Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Sci Rep ; 13(1): 5964, 2023 04 12.
Article in English | MEDLINE | ID: mdl-37045940

ABSTRACT

Role of DNA damage and demethylation on anticancer activity of DNA methyltransferase inhibitors (DNMTi) remains undefined. We report the effects of DNMT1 gene deletion/disruption (DNMT1-/-) on anticancer activity of a class of DNMTi in vitro, in vivo and in human cancers. The gene deletion markedly attenuated cytotoxicity and growth inhibition mediated by decitabine, azacitidine and 5-aza-4'-thio-2'-deoxycytidine (aza-T-dCyd) in colon and breast cancer cells. The drugs induced DNA damage that concurred with DNMT1 inhibition, subsequent G2/M cell-cycle arrest and apoptosis, and upregulated p21 in DNMT1+/+ versus DNMT1-/- status, with aza-T-dCyd the most potent. Tumor growth and DNMT1 were significantly inhibited, and p21 was upmodulated in mice bearing HCT116 DNMT1+/+ xenograft and bladder PDX tumors. DNMT1 gene deletion occurred in ~ 9% human colon cancers and other cancer types at varying degrees. Decitabine and azacitidine demethylated CDKN2A/CDKN2B genes in DNMT1+/+ and DNMT1-/- conditions and increased histone-H3 acetylation with re-expression of p16INK4A/p15INK4B in DNMT1-/- state. Thus, DNMT1 deletion confers resistance to DNMTi, and their anti-cancer activity is determined by DNA damage effects. Patients with DNMT1 gene deletions may not respond to DNMTi treatment.


Subject(s)
Azacitidine , DNA (Cytosine-5-)-Methyltransferases , Humans , Mice , Animals , Decitabine/pharmacology , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Azacitidine/pharmacology , DNA Damage , Demethylation , DNA , DNA Methylation , Cell Line, Tumor
2.
NPJ Precis Oncol ; 6(1): 22, 2022 Apr 05.
Article in English | MEDLINE | ID: mdl-35383277

ABSTRACT

The role of lymph node involvement and tumor size in metastatic disease including breast cancer is unclear. Here, nodal metastasis and T stage on the risk of mortality were investigated in de novo metastatic breast cancer population (35812 patients) in the Surveillance, Epidemiology, and End Results (SEER) Program database in the United States. We found an association between all-cause mortality and regional node involvement (adjusted hazard ratio [HR] = 1.45, 95% confidence interval [CI] 1.36-1.55, p < 0.0001) or T stage (HR = 1.20, 95% CI 1.14-1.25, p < 0.0001), independent of known clinicopathologic measurements. Number of positive nodes, and size and chest wall involvement of the breast tumors exhibited similar significance for breast cancer-specific mortality in the population (p < 0.0001 each), and all-cause mortality in hormone receptor (HR)-positive/HER2-negative (HR+/HER2-), HR+/HER2+, HR-/HER2+ and triple-negative metastatic breast cancer subtypes. Thus, nodal involvement and T stage are independent risk factors for mortality in the population of de novo metastatic breast cancer.

3.
J Hematol Oncol ; 14(1): 83, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34039392

ABSTRACT

BACKGROUND: Challenges remain on the selection of patients who potentially respond to a class of drugs that target epigenetics for cancer treatment. This study aims to investigate TET2/DNMT3A mutations and antitumor activity of a novel epigenetic agent in multiple human cancer cell lines and animal models. METHODS: Seventeen cancer cell lines and multiple xenograft models bearing representative human solid tumors were subjected to 4'-thio-2'-deoxycytidine (T-dCyd) or control treatment. Gene mutations in cell lines were examined by whole exome and/or Sanger sequencing. Specific gene expression was measured in cells and xenograft tumor samples by Western blotting and immunohistochemistry. TET2/DNMT3A mutation status in 47,571 human tumor samples was analyzed at cBioPortal for Cancer Genomics. RESULTS: Cell survival was significantly inhibited by T-dCyd in breast BT549, lung NCI-H23, melanoma SKMEL5 and renal ACHN cancer lines harboring deleterious TET2 and nonsynonymous DNMT3A mutations compared to 13 lines without such mutation pattern (P = 0.007). The treatment upregulated p21 and induced cell cycle arrest in NCI-H23 cells, and dramatically inhibited their xenograft tumor growth versus wildtype models. T-dCyd administrations led to a significant p21 increase and near eradication of tumor cells in the double-mutant xenografts by histological evaluation. TET2/DNMT3A was co-mutated in human lung, breast, skin and kidney cancers and frequently in angioimmunoblastic and peripheral T cell lymphomas and several types of leukemia. CONCLUSIONS: Cell and animal models with concurrent mutations in TET2 and DNMT3A were sensitive to T-dCyd treatment. The mutations were detectable in human solid tumors and frequently occur in some hematological malignancies.


Subject(s)
DNA (Cytosine-5-)-Methyltransferases/genetics , DNA-Binding Proteins/genetics , Deoxycytidine/analogs & derivatives , Mutation , Neoplasms/drug therapy , Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Thionucleosides/pharmacology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methyltransferase 3A , DNA-Binding Proteins/metabolism , Deoxycytidine/pharmacology , Dioxygenases , Female , HCT116 Cells , Humans , MCF-7 Cells , Mice , Mice, Nude , Neoplasms/metabolism , Proto-Oncogene Proteins/metabolism , Xenograft Model Antitumor Assays
4.
JAMA Netw Open ; 3(7): e207213, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32644137

ABSTRACT

Importance: It is not well understood whether prognostic factors in breast cancer are affected by specific treatment and vary by clinical outcome type compared with untreated patients. Objective: To identify independent clinical and molecular measurements associated with overall survival (OS) and recurrence-free survival (RFS) by homogeneous treatment in women with breast cancer. Design, Setting, and Participants: This prognostic study included 956 patients diagnosed with invasive breast cancer from hospital centers across 4 geographical regions of the United States who participated in the accreditation program of the Commission on Cancer of the American College of Surgeons from 1985 to 1997. The duration of follow-up ranged from 1 to 282 months. The study analysis was conducted from June 10, 2019, to March 18, 2020. Main Outcomes and Measures: Analysis of OS and RFS in patients who underwent chemotherapy, radiotherapy, or endocrine therapy alone compared with no systemic or locoregional therapy. Cox proportional hazards regression models were used to estimate independent performance and 95% CI of age, tumor size, number of positive nodes (nodal status), tumor grades 2 and 3, p53 status, estrogen receptor (ER) status, and ERBB2 (formerly HER2) status. Results: Among 956 participants, median age was 61 (range, 25-96) years. Age (adjusted hazard ratio [AHR], 2.24; 95% CI, 1.27-3.94; P = .01) and high grade (AHR, 2.05; 95% CI, 1.09-3.86; P = .02), in addition to nodal status and tumor size, were independently associated with OS and RFS, respectively, in untreated patients. p53 status (AHR, 2.11; 95% CI, 1.07-4.18; P = .03) and ER status (AHR, 0.46; 95% CI, 0.23-0.92; P = .03) were associated with higher and lower risks of death, respectively, whereas nodal status (AHR, 1.13; 95% CI, 1.06-1.20; P < .005), high grade (AHR, 4.01; 95% CI, 1.51-10.70; P = .01), and ERBB2 positivity (AHR, 2.67; 95% CI, 1.25-5.70; P = .01) were associated with the risk of recurrence after endocrine therapy. Tumor size (AHR for OS, 2.76 [95% CI, 1.79-4.31; P < .005]; AHR for RFS, 2.27 [95% CI, 1.23-4.18; P = .01]) and ERBB2 status (AHR for OS, 5.35 [95% CI, 1.31-21.98; P = .02]; AHR for RFS, 6.05 [95% CI, 1.48-24.78; P = .01]) were independently associated with radiotherapy outcomes, and nodal status was significantly associated with chemotherapy outcomes (AHR for OS, 1.06 [95% CI, 1.02-1.09; P < .005]; AHR for RFS, 1.05 [95% CI, 1.01-1.09; P = .01]). Conclusions and Relevance: In this study, independent prognostic factors were associated with specific treatment and weighted by the outcome category with reference to untreated patients within biological and clinical contexts.


Subject(s)
Breast Neoplasms , Neoplasm Recurrence, Local/diagnosis , Patient Care Management/methods , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Disease-Free Survival , Female , Humans , Kaplan-Meier Estimate , Middle Aged , Neoplasm Staging , Outcome Assessment, Health Care , Prognosis , Tumor Burden
5.
Clin Transl Gastroenterol ; 10(10): e00098, 2019 10.
Article in English | MEDLINE | ID: mdl-31663905

ABSTRACT

OBJECTIVES: Preoperative decision-making for differentiating malignant from benign lesions in the gallbladder remains challenging. We aimed to create a diagnostic nomogram to identify gallbladder cancer (GBC), especially for incidental GBC (IGBC), before surgical resection. METHODS: A total of 587 consecutive patients with pathologically confirmed gallbladder lesions from a hospital were randomly assigned to a training cohort (70%) and an internal validation cohort (30%), with 287 patients from other centers as an external validation cohort. Radiological features were developed by the least absolute shrinkage and selection operator logistic regression model. Significant radiological features and independent clinical factors, identified by multivariate analyses, were used to construct a nomogram. RESULTS: A diagnostic nomogram was established by age, CA19.9, and 6 radiological features. The values of area under the curve in the internal and external validation cohorts were up to 0.91 and 0.89, respectively. The calibration curves for probability of GBC showed optimal agreement between nomogram prediction and actual observation. Compared with previous methods, it demonstrated superior sensitivity (91.5%) and accuracy (85.1%) in the diagnosis of GBC. The accuracy using the nomogram was significantly higher in GBC groups compared with that by radiologists in the training cohort (P < 0.001) and similarly in each cohort. Notably, most of the IGBC, which were misdiagnosed as benign lesions, were successfully identified using this nomogram. DISCUSSION: A novel nomogram provides a powerful tool for detecting the presence of cancer in gallbladder masses, with an increase in accuracy and sensitivity. It demonstrates an unprecedented potential for IGBC identification.


Subject(s)
CA-19-9 Antigen/blood , Early Detection of Cancer/methods , Gallbladder Neoplasms/diagnosis , Gallbladder/diagnostic imaging , Nomograms , Adult , Aged , Aged, 80 and over , Cystectomy , Female , Gallbladder/pathology , Gallbladder Neoplasms/blood , Gallbladder Neoplasms/pathology , Humans , Male , Middle Aged , Multivariate Analysis , Preoperative Period , ROC Curve , Retrospective Studies , Tomography, X-Ray Computed , Young Adult
6.
Breast Cancer Res Treat ; 175(2): 287-295, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30746635

ABSTRACT

BACKGROUND: It remains unclear whether breast cancer subtypes are associated with clinical outcome in patients without any treatment including systemic and radiation therapy as an independent entity. Understanding the survival profiles among subtypes by treatment status could impact optimal selection of treatments. METHODS: Patients were diagnosed with invasive breast cancer from the community hospitals across four geographical regions of the United States. Expression of hormone receptor (HR) and HER2 in tumor specimens from 1169 patients was centrally determined by immunohistochemistry and fluorescence in situ; breast cancer was classified into HR+/HER2-, HR+/HER2+, triple-negative, and HER2+ subtypes. Overall survival (OS) at a median follow-up of about 15 years among subtypes in untreated patients and those with systemic treatments and radiotherapy was analyzed by Kaplan-Meier method and multivariable analysis adjusting for age, tumor size and grade, number of positive nodes, stage and breast cancer subtypes. RESULTS: Without treatment, breast cancer subtypes were not associated with OS (P = 0.983) and remained insignificant for prognosis by multivariable analysis after adjusting for confounders. This contrasted with a significant survival difference across the subtypes in patients with conventional therapies (P < 0.0001). Compared with HR+/HER2- subtype, triple-negative subtype (HR 1.5, 95% CI 1.11-2.04; P = 0.009) and HER2+ subtype (HR 2.18, 95% CI 1.48-3.28; P = 0.0001) were significantly associated with worse survival by multivariable analyses. CONCLUSION: Breast cancer subtypes are not associated with survival in untreated patient population and, in contrast, significantly associated with prognosis in patients with conventional therapy. The data provide evidence of treatment-associated differential outcomes among breast cancer subtypes.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/therapy , Prognosis , Triple Negative Breast Neoplasms/therapy , Adult , Aged , Breast Neoplasms/classification , Breast Neoplasms/genetics , Chemoradiotherapy, Adjuvant/adverse effects , Chemotherapy, Adjuvant/adverse effects , Disease-Free Survival , Female , Follow-Up Studies , Humans , Middle Aged , Receptor, ErbB-2/genetics , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Trastuzumab/therapeutic use , Treatment Outcome , Triple Negative Breast Neoplasms/classification , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
7.
Skinmed ; 16(1): 71-72, 2018.
Article in English | MEDLINE | ID: mdl-29551121

ABSTRACT

An otherwise healthy man in his 50s presented complaining of pruritic lesions on the left side of his scalp. The lesions had slowly been growing in size over the preceding 30 years. They would occasionally bleed, and this is what ultimately prompted him to seek medical advice. Physical examination revealed multiple aggregated and soft, flesh-colored nodules on the left posterior auricular area of the scalp (Figure 1). No appreciable clinical lymphadenopathy was identified on examination. A shave biopsy of one of the nodules was performed for diagnostic clarification.


Subject(s)
Angiolymphoid Hyperplasia with Eosinophilia/pathology , Angiolymphoid Hyperplasia with Eosinophilia/therapy , Scalp/pathology , Biopsy , Humans , Male , Middle Aged , Skin/pathology
8.
Theranostics ; 7(4): 945-951, 2017.
Article in English | MEDLINE | ID: mdl-28382166

ABSTRACT

γH2AX plays a role in DNA damage response signaling and facilitates the repair of DNA double strand breaks. However, it remains unknown whether constitutive tumor γH2AX expression is associated with treatment outcome in patients. γH2AX status was detected in primary tumors from 24% of 826 patients with stage I, II and III breast cancer by immunohistochemistry; overall survival was analyzed by Kaplan-Meier method. At median follow-up of 176 months (range 13 - 282 months), we found substantial survival heterogeneity in γH2AX-positive patients (P=0.002) among uniform treatment groups including radiation or endocrine therapy alone and no-treatment, as well as chemotherapy alone (being worst), in contrast to γH2AX-negative patients (P=0.2). In the chemotherapy group (n=118), median survival was 63 months (95% confidence interval [CI], 29 - 83) in patients with γH2AX-positive tumors compared with 170 months (95% CI 94 - 235) in those with γH2AX-negative tumors (P=0.0017). γH2AX remained a poor prognosis factor in the group by multivariable analysis (adjusted hazard ratio 2.12, P=0.009). Our data demonstrate that constitutive γH2AX positivity is significantly associated with survival heterogeneity in patients among uniform treatment groups, and its expression at diagnosis independently predicts poor chemotherapy outcome in breast cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/diagnosis , Breast Neoplasms/drug therapy , Drug Therapy/methods , Histones/analysis , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Female , Humans , Immunohistochemistry , Prognosis , Survival Analysis , Treatment Outcome
9.
Expert Opin Drug Discov ; 11(11): 1071-1080, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27626707

ABSTRACT

INTRODUCTION: Cancer stem cells (CSCs), also known as tumor initialing cells, have self-renewal capacity and are believed to play an important role in residual disease or tumor relapse. CSCs exhibit characteristic slow growth rate and are resistant to conventional chemotherapy/radiotherapy in experimental models. The type of cells commonly employs aberrant activity of the embryonic signal transduction pathways - Notch, Hedgehog (Hh), and Wnt - for uncontrolled proliferation and survival. Areas covered: The following article discusses key genetic and molecular alterations in Notch, Hh and Wnt pathways and drugs targeting the alterations for the treatment of leukemia and lymphoma. Expert opinion: Early signs of signal agent activity have been observed in certain types of leukemia and lymphoma with experimental therapeutics targeting the embryonic pathways in the CSC signaling network. However, clinical development of agents that inhibit the Wnt/ß-catenin, Notch and Hh signaling appear to be more complex in relapsed or refractory malignancies. A strategy to effectively target signaling may rely on early application of biomarkers representative of the active signaling nodes companion to the molecularly targeted agents. Biomarkers for efficacy could potentially guide selective treatment of hematological malignancies or cancer with drugs that target the embryonic pathways.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia/drug therapy , Lymphoma/drug therapy , Animals , Drug Design , Hedgehog Proteins/antagonists & inhibitors , Humans , Leukemia/pathology , Lymphoma/pathology , Molecular Targeted Therapy , Neoplastic Stem Cells/metabolism , Receptors, Notch/antagonists & inhibitors , Signal Transduction/drug effects , Wnt Signaling Pathway/drug effects
10.
Cancer Treat Rev ; 45: 87-96, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26995633

ABSTRACT

PI3K/AKT signaling pathway plays an important role in tumorigenesis and regulates critical cellular functions including survival, proliferation and metabolism. PIK3CA mutations and AKT activation by phosphorylation (pAKT) are often detected in many cancers and especially at high frequencies in breast cancer. Mounting data suggest that PIK3CA mutations or pAKT are mostly associated with better or insignificant outcomes in estrogen receptor-positive (ER+) early stage breast cancer and tend to be with worse prognosis in ER- disease. pAKT expression has been identified to predict paclitaxel chemotherapy benefit in node-positive breast cancer. Preclinical and neoadjuvant trial data suggest that PIK3CA alterations confer resistance to HER2-targeted therapy and are associated with lower pathological complete response (pCR) rate in HER2-positive breast cancer. However, recent results from randomized clinical trials of adjuvant and metastatic settings show that patients with mutant and wildtype PIK3CA tumors derived similar benefit from anti-HER2 therapy. This article, with our new insights, aims to decipher the mixed data and discusses the influence of the potential confounding factors in the assessments. We also share our views for validation of PI3K/AKT alterations in relation to clinical outcome in the context of specific breast cancer subtypes and treatment modalities towards further advance of the precision medicine for breast cancer treatment.


Subject(s)
Breast Neoplasms , Oncogene Protein v-akt/genetics , Paclitaxel/pharmacology , Phosphatidylinositol 3-Kinases/genetics , Receptor, ErbB-2 , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Class I Phosphatidylinositol 3-Kinases , Female , Humans , Mutation , Neoplasm Staging , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Signal Transduction/genetics , Treatment Outcome
12.
Cancer Chemother Pharmacol ; 76(6): 1273-83, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26486517

ABSTRACT

PURPOSE: P-glycoprotein (Pgp), an ATP-dependent transport protein, confers multidrug resistance in cancer cells. Tariquidar binds and inhibits Pgp. To assess the toxicity, pharmacokinetics (PK), and pharmacodynamics of tariquidar, we conducted a phase I trial of tariquidar in combination with doxorubicin, docetaxel, or vinorelbine in children and adolescents with recurrent or refractory solid tumors. METHODS: Patients less than 19 years of age with refractory or recurrent solid tumors were eligible. Tariquidar (1, 1.5, or 2 mg/kg) was administered alone and in combination with doxorubicin, docetaxel, or vinorelbine. PK of tariquidar and cytotoxic drugs was performed. Pgp function was assessed by a rhodamine efflux assay and (99m)Tc-sestamibi scintigraphy. Tumor Pgp expression was assessed by immunohistochemistry. Response was assessed using Response Evaluation Criteria in Solid Tumors. RESULTS: Twenty-nine subjects were enrolled. No tariquidar-related dose-limiting toxicity (DLT) was observed. DLT related to cytotoxic drugs occurred in 12 % of subjects receiving tariquidar 2 mg/kg. When administered in combination with tariquidar, the clearance of docetaxel and vinorelbine was reduced compared to prior studies. Inhibition of rhodamine efflux was dose dependent. After tariquidar administration, (99m)Tc-sestamibi accumulation in tumor increased by 22 %. Objective responses (1 complete, 2 partial) were observed. There was no association between tumor Pgp expression and response. CONCLUSION: A tolerable and biologically active dose of tariquidar was established in children and adolescents. This trial demonstrates that modulators of resistance can be evaluated in combination with chemotherapy, and pharmacokinetic and pharmacodynamic endpoints can be useful in determination of recommended dose in children and adolescents.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoplasms/drug therapy , Quinolines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adolescent , Anemia/chemically induced , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Area Under Curve , Child , Child, Preschool , Docetaxel , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Drug Resistance, Neoplasm , Female , Humans , Male , Metabolic Clearance Rate , Neoplasms/metabolism , Neoplasms/pathology , Neutropenia/chemically induced , Quinolines/administration & dosage , Quinolines/adverse effects , Taxoids/administration & dosage , Taxoids/adverse effects , Treatment Outcome , Vinblastine/administration & dosage , Vinblastine/adverse effects , Vinblastine/analogs & derivatives , Vinorelbine , Vomiting/chemically induced
13.
Nat Rev Clin Oncol ; 12(8): 445-64, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25850553

ABSTRACT

During the past decade, cancer stem cells (CSCs) have been increasingly identified in many malignancies. Although the origin and plasticity of these cells remain controversial, tumour heterogeneity and the presence of small populations of cells with stem-like characteristics is established in most malignancies. CSCs display many features of embryonic or tissue stem cells, and typically demonstrate persistent activation of one or more highly conserved signal transduction pathways involved in development and tissue homeostasis, including the Notch, Hedgehog (HH), and Wnt pathways. CSCs generally have slow growth rates and are resistant to chemotherapy and/or radiotherapy. Thus, new treatment strategies targeting these pathways to control stem-cell replication, survival and differentiation are under development. Herein, we provide an update on the latest advances in the clinical development of such approaches, and discuss strategies for overcoming CSC-associated primary or acquired resistance to cancer treatment. Given the crosstalk between the different embryonic developmental signalling pathways, as well as other pathways, designing clinical trials that target CSCs with rational combinations of agents to inhibit possible compensatory escape mechanisms could be of particular importance. We also share our views on the future directions for targeting CSCs to advance the clinical development of these classes of agents.


Subject(s)
Hedgehog Proteins/antagonists & inhibitors , Neoplasms/therapy , Neoplastic Stem Cells/physiology , Receptors, Notch/antagonists & inhibitors , Wnt Signaling Pathway/drug effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/metabolism , Clinical Trials as Topic , Drug Design , Drug Resistance, Neoplasm , Humans , Molecular Targeted Therapy/methods , Molecular Targeted Therapy/trends , Neoplasms/pathology , Signal Transduction/drug effects
14.
J Hematol Oncol ; 8: 9, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25653136

ABSTRACT

BACKGROUND: Notch1 transmembrane receptor is activated through ligand-binding- triggered proteolytic cleavages and, upon release, the intracellular domain (N1-ICD) translocates into the nucleus and modulates target gene transcriptions. Notch activation has been implicated in tumorigenesis in an increasing number of human malignancies including non-small cell lung cancer (NSCLC). However, Notch1 in distinct expression patterns and activation status with tumor progression remains to be defined in NSCLC. METHODS: Notch1 and activated Notch1, N1-ICD, were examined by immunohistochemistry in 58 cases of stage I to IV NSCLC tumors. Association between Notch1 or N1-ICD expression and clinicopathological factors was assessed via correlation coefficient r statistics. P-values are two-sided. RESULTS: Detectable tumor Notch1, predominantly localized to the membrane and cytoplasm, was observed in 29 cases (50%, 95% Blyth-Still-Casella confidence interval 37 - 63%). It was negatively associated with stage (r=- 0.43, P<0.001) and nodal status (r=- 0.33, P = 0.01), but not tumor size. In contrast, nuclear N1-ICD expression level was low and found in 12% of NSCLC patients, neither significantly associated with stage nor nodal status. Upon Notch1 activation in vitro, a mostly extra-nuclear staining was substantially turned into the nuclear signal in cancer cells. CONCLUSIONS: Notch1 in the largely inactivated phenotype is inversely associated with clinical stage progression in NSCLC. Notch1, rather than activated N1-ICD, may be a context-dependent restrictive factor to nodal metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Receptor, Notch1/metabolism , Blotting, Western , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Disease Progression , Female , Humans , Immunohistochemistry , Lung Neoplasms/metabolism , Male , Middle Aged , Neoplasm Staging , Phenotype , Tissue Array Analysis
15.
J Clin Endocrinol Metab ; 99(4): 1291-7, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24423320

ABSTRACT

CONTEXT: Adrenocortical carcinoma (ACC) is a rare malignancy with a poor prognosis in need of more effective treatment options. Published evidence indicates many ACCs express the vascular endothelial growth factor receptor (VEGFR), suggesting inhibiting vascular endothelial growth factor signaling could potentially impact tumor growth. OBJECTIVE: The objective of the study was to determine the antitumor efficacy of axitinib (AG-013736), a potent, selective inhibitor of VEGFR1, -2, and -3. DESIGN: This was a phase II, open-label trial using a two-stage design. PATIENTS: Thirteen patients with metastatic ACC previously treated with at least one chemotherapy regimen with or without mitotane participated in the study. INTERVENTION: Starting axitinib dose was 5 mg orally twice daily. Dose escalations were permitted if the administered dose was tolerable. RESULTS: Thirteen patients were enrolled. Dose escalation was possible in seven patients, but the majority could not tolerate a dose higher than the starting 5 mg, twice-daily dose for prolonged periods of time. All patients experienced known grade 1/2 toxicities, and 10 of 13 patients had at least one grade 3/4 adverse event. No patient tumor could be scored as a Response Evaluation Criteria in Solid Tumors response, although the growth rate on therapy compared with that prior to starting axitinib was reduced in 4 of the 13 patients. The median progression-free survival was 5.48 months, and the median overall survival was longer than 13.7 months. CONCLUSION: Axitinib has limited effectiveness in ACC. Together with 48 patients previously reported who received either sorafenib or sunitinib, a total of 61 ACC patients have now been treated with a VEGFR tyrosine kinase inhibitor without an objective Response Evaluation Criteria in Solid Tumors response. Future trials in ACC should look to other targets for possible active agents.


Subject(s)
Adrenal Cortex Neoplasms/drug therapy , Adrenocortical Carcinoma/drug therapy , Angiogenesis Inhibitors/therapeutic use , Imidazoles/therapeutic use , Indazoles/therapeutic use , Adrenal Cortex Neoplasms/mortality , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/mortality , Adrenocortical Carcinoma/pathology , Axitinib , Dose-Response Relationship, Drug , Drug Administration Schedule , Humans , Receptors, Vascular Endothelial Growth Factor/metabolism , Survival Analysis , Treatment Outcome , Tumor Burden/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors
16.
Pharmacol Ther ; 141(2): 140-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24076266

ABSTRACT

Notch signaling plays an important role in development and cell fate determination, and it is deregulated in human hematologic malignancies and solid tumors. This review includes a brief introduction of the relevant pathophysiology of Notch signaling pathway and primarily focuses on the clinical development of promising agents that either obstruct Notch receptor cleavages such as γ-secretase inhibitors (GSIs) or interfere with the Notch ligand-receptor interaction by monoclonal antibodies (mAbs). Antitumor activity by GSIs and mAbs administered as single agent in early phases of clinical trials has been observed in advanced or metastatic thyroid cancer, non-small cell lung cancer, intracranial tumors, sarcoma or desmoid tumors, colorectal cancer with neuroendocrine features, melanoma and ovarian cancer. A number of mechanism-based adverse events particularly gastrointestinal toxicities emerged and mitigation strategies are developed after testing multiple GSIs and Notch targeting mAbs. We also discuss pharmacodynamic biomarkers in conjunction with methods of assessment of the molecular target inhibition validation. Biomarkers of efficacy or benefit may be of importance for a successful development of this class of drugs.


Subject(s)
Neoplasms/metabolism , Receptors, Notch/metabolism , Animals , Antineoplastic Agents/therapeutic use , Biomarkers/metabolism , Humans , Neoplasms/drug therapy , Neoplastic Stem Cells/metabolism , Neovascularization, Pathologic , Signal Transduction
17.
Clin Cancer Res ; 19(13): 3681-92, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23685835

ABSTRACT

PURPOSE: The aim of this study was to identify conserved pharmacodynamic and potential predictive biomarkers of response to anti-VEGF therapy using gene expression profiling in preclinical tumor models and in patients. EXPERIMENTAL DESIGN: Surrogate markers of VEGF inhibition [VEGF-dependent genes or VEGF-dependent vasculature (VDV)] were identified by profiling gene expression changes induced in response to VEGF blockade in preclinical tumor models and in human biopsies from patients treated with anti-VEGF monoclonal antibodies. The potential value of VDV genes as candidate predictive biomarkers was tested by correlating high or low VDV gene expression levels in pretreatment clinical samples with the subsequent clinical efficacy of bevacizumab (anti-VEGF)-containing therapy. RESULTS: We show that VDV genes, including direct and more distal VEGF downstream endothelial targets, enable detection of VEGF signaling inhibition in mouse tumor models and human tumor biopsies. Retrospective analyses of clinical trial data indicate that patients with higher VDV expression in pretreatment tumor samples exhibited improved clinical outcome when treated with bevacizumab-containing therapies. CONCLUSIONS: In this work, we identified surrogate markers (VDV genes) for in vivo VEGF signaling in tumors and showed clinical data supporting a correlation between pretreatment VEGF bioactivity and the subsequent efficacy of anti-VEGF therapy. We propose that VDV genes are candidate biomarkers with the potential to aid the selection of novel indications as well as patients likely to respond to anti-VEGF therapy. The data presented here define a diagnostic biomarker hypothesis based on translational research that warrants further evaluation in additional retrospective and prospective trials.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Antineoplastic Agents/therapeutic use , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism , Angiogenesis Inhibitors/pharmacology , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/pharmacology , Bevacizumab , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Disease Models, Animal , Drug Evaluation, Preclinical , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Neoplasms/genetics , Neoplasms/mortality , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/genetics , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism
18.
Int J Oncol ; 42(4): 1229-38, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23404210

ABSTRACT

Dual oxidase 2 (Duox2), one of the seven members of the NADPH oxidase gene family, plays a critical role in generating H2O2 for thyroid hormone biosynthesis and as an integral part of the host defense system of the respiratory epithelium and the gastrointestinal tract. Recent evidence suggests that the regulation of Duox2 expression is under the control of pro-inflammatory cytokines and that Duox2-induced reactive oxygen species (ROS) contribute to the inflammation-related tissue injury that occurs in two pre-malignant, inflammatory conditions: chronic pancreatitis and inflammatory bowel disease. Because no reliable Duox antibodies are commercially available, we report the development of a murine monoclonal antibody (MAb) to Duox2 (clone Duox S-12) and its use for the characterization of Duox2 expression in human tumors, tumor cell lines and normal tissues. Duox S-12 specifically detected both endogenously- and ectopically-expressed Duox2 protein by immunoblotting, immunofluorescence microscopy and immunohistochemistry (where both membranous and cytoplasmic staining were present). Duox2 expression detected by Duox S-12 was functionally coupled to the generation of H(2)O(2) in pancreatic cancer cells that expressed Duox2 and its cognate maturation factor DuoxA2. Although Duox S-12 recognizes ectopically expressed Duox1 protein because of the extensive amino acid homology between Duox1 and Duox2, the lack of substantial Duox1 mRNA expression in human tumors (except thyroid cancer) allowed us to evaluate Duox2 expression across a wide range of normal and malignant tissues by immuno-histochemistry. Duox2 was expressed at elevated levels in many human cancers, most notably tumors of the prostate, lung, colon and breast while brain tumors and lymphomas demonstrated the lowest frequency of expression. The Duox-specific monoclonal antibody described here provides a promising tool for the further examination of the role of Duox-dependent reactive oxygen production in inflammation-related carcinogenesis, where alterations in oxidant tone play a critical role in cell growth and proliferation.


Subject(s)
Adenocarcinoma/enzymology , Antibodies, Monoclonal, Murine-Derived/immunology , Breast Neoplasms/enzymology , NADPH Oxidases/metabolism , Pancreatic Neoplasms/enzymology , Amino Acid Sequence , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibody Specificity , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Dual Oxidases , Female , Humans , Hybridomas , Hydrogen Peroxide/metabolism , Immunohistochemistry , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Molecular Sequence Data , NADPH Oxidases/immunology , Tissue Array Analysis
19.
Clin Breast Cancer ; 12(4): 232-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22694825

ABSTRACT

BACKGROUND: Inflammatory breast cancer (IBC) is an aggressive form of breast cancer that on presentation resembles locally advanced breast cancer (LABC). This study identified molecular features of IBC and LABC to investigate pathogenesis. MATERIALS AND METHODS: This study involved 100 IBC cases identified in a national IBC registry and 107 non-IBC LABC cases from the National Cancer Institute's Cooperative Breast Cancer Tissue Resource (CBCTR). Vascular endothelial growth factor D (VEGF-D) and E-cadherin levels and lymphatic vessel density (LVD) measured by podoplanin staining were examined by immunohistochemistry on paraffin-embedded tumor specimens. Intralymphatic tumor emboli (ILTE) were assessed in IBC and non-IBC tumors. IBC cases diagnosed by clinicians but not meeting the case definitions of the American Joint Committee on Cancer (AJCC) or the Surveillance, Epidemiology and End Results (SEER) Program of the National Cancer Institute (NCI)(designated atypical IBC) were compared with AJCC- and/or SEER-defined cases (designated classic IBC). RESULTS: E-cadherin levels were significantly higher in classic IBC cases compared with non-IBC cases (P = .031), whereas compared with classic IBC, patients with non-IBC LABC had significantly higher LVD (P = .0017) and VEGF-D levels (P < .0001). ILTE was marginally greater in classic IBC than in non-IBC (P = .046). The profile of laboratory values in atypical IBC cases more closely resembled those fitting classic IBC than LABC. CONCLUSION: E-cadherin levels, LVD, VEGF-D expression, and to a lesser extent, ILTE differed between classic IBC and non-IBC LABC. The similarity of laboratory results between atypical IBC and classic IBC vs. LABC suggests the need for broadening both the AJCC and SEER case definitions for this disease.


Subject(s)
Biomarkers, Tumor/metabolism , Inflammatory Breast Neoplasms/metabolism , Inflammatory Breast Neoplasms/pathology , Lymphatic Vessels/pathology , Receptor, ErbB-2/metabolism , Vascular Endothelial Growth Factor D/metabolism , Adult , Aged , Canada , Diagnosis, Differential , Female , Humans , Lymphangiogenesis , Middle Aged , Neoplasm Staging , United States
SELECTION OF CITATIONS
SEARCH DETAIL
...