathology , Endostatins/physiology , Lens, Crystalline/pathology , Skin/pathology , Angiogenesis Inhibitors , Animals , Basement Membrane/ultrastructure , DNA Primers , Humans , Lens, Crystalline/ultrastructure , Mice , Mice, Transgenic , Microscopy, Immunoelectron , Polymerase Chain Reaction , Skin/ultrastructure
17.
Science ; 304(5668): 300-4, 2004 Apr 09.
Article En | MEDLINE | ID: mdl-15073379

Susceptibility to asthma depends on variation at an unknown number of genetic loci. To identify susceptibility genes on chromosome 7p, we adopted a hierarchical genotyping design, leading to the identification of a 133-kilobase risk-conferring segment containing two genes. One of these coded for an orphan G protein-coupled receptor named GPRA (G protein-coupled receptor for asthma susceptibility), which showed distinct distribution of protein isoforms between bronchial biopsies from healthy and asthmatic individuals. In three cohorts from Finland and Canada, single nucleotide polymorphism-tagged haplotypes associated with high serum immunoglobulin E or asthma. The murine ortholog of GPRA was up-regulated in a mouse model of ovalbumin-induced inflammation. Together, these data implicate GPRA in the pathogenesis of atopy and asthma.


Asthma/genetics , Chromosomes, Human, Pair 7/genetics , Genetic Predisposition to Disease , Haplotypes , Receptors, G-Protein-Coupled/genetics , Algorithms , Alternative Splicing , Animals , Asthma/metabolism , Bronchi/chemistry , Bronchi/cytology , Epithelial Cells/chemistry , Female , Finland , Gene Expression , Genes , Genetic Linkage , Genetic Variation , Genotype , Humans , Hypersensitivity/genetics , Hypersensitivity/metabolism , Immunoglobulin E/blood , Inflammation/genetics , Lung/metabolism , Male , Mice , Myocytes, Smooth Muscle/chemistry , Polymorphism, Single Nucleotide , Quebec , Receptors, G-Protein-Coupled/analysis
18.
Matrix Biol ; 22(5): 427-42, 2003 Sep.
Article En | MEDLINE | ID: mdl-14614989

Human type XVIII collagen was found to be expressed as three variants, termed NC1-303, NC1-493 and NC1-728, differing in their N-terminal non-collagenous domains (NC1). The corresponding gene was found to be approximately 105 kb in size and contain 43 exons. The short variant is derived from utilization of an upstream promoter associated with the first two exons of the gene. The two other variants are derived from a downstream promoter and alternative splicing of exon 3, resulting in 192 residues of shared sequences characterized by a putative approximately 30 residue conserved coiled-coil motif and 235 residues of sequences specific to NC1-728. The NC1-728 variant has a conserved cysteine-rich domain homologous with the ligand-binding part of the frizzled proteins. A polyclonal antibody specific to the NC1-728 variant was generated, and immunostaining of fetal tissues revealed staining in lung and skeletal muscle. Human serum contained 173- and 144-kDa alpha1(XVIII) chains corresponding to the NC1-728 and NC1-493 variants, respectively. A 200-kDa polypeptide was detected in cells transfected with a cDNA construct corresponding to the full-length NC1-728 variant, and EBNA-293 cells endogenously synthesizing low amounts of type XVIII collagen had a 45-kDa fragment in their culture medium that corresponded to most of the NC1 domain of the NC1-728 variant, suggesting processing of the N-terminal frizzled-containing domain.


Collagen Type XVIII/chemistry , Collagen Type XVIII/genetics , Alternative Splicing , Amino Acid Motifs , Amino Acid Sequence , Animals , Cell Line , Cloning, Molecular , Cysteine/chemistry , DNA, Complementary/metabolism , Exons , Genetic Variation , Humans , Immunohistochemistry , Introns , Mice , Models, Genetic , Molecular Sequence Data , Peptides/chemistry , Precipitin Tests , Promoter Regions, Genetic , Protein Structure, Tertiary , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Tissue Distribution
19.
J Biol Chem ; 278(37): 35636-43, 2003 Sep 12.
Article En | MEDLINE | ID: mdl-12832404

Bmx/Etk, a member of the Tec/Btk family of nonreceptor kinases, has recently been shown to mediate cell motility in signaling pathways that become activated upon integrin-mediated cell adhesion (Chen, R., Kim, O., Li, M., Xiong, X., Guan, J. L., Kung, H. J., Chen, H., Shimizu, Y., and Qiu, Y. (2001) Nat Cell Biol. 3, 439-444). The molecular mechanisms of Bmx-induced cell motility have so far remained unknown. Previous studies by us and others have demonstrated that a complex formation between the docking protein p130Cas (Cas) and the adapter protein Crk is instrumental in connecting several stimuli to the regulation of actin cytoskeleton and cell motility. We demonstrate here that expression of Bmx leads to an interaction between Bmx and Cas at membrane ruffles, which are sites of active actin remodeling in motile cells. Expression of Bmx also enhances tyrosine phosphorylation of Cas and Cas.Crk complex formation, and coexpression of Bmx with Cas results in an enhanced membrane ruffling and haptotactic cell migration. Importantly, a mutant form of Bmx that fails to interact with Cas also fails to induce cell migration. Furthermore, expression of a dominant-negative form of Cas that is incapable of interacting with Crk inhibits Bmx-induced membrane ruffling and cell migration. These studies suggest that Bmx-Cas interaction, phosphorylation of Cas by Bmx, and subsequent Cas.Crk complex formation functionally couple Bmx to the regulation of actin cytoskeleton and cell motility.


Actins/metabolism , Cell Movement/physiology , Phosphoproteins/metabolism , Protein-Tyrosine Kinases/metabolism , Proteins , Animals , Binding Sites , COS Cells , Cell Line , Chlorocebus aethiops , Crk-Associated Substrate Protein , Culture Media, Serum-Free , Cytoskeleton/physiology , Endothelium, Vascular/physiology , Humans , Mutagenesis, Site-Directed , Phosphoproteins/genetics , Phosphotyrosine/metabolism , Protein-Tyrosine Kinases/genetics , Recombinant Proteins/metabolism , Retinoblastoma-Like Protein p130 , Transfection
20.
J Biol Chem ; 278(15): 13069-76, 2003 Apr 11.
Article En | MEDLINE | ID: mdl-12556525

Leukocyte infiltration during inflammation is mediated by the sequential actions of adhesion molecules and chemokines. By using a rat ureteral obstruction model, we showed previously that L-selectin plays an important role in leukocyte infiltration into the kidney. Here we report the purification, identification, and characterization of an L-selectin-binding heparan sulfate proteoglycan (HSPG) expressed in the rat kidney. Partial amino acid sequencing and Western blotting analyses showed that the L-selectin-binding HSPG is collagen XVIII, a basement membrane HSPG. The binding of L-selectin to isolated collagen XVIII was specifically inhibited by an anti-L-selectin monoclonal antibody, EDTA, treatment of the collagen XVIII with heparitinase or heparin but not by chemically desulfated heparin. A cell binding assay showed that the L-selectin-collagen XVIII interaction mediates cell adhesion. Interestingly, collagen XVIII also interacted with a chemokine, monocyte chemoattractant protein-1, and presented it to a monocytic cell line, THP-1, which enhanced the alpha(4)beta(1) integrin-mediated binding of the THP-1 cells to vascular cell adhesion molecule-1. Thus, collagen XVIII may provide a link between selectin-mediated cell adhesion and chemokine-induced cellular activation and accelerate the progression of leukocyte infiltration in renal inflammation.


Basement Membrane/physiology , Chemokine CCL2/metabolism , Collagen/metabolism , L-Selectin/metabolism , Peptide Fragments/metabolism , Amino Acid Sequence , Animals , Binding Sites , Collagen Type XVIII , Edetic Acid/pharmacology , Endostatins , Humans , Kidney/metabolism , Kinetics , Polysaccharide-Lyases/metabolism , Rats , Recombinant Proteins/metabolism , Transfection
...