Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 212
Filter
Add more filters

Complementary Medicines
Publication year range
1.
J Agric Food Chem ; 72(17): 9717-9734, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38624258

ABSTRACT

Plants have a history of being employed in managing breast cancer. However, no scientific evidence supports the idea that these plants can effectively reduce the level of HER2 expression. In this study, extracts from 10 medicinal plants were evaluated for their anticancer properties against HER2-positive breast cancer cells through various methods, including the SRB assay, comet assay, annexin V-FITC dual staining, and immunoblotting. All extracts exerted antiproliferative activity against HER2-positive breast cancer cells. Furthermore, Terminalia chebula (T. chebula), Berberis aristata (B. aristata), and Mucuna pruriens (M. pruriens) reduced HER2 expression in tested cell lines. In addition, an increased Bax/Bcl-2 ratio was observed after the treatment. A comparative proteomics study showed modulation in the proteome profile of breast cancer cells after treatment with T. chebula, B. aristata, Punica granatum, M. pruriens, and Acorus calamus. Metabolic profiling of lead plants revealed the existence of multiple anticancer compounds. Our study demonstrates the considerable potential of the mentioned plants as innovative therapies for HER2-positive breast cancer.


Subject(s)
Breast Neoplasms , Cell Proliferation , Down-Regulation , Plant Extracts , Plants, Medicinal , Receptor, ErbB-2 , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Plants, Medicinal/chemistry , Female , Plant Extracts/pharmacology , Plant Extracts/chemistry , Cell Line, Tumor , Down-Regulation/drug effects , Cell Proliferation/drug effects , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/chemistry , Apoptosis/drug effects , Terminalia/chemistry , Mucuna/chemistry
2.
Environ Toxicol ; 39(6): 3389-3399, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38445457

ABSTRACT

Breast cancer stands as the predominant malignancy and primary cause of cancer-related mortality among females globally. Approximately 25% of breast cancers exhibit HER2 overexpression, imparting a more aggressive tumor phenotype and correlating with poor prognoses. Patients with metastatic breast cancer receiving HER2 tyrosine kinase inhibitors (HER2 TKIs), such as Lapatinib, develop acquired resistance within a year, posing a critical challenge in managing this disease. Here, we explore the potential of Artemisia argyi, a Chinese herbal medicine known for its anti-cancer properties, in mitigating HER2 TKI resistance in breast cancer. Analysis of the Cancer Genome Atlas (TCGA) revealed diminished expression of transmembrane serine protease 2 (TMPRSS2), a subfamily of membrane proteolytic enzymes, in breast cancer patients, correlating with unfavorable outcomes. Intriguingly, lapatinib-responsive patients exhibited higher TMPRSS2 expression. Our study unveiled that the compounds from Artemisia argyi, eriodictyol, and umbelliferone could inhibit the growth of lapatinib-resistant HER2-positive breast cancer cells. Mechanistically, they suppressed HER2 kinase activation by enhancing TMPRSS2 activity. Our findings propose TMPRSS2 as a critical determinant in lapatinib sensitivity, and Artemisia argyi emerges as a potential agent to overcome lapatinib via activating TMPRSS2 in HER2-positive breast cancer. This study not only unravels the molecular mechanisms driving cell death in HER2-positive breast cancer cells induced by Artemisia argyi but also lays the groundwork for developing novel inhibitors to enhance therapy outcomes.


Subject(s)
Artemisia , Breast Neoplasms , Drug Resistance, Neoplasm , Lapatinib , Plant Extracts , Receptor, ErbB-2 , Serine Endopeptidases , Lapatinib/pharmacology , Lapatinib/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Humans , Drug Resistance, Neoplasm/drug effects , Artemisia/chemistry , Female , Serine Endopeptidases/metabolism , Serine Endopeptidases/genetics , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Cell Line, Tumor , Plant Extracts/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
3.
Adv Sci (Weinh) ; 11(16): e2308316, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38380506

ABSTRACT

Anti-HER2 (human epidermal growth factor receptor 2) therapies significantly increase the overall survival of patients with HER2-positive breast cancer. Unfortunately, a large fraction of patients may develop primary or acquired resistance. Further, a multidrug combination used to prevent this in the clinic places a significant burden on patients. To address this issue, this work develops a nanotherapeutic platform that incorporates bimetallic gold-silver hollow nanoshells (AuAg HNSs) with exceptional near-infrared (NIR) absorption capability, the small-molecule tyrosine kinase inhibitor pyrotinib (PYR), and Herceptin (HCT). This platform realizes targeted delivery of multiple therapeutic effects, including chemo-and photothermal activities, oxidative stress, and immune response. In vitro assays reveal that the HCT-modified nanoparticles exhibit specific recognition ability and effective internalization by cells. The released PYR inhibit cell proliferation by downregulating HER2 and its associated pathways. NIR laser application induces a photothermal effect and tumor cell apoptosis, whereas an intracellular reactive oxygen species burst amplifies oxidative stress and triggers cancer cell ferroptosis. Importantly, this multimodal therapy also promotes the upregulation of genes related to TNF and NF-κB signaling pathways, enhancing immune activation and immunogenic cell death. In vivo studies confirm a significant reduction in tumor volume after treatment, substantiating the potential effectiveness of these nanocarriers.


Subject(s)
Breast Neoplasms , Gold , Hyperthermia, Induced , Receptor, ErbB-2 , Breast Neoplasms/drug therapy , Breast Neoplasms/therapy , Breast Neoplasms/metabolism , Breast Neoplasms/genetics , Female , Humans , Mice , Animals , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/genetics , Hyperthermia, Induced/methods , Gold/chemistry , Trastuzumab/pharmacology , Trastuzumab/therapeutic use , Silver/chemistry , Cell Line, Tumor , Disease Models, Animal , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Combined Modality Therapy/methods , Metal Nanoparticles/therapeutic use , Metal Nanoparticles/chemistry , Cell Proliferation/drug effects
4.
Small ; 19(50): e2303934, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37632323

ABSTRACT

Treatment failure in breast cancers overexpressing human epidermal growth factor receptor 2 (HER2) is associated mainly to the upregulation of human epidermal growth factor receptor 3 (HER3) oncoprotein linked to chemoresitence. Therefore, to increase patient survival, here a multimodal theranostic nanoplatform targeting both HER2 and HER3 is developed. This consists of doxorubicin-loaded branched gold nanoshells functionalized with the near-infrared (NIR) fluorescent dye indocyanine green, a small interfering RNA (siRNA) against HER3, and the HER2-specific antibody Transtuzumab, able to provide a combined therapeutic outcome (chemo- and photothermal activities, RNA silencing, and immune response). In vitro assays in HER2+ /HER3+ SKBR-3 breast cancer cells have shown an effective silencing of HER3 by the released siRNA and an inhibition of HER2 oncoproteins provided by Trastuzumab, along with a decrease of the serine/threonine protein kinase Akt (p-AKT) typically associated with cell survival and proliferation, which helps to overcome doxorubicin chemoresistance. Conversely, adding the NIR light therapy, an increment in p-AKT concentration is observed, although HER2/HER3 inhibitions are maintained for 72 h. Finally, in vivo studies in a tumor-bearing mice model display a significant progressively decrease of the tumor volume after nanoparticle administration and subsequent NIR light irradiation, confirming the potential efficacy of the hybrid nanocarrier.


Subject(s)
Breast Neoplasms , Nanoshells , Humans , Animals , Mice , Female , Breast Neoplasms/metabolism , Proto-Oncogene Proteins c-akt , Gold , Receptor, ErbB-2/genetics , Doxorubicin/pharmacology , Doxorubicin/therapeutic use , RNA, Small Interfering , Cell Line, Tumor
5.
Br J Cancer ; 128(2): 245-254, 2023 01.
Article in English | MEDLINE | ID: mdl-36352028

ABSTRACT

BACKGROUND: Epidermal growth factor receptor (EGFR) is a therapeutic target to which HER2/HER3 activation may contribute resistance. This Phase I/II study examined the toxicity and efficacy of high-dose pulsed AZD8931, an EGFR/HER2/HER3 inhibitor, combined with chemotherapy, in metastatic colorectal cancer (CRC). METHODS: Treatment-naive patients received 4-day pulses of AZD8931 with irinotecan/5-FU (FOLFIRI) in a Phase I/II single-arm trial. Primary endpoint for Phase I was dose limiting toxicity (DLT); for Phase II best overall response. Samples were analysed for pharmacokinetics, EGFR dimers in circulating exosomes and Comet assay quantitating DNA damage. RESULTS: Eighteen patients received FOLFIRI and AZD8931. At 160 mg bd, 1 patient experienced G3 DLT; 160 mg bd was used for cohort expansion. No grade 5 adverse events (AE) reported. Seven (39%) and 1 (6%) patients experienced grade 3 and grade 4 AEs, respectively. Of 12 patients receiving 160 mg bd, best overall response rate was 25%, median PFS and OS were 8.7 and 21.2 months, respectively. A reduction in circulating HER2/3 dimer in the two responding patients after 12 weeks treatment was observed. CONCLUSIONS: The combination of pulsed high-dose AZD8931 with FOLFIRI has acceptable toxicity. Further studies of TKI sequencing may establish a role for pulsed use of such agents rather than continuous exposure. TRIAL REGISTRATION NUMBER: ClinicalTrials.gov number: NCT01862003.


Subject(s)
Colorectal Neoplasms , Receptor, ErbB-3 , Humans , Receptor, ErbB-3/metabolism , Signal Transduction , Quinazolines/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/chemically induced , Fluorouracil , Leucovorin/adverse effects , Camptothecin , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism
6.
Nihon Shokakibyo Gakkai Zasshi ; 119(10): 937-945, 2022.
Article in Japanese | MEDLINE | ID: mdl-36216544

ABSTRACT

Intratumoral HER2 heterogeneity is a well-described gastric cancer feature and may explain many false-negative results related to this oncogene. An 81-year-old man was diagnosed at our hospital with stage IV gastric cancer with multiple lymph node metastases. Immunohistochemistry (IHC) analysis indicated that the primary tumor was HER2-negative. After a chemotherapy course, we submitted a pretreatment biopsy specimen for comprehensive cancer genome profiling (CGP) to determine the last-line therapy. This revealed HER2 amplification. The specimen was reevaluated using fluorescence in situ hybridization and IHC with deeper-cut specimens, which confirmed that the tumor was indeed HER2-positive. Therefore, the patient was treated with chemotherapy plus trastuzumab, which elicited tumor shrinkage and conferred long-term survival. Our current data underscore the CGP importance, which can provide more accurate tumor profilings and inform subsequent treatment decisions.


Subject(s)
Stomach Neoplasms , Aged, 80 and over , Biomarkers, Tumor , Humans , In Situ Hybridization, Fluorescence , Male , Receptor, ErbB-2/genetics , Receptor, ErbB-2/therapeutic use , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Trastuzumab/therapeutic use
7.
J Med Food ; 25(7): 770-777, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35834632

ABSTRACT

Umbilicaria esculenta (UE), an edible lichen, is widespread in northeast Asian countries, including China, Japan, and Korea. In the present study, we examined the antiwrinkle activity of UE. We observed that the UE extract (UEE) suppressed ultraviolet (UV)-induced matrix metalloprotein-1 (MMP-1) expression and reactive oxygen species (ROS) generation in a human keratinocyte cell line (HaCaT) and human skin tissue. In addition, UEE reversed the UV-induced decrease in collagen in the human skin tissue. Excessive and chronic UV exposure is a key factor underlying skin wrinkle formation via MMP-1 expression. As treatment with UEE disrupted the UV-activated mitogen-activated protein kinase (MAPK) signaling pathway, we applied an antibody array to unveil the underlying mechanism of UEE. Interestingly, UEE treatment inhibited ErbB2 phosphorylation, but not epidermal growth factor receptor phosphorylation, a heterodimerization partner with ErbB2. Furthermore, UEE treatment enhanced UV-suppressed phosphatase activity via ROS suppression. Collectively, our findings indicate that UEE enhances ErbB2 dephosphorylation to suppress UV-induced MMP-1 expression.


Subject(s)
Ascomycota , Receptor, ErbB-2 , Skin Aging , Skin , Tissue Extracts , Fibroblasts/drug effects , Fibroblasts/metabolism , HaCaT Cells/drug effects , HaCaT Cells/metabolism , Humans , Lichens , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , Phosphorylation/drug effects , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Skin/drug effects , Skin/metabolism , Skin/pathology , Skin Aging/drug effects , Tissue Extracts/pharmacology , Ultraviolet Rays/adverse effects
8.
Oncologist ; 27(8): 646-654, 2022 08 05.
Article in English | MEDLINE | ID: mdl-35666660

ABSTRACT

BACKGROUND: A study was initiated at Roswell Park Comprehensive Cancer Center to capture the real-world experience related to the use of CDK4/6 inhibitors (Ciclibs) for the treatment of metastatic hormone receptor-positive and HER2-negative breast cancer (HR+/HER2-). PATIENTS AND METHODS: A total of 222 patients were evaluated who received CDK4/6 inhibitors in the period from 2015 to 2021. Detailed clinical and demographic information was obtained on each patient and used to define clinical and demographic features associated with progression-free survival on CDK4/6 inhibitor-based therapies. RESULTS: In this real-world analysis, the majority of patients received palbociclib as the CDK4/6 inhibitor with letrozole or fulvestrant as the predominant endocrine therapies. The median progression-free survival (PFS) in the letrozole (27.6 months) and fulvestrant (17.2 months) groups were comparable to that observed in clinical trials. As expected, age at start of the treatment and menopausal status influenced endocrine therapy utilization but were not associated with PFS. Patients with recurrent disease had shorter PFS (P = .0024) than those presenting with de novo metastasis. The presence of visceral metastasis trended toward shorter PFS (P = .051). Similarly, prior endocrine therapy (P = .003) or chemotherapy (P = .036) was associated with shorter PFS. Body mass index was not associated with PFS or with dose interruption and/or modification. While the number of minorities in this analysis is limited (n = 26), these patients as a group had statistically shorter PFS on treatment (P = .002). CONCLUSIONS: The real-world progression-free survival with CDK4/6 inhibitors mimics that observed in the clinical trial. A number of clinical and demographic features were associated with PFS on CDK4/6 inhibitor-based therapy. Further studies are ongoing to validate these findings incorporating additional cancer centers.


Subject(s)
Breast Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Cyclin-Dependent Kinase 4 , Female , Fulvestrant/therapeutic use , Humans , Letrozole/therapeutic use , Receptor, ErbB-2/genetics , Receptor, ErbB-2/therapeutic use , Receptors, Estrogen , Receptors, Progesterone
9.
J Mol Diagn ; 24(7): 775-783, 2022 07.
Article in English | MEDLINE | ID: mdl-35526835

ABSTRACT

Novel human epidermal growth factor receptor 2 (HER2)-directed antibody-drug conjugates have demonstrated efficacy in HER2-low expressing breast cancers, which are currently defined as those with immunohistochemistry (IHC) scores of 1+ or 2+ with a negative in situ hybridization assay. However, current HER2 testing methods are designed to identify HER2-amplified tumors with high expression levels. The true definition of HER2-low expressing breast cancers remains controversial. Using quantitative molecular analysis of breast cancers based on RNA expression, the dynamic range of HER2 expression exceeds that detected by in situ IHC approaches. Erb-B2 receptor tyrosine kinase 2 (ERBB2) mRNA expression levels across IHC groups using patient samples derived from the Tamoxifen Exemestane Adjuvant Multicenter Trial were investigated. The standardized mean differences in ERBB2 mRNA scores in log base 2 are 0.47 (95% CI, 0.36-0.57), 0.58 (95% CI, 0.26-0.70), and 0.32 (95% CI, -0.12 to 0.75) when comparing IHC 0+ without staining versus IHC 0+ with some staining, IHC 0+ with some staining versus IHC 1+, and IHC 1+ versus IHC 2+/fluorescence in situ hybridization-negative, respectively. The results showed immunohistochemical methods have a comparatively limited dynamic range for measuring HER2 protein expression. The range of expression based on RNA abundance suggests a molecular method defining HER2-low cancers may better serve the treatment decision needs of this group. Indeed, the validity of RNA abundance to identify HER2-low cancers and predict treatment response needs to be further evaluated by prospective clinical trials.


Subject(s)
Breast Neoplasms , Receptor, ErbB-2 , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence/methods , Prospective Studies , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
10.
Breast Cancer Res Treat ; 191(1): 191-207, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34687411

ABSTRACT

PURPOSE: A major question when treating HR+/HER2- metastatic breast cancer (MBC) is whether early introduction of chemotherapy (CT) increases endocrine resistance. We aimed to describe progression-free survival (PFS) under first endocrine therapy (ET) depending on whether given before or after CT in a large nationwide cohort, in the pre-CDK era. METHODS: The real-life retrospective ESME database includes all patients with MBC whose first-line treatment was initiated between 2008 and 2014 in one of the 18 French Comprehensive Cancer Centres. Our primary objective was to compare PFS from start of first ET in patients with HR+/HER2- MBC who received ET or CT first. RESULTS: We identified 6293 patients who received at least one ET line during their first two therapeutic lines for MBC. As first-line therapy, 3832 (60.9%) received ET alone (ET1 1st group), whilst 2461 (39.1%) received CT, including 2024 patients (32.2%) with maintenance ET after CT (ET1 after CT group). Median PFS under first ET was 12.4 months (95% CI 11.9-13.1) in ET 1st group vs. 12.6 months in ET1 after CT group (95% CI 12.1-13.4), HR 0.96 (95% CI 0.90-1.01, P = 0.1277). CONCLUSIONS: PFS under first ET appears identical whether prescribed before or after chemotherapy. These data suggest chemotherapy does not promote endocrine resistance.


Subject(s)
Breast Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Female , Hormones , Humans , Progression-Free Survival , Receptor, ErbB-2/genetics , Retrospective Studies
11.
Br J Cancer ; 125(9): 1285-1298, 2021 10.
Article in English | MEDLINE | ID: mdl-34462586

ABSTRACT

BACKGROUND: Metastatic triple-negative breast cancer (mTNBC) is treated mainly with chemotherapy. However, resistance frequently occurs as tumours enter dormancy. Statins have been suggested as effective against cancer but as they prolong and promote dormancy, it is an open question of whether the concomitant use would interfere with chemotherapy in primary and mTNBC. We examined this question in animal models and clinical correlations. METHODS: We used a xenograft model of spontaneous metastasis to the liver from an ectopic tumour employing a mTNBC cell line. Atorvastatin was provided to sensitise metastatic cells, followed by chemotherapy. The effects of statin usage on outcomes in women with metastatic breast cancer was assessed respectively by querying a database of those diagnosed from 1999 to 2019. RESULTS: Atorvastatin had limited influence on tumour growth or chemotherapy effects in ectopic primary tumours. Interestingly, atorvastatin was additive with doxorubicin (but not paclitaxel) when targeting liver metastases. E-cadherin-expressing, dormant, breast cancer cells were resistant to the use of either statins or chemotherapy as compared to wild-type cells; however, the combination of both did lead to increased cell death. Although prospective randomised studies are needed for validation, our retrospective clinical analysis suggested that patients on statin treatment could experience prolonged dormancy and overall survival; still once the tumour recurred progression was not affected by statin use. CONCLUSION: Atorvastatin could be used during adjuvant chemotherapy and also in conjunction with metastatic chemotherapy to reduce mTNBC cancer progression. These preclinical data establish a rationale for the development of randomised studies.


Subject(s)
Atorvastatin/administration & dosage , Doxorubicin/administration & dosage , Liver Neoplasms/drug therapy , Liver Neoplasms/secondary , Triple Negative Breast Neoplasms/drug therapy , Animals , Atorvastatin/pharmacology , Cell Line, Tumor , Doxorubicin/pharmacology , Drug Synergism , Female , Humans , Liver Neoplasms/genetics , Mice , Prospective Studies , Receptor, ErbB-2/genetics , Retrospective Studies , Survival Analysis , Treatment Outcome , Triple Negative Breast Neoplasms/genetics , Xenograft Model Antitumor Assays
12.
Molecules ; 26(15)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34361688

ABSTRACT

Trans-(-)-kusunokinin, an anticancer compound, binds CSF1R with low affinity in breast cancer cells. Therefore, finding an additional possible target of trans-(-)-kusunokinin remains of importance for further development. Here, a computational study was completed followed by indirect proof of specific target proteins using small interfering RNA (siRNA). Ten proteins in breast cancer were selected for molecular docking and molecular dynamics simulation. A preferred active form in racemic trans-(±)-kusunokinin was trans-(-)-kusunokinin, which had stronger binding energy on HER2 trans-(+)-kusunokinin; however, it was weaker than the designed HER inhibitors (03Q and neratinib). Predictively, trans-(-)-kusunokinin bound HER2 similarly to a reversible HER2 inhibitor. We then verified the action of (±)-kusunokinin compared with neratinibon breast cancer cells (MCF-7). (±)-Kusunokinin exhibited less cytotoxicity on normal L-929 and MCF-7 than neratinib. (±)-Kusunokinin and neratinib had stronger inhibited cell proliferation than siRNA-HER2. Moreover, (±)-kusunokinin decreased Ras, ERK, CyclinB1, CyclinD and CDK1. Meanwhile, neratinib downregulated HER, MEK1, ERK, c-Myc, CyclinB1, CyclinD and CDK1. Knocking down HER2 downregulated only HER2. siRNA-HER2 combination with (±)-kusunokinin suppressed HER2, c-Myc, CyclinB1, CyclinD and CDK1. On the other hand, siRNA-HER2 combination with neratinib increased HER2, MEK1, ERK, c-Myc, CyclinB1, CyclinD and CDK1 to normal levels. We conclude that trans-(±)-kusunokinin may bind HER2 with low affinity and had a different action from neratinib.


Subject(s)
Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Breast Neoplasms/metabolism , Lignans/metabolism , Lignans/pharmacology , Piper nigrum/chemistry , Plant Extracts/metabolism , Plant Extracts/pharmacology , Receptor, ErbB-2/metabolism , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Female , Gene Knockdown Techniques , Humans , MCF-7 Cells , Molecular Docking Simulation , Molecular Dynamics Simulation , Quinolines/pharmacology , RNA, Small Interfering/genetics , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Transfection
13.
Breast Cancer Res Treat ; 189(1): 213-224, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33945043

ABSTRACT

PURPOSE: The presence of extensive ductal carcinoma in situ (DCIS) adjacent to HER2-positive invasive breast cancer (IBC) is often a contra-indication for breast-conserving surgery, even in case of excellent treatment response of the invasive component. Data on the response of DCIS to neoadjuvant systemic treatment (NST) are limited. Therefore, we estimated the response of adjacent DCIS to NST-containing HER2-blockade in HER2-positive breast cancer patients and assessed the association of clinicopathological and radiological factors with response. METHODS: Pre-NST biopsies were examined to determine presence of DCIS in all women with HER2-positive IBC treated with trastuzumab-containing NST ± pertuzumab between 2004 and 2017 in a comprehensive cancer center. When present, multiple DCIS factors, including grade, calcifications, necrosis, hormone receptor, and Ki-67 expression, were scored. Associations of clinicopathological and radiological factors with complete response were assessed using logistic regression models. RESULTS: Adjacent DCIS, observed in 138/316 patients with HER2-positive IBC, was eradicated after NST in 46% of patients. Absence of calcifications suspicious for malignancy on pre-NST mammography (odds ratio (OR) 3.75; 95% confidence interval (95% CI) 1.72-8.17), treatment with dual HER2-blockade (OR 2.36; 95% CI 1.17-4.75), a (near) complete response on MRI (OR 3.55; 95% CI 1.31-9.64), and absence of calcifications (OR 3.19; 95% CI 1.34-7.60) and Ki-67 > 20% in DCIS (OR 2.74; 95% CI 1.09-6.89) on pre-NST biopsy were significantly associated with DCIS response. CONCLUSIONS: As DCIS can respond to NST containing HER2-blockade, the presence of extensive DCIS in HER2-positive breast cancer before NST should not always indicate a mastectomy. The predictive factors we found could be helpful when considering breast-conserving surgery in these patients.


Subject(s)
Breast Neoplasms , Carcinoma, Ductal, Breast , Carcinoma, Intraductal, Noninfiltrating , Breast Neoplasms/drug therapy , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/surgery , Carcinoma, Intraductal, Noninfiltrating/drug therapy , Carcinoma, Intraductal, Noninfiltrating/surgery , Female , Humans , Mastectomy , Mastectomy, Segmental , Neoadjuvant Therapy , Receptor, ErbB-2/genetics
14.
Life Sci ; 273: 119239, 2021 May 15.
Article in English | MEDLINE | ID: mdl-33652033

ABSTRACT

Our previous work revealed the protective effect of Qiliqiangxin (QLQX) on cardiac microvascular endothelial cells (CMECs), but the underlying mechanisms remain unclear. We aimed to investigate whether QLQX exerts its protective effect against high-concentration angiotensin II (Ang II)-induced CMEC apoptosis through the autophagy machinery. CMECs were cultured in high-concentration Ang II (1 µM) medium in the presence or absence of QLQX for 48 h. We found that QLQX obviously inhibited Ang II-triggered autophagosome synthesis and apoptosis in cultured CMECs. QLQX-mediated protection against Ang II-induced CMEC apoptosis was reversed by the autophagy activator rapamycin. Specifically, deletion of ATG7 in cultured CMECs indicated a detrimental role of autophagy in Ang II-induced CMEC apoptosis. QLQX reversed Ang II-mediated ErbB2 phosphorylation impairment. Furthermore, inhibition of ErbB2 phosphorylation with lapatinib in CMECs revealed that QLQX-induced downregulation of Ang II-activated autophagy and apoptosis was ErbB2 phosphorylation-dependent via the AKT-FoxO3a axis. Activation of ErbB2 phosphorylation by Neuregulin-1ß achieved a similar CMEC-protective effect as QLQX in high-concentration Ang II medium, and this effect was also abolished by autophagy activation. These results show that the CMEC-protective effect of QLQX under high-concentration Ang II conditions could be partly attributable to QLQX-mediated ErbB2 phosphorylation-dependent downregulation of autophagy via the AKT-FoxO3a axis.


Subject(s)
Angiotensin II/toxicity , Autophagy , Drugs, Chinese Herbal/pharmacology , Endothelial Cells/drug effects , Forkhead Box Protein O3/metabolism , Myocytes, Cardiac/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/metabolism , Animals , Apoptosis , Endothelial Cells/metabolism , Endothelial Cells/pathology , Forkhead Box Protein O3/genetics , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Rats , Rats, Sprague-Dawley , Receptor, ErbB-2/genetics , Signal Transduction , Vasoconstrictor Agents/toxicity
15.
Arch Pathol Lab Med ; 145(6): 687-691, 2021 06 01.
Article in English | MEDLINE | ID: mdl-32649220

ABSTRACT

CONTEXT.­: Endometrial serous carcinoma is an aggressive subtype of endometrial cancer with the highest rate of recurrence and mortality among all histotypes. A recent clinical trial showed prolonged progression-free survival in advanced-stage and recurrent human epidermal growth factor receptor 2 (HER2)-positive endometrial serous carcinoma when trastuzumab was added to the standard chemotherapy regimen. This targeted therapeutic approach was recently endorsed by the National Comprehensive Cancer Network clinical guidelines. There is a growing interest among clinicians to obtain HER2 testing in endometrial serous carcinoma, and pathologists need to be prepared to recognize the unique characteristics of HER2 protein expression and gene amplification in these tumors and apply specific HER2 scoring criteria. OBJECTIVE.­: To provide a historical overview of targeted HER2 therapy in endometrial serous carcinoma and to summarize key findings from recent studies on the specific features of HER2 protein expression and gene amplification relative to other tumor types. Endometrial carcinoma-specific HER2 testing criteria are proposed based on evidence in the existing literature. DATA SOURCES.­: Sources comprise review of the literature and personal experience of the author. CONCLUSIONS.­: HER2 protein overexpression and/or gene amplification is present in approximately 25% to 30% of endometrial serous carcinomas, providing an opportunity for targeted therapy. Pathologists play a key role in tumor HER2 testing and scoring to ensure appropriate patient selection and successful clinical outcome.


Subject(s)
Cystadenocarcinoma, Serous/genetics , Endometrial Neoplasms/genetics , Gene Amplification , Genetic Testing/methods , Pathology, Clinical/methods , Receptor, ErbB-2/genetics , Antineoplastic Agents, Immunological/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cystadenocarcinoma, Serous/diagnosis , Cystadenocarcinoma, Serous/metabolism , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/metabolism , Endometrium/drug effects , Endometrium/metabolism , Endometrium/pathology , Female , Humans , Molecular Targeted Therapy/methods , Pathology, Clinical/standards , Pathology, Clinical/trends , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/metabolism , Reference Standards , Trastuzumab/therapeutic use
16.
J Nutr ; 151(1): 73-84, 2021 01 04.
Article in English | MEDLINE | ID: mdl-33188406

ABSTRACT

BACKGROUND: Aberrations in the regulation of cell proliferation perturb cellular homeostasis and lead to malignancies in which dysregulation of the cell cycle and suppressed apoptosis are 2 common phenomena. Combinatorial nutritional approaches could be efficacious in ameliorating these aberrations. OBJECTIVES: We sought to investigate the effect of dietary broccoli sprouts (BSp) and green tea polyphenol (GTP) administration on cell cycle progression and apoptosis in mammary tumors. METHODS: Forty female HER2/neu transgenic mice were randomly divided into 4 groups and treated with control, 26% BSp (wt:wt) in food, 0.5% GTPs (wt:vol) in drinking water, or combined BSp and GTPs from dams' conception until their pups were killed at 29 wk of age. Pups' tumor growth was monitored weekly for 27 wk. Tumor cell cycle- and apoptosis-related protein expression was measured. Data were analyzed with 2-factor or 3-factor (repeated-measures) ANOVA. RESULTS: Compared with the control group, BSp and/or GTPs decreased tumor incidence (P < 0.05) and combined BSp and GTPs synergistically [combination index (CIn) < 1] reduced tumor volume over time (P-time < 0.01). BSp and/or GTPs upregulated the expression of phosphatase and tension homolog, P16, and P53 (P < 0.05) and downregulated myelocytomatosis oncogene, Bmi1 polycomb ring finger oncogene, and telomerase reverse transcriptase (P < 0.05) compared with the control group. Combined BSp and GTPs synergistically (CIn < 1) downregulated the expression of cyclin B1, D1, and E1 and cyclin-dependent kinase 1, 2, and 4 (P < 0.05) compared with the control group. Moreover, combined BSp and GTPs induced apoptosis by regulating Bcl-2-associated X protein and B-cell lymphoma 2 (P < 0.05). BSp and/or GTPs also reduced the expression of DNA methyltransferase 1, 3A, and 3B and histone deacetylase 1 compared with the control group (P < 0.05). CONCLUSIONS: Collectively, lifelong BSp and GTP administration can prevent estrogen receptor-negative mammary tumorigenesis through cell cycle arrest and inducing apoptosis in HER2/neu mice.


Subject(s)
Brassica , Mammary Neoplasms, Animal/prevention & control , Polyphenols , Receptors, Estrogen/metabolism , Tea/chemistry , Animals , Cell Cycle Checkpoints/drug effects , Female , Gene Expression Regulation , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genotype , Mice , Mice, Transgenic , Random Allocation , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Estrogen/genetics
17.
PLoS One ; 15(12): e0234893, 2020.
Article in English | MEDLINE | ID: mdl-33382695

ABSTRACT

Breast cancer is the second leading cause of cancer-related mortality in women. Various nutritional compounds possess anti-carcinogenic properties which may be mediated through their effects on the gut microbiota and its production of short-chain fatty acids (SCFAs) for the prevention of breast cancer. We evaluated the impact of broccoli sprouts (BSp), green tea polyphenols (GTPs) and their combination on the gut microbiota and SCFAs metabolism from the microbiota in Her2/neu transgenic mice that spontaneously develop estrogen receptor-negative [ER(-)] mammary tumors. The mice were grouped based on the dietary treatment: control, BSp, GTPs or their combination from beginning in early life (BE) or life-long from conception (LC). We found that the combination group showed the strongest inhibiting effect on tumor growth volume and a significant increase in tumor latency. BSp treatment was integrally more efficacious than the GTPs group when compared to the control group. There was similar clustering of microbiota of BSp-fed mice with combination-fed mice, and GTPs-fed mice with control-fed mice at pre-tumor in the BE group and at pre-tumor and post-tumor in the LC group. The mice on all dietary treatment groups incurred a significant increase of Adlercreutzia, Lactobacillus genus and Lachnospiraceae, S24-7 family in the both BE and LC groups. We found no change in SCFAs levels in the plasma of BSp-fed, GTPs-fed and combination-fed mice of the BE group. Marked changes were observed in the mice of the LC group consisting of significant increases in propionate and isobutyrate in GTPs-fed and combination-fed mice. These studies indicate that nutrients such as BSp and GTPs differentially affect the gut microbial composition in both the BE and LC groups and the key metabolites (SCFAs) levels in the LC group. The findings also suggest that temporal factors related to different time windows of consumption during the life-span can have a promising influence on the gut microbial composition, SCFAs profiles and ER(-) breast cancer prevention.


Subject(s)
Diet/methods , Fatty Acids, Volatile/blood , Gastrointestinal Microbiome/drug effects , Mammary Neoplasms, Experimental/prevention & control , Polyphenols/pharmacology , Seedlings/chemistry , Actinobacteria/drug effects , Actinobacteria/isolation & purification , Actinobacteria/physiology , Animals , Brassica/chemistry , Clostridiales/drug effects , Clostridiales/isolation & purification , Clostridiales/physiology , Female , Gastrointestinal Microbiome/physiology , Gene Expression , Lactobacillus/drug effects , Lactobacillus/isolation & purification , Lactobacillus/physiology , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental/blood , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Knockout , Polyphenols/chemistry , Receptor, ErbB-2/deficiency , Receptor, ErbB-2/genetics , Receptors, Estrogen/deficiency , Receptors, Estrogen/genetics , Tea/chemistry
18.
Cell Mol Biol (Noisy-le-grand) ; 66(6): 105-111, 2020 Sep 30.
Article in English | MEDLINE | ID: mdl-33040794

ABSTRACT

Surface functionalization of nanoparticles (NPs) for therapeutic siRNA delivery into cancer cells has gained interest. The present study was designed for surface functionalization of gold nanoparticles (AuNPs) for efficient siRNA delivery and knockdown in cancer cells. In order to achieve this objective, AuNPs were coated with HER2-siRNA in the presence of 11-mercaptoundecanoic acid (11-MUA), calcium chloride (CaCl2) and polyethyleneimine (PEI) in alternate charge bearing successive layers. MCF-7 cells were cultured and transfected with fabricated assembly of AuNPs. Cytotoxicity analysis revealed that the half inhibitory concentration (IC50) for the formulation was 45.35 nM  . Total RNA was isolated from transfected cells, reverse transcribed into complementary DNA (cDNA) and real-time polymerase chain reaction (RT-PCR) was performed. The RT-PCR based delta-delta Ct analysis in treated cells revealed a significant 18.94 times decrease (p<0.001) in the expression of HER2 gene standardized with ACTB housekeeping gene as compared to untreated cells, which makes this formulation a potent approach for siRNA delivery and  gene knockout.


Subject(s)
Calcium/metabolism , Gold/chemistry , Metal Nanoparticles/chemistry , RNA, Small Interfering/genetics , Receptor, ErbB-2/genetics , Cell Line, Tumor , Fatty Acids/chemistry , Gene Transfer Techniques , Humans , MCF-7 Cells , Polyethyleneimine/chemistry , Sulfhydryl Compounds/chemistry
19.
Am J Chin Med ; 48(6): 1475-1489, 2020.
Article in English | MEDLINE | ID: mdl-32907364

ABSTRACT

Inadequate responses to traditional chemotherapeutic agents in cholangiocarcinoma (CCA) emphasize a requirement for new effective compounds for the treatment of this malignancy. This study aimed to investigate the antiproliferative property of cucurbitacin B on KKU-100 CCA cells. The determination of underlying molecular mechanisms was also carried out. The results revealed that cucurbitacin B suppressed growth and replicative ability to form colonies of CCA cells, suggesting the antiproliferative effect of this compound against the cells. Flow cytometry analysis demonstrated that the interfering effect of cucurbitacin B on the CCA cell cycle at the G2/M phase was accountable for its antiproliferation property. Accompanied with cell cycle disruption, cucurbitacin B altered the expression of proteins involved in the G2/M phase transition including downregulation of cyclin A, cyclin D1, and cdc25A, and upregulation of p21. Additional molecular studies demonstrated that cucurbitacin B suppressed the activation of focal adhesion kinase (FAK) which consequently resulted in inhibition of its kinase-dependent and kinase-independent downstream targets contributing to the regulation of cell proliferation including PI3K/PDK1/AKT and p53 proteins. In this study, the transient knockdown of FAK using siRNA was employed to ascertain the role of FAK in CCA cell proliferation. Finally, the effect of cucurbitacin B on upstream receptor tyrosine kinases regulating FAK activation was elucidated. The results showed that the inhibitory effect of cucurbitacin B on FAK activation in CCA cells is mediated via interference of EGFR and HER2 expression. Collectively, cucurbitacin B might be a promising drug for CCA treatment by targeting FAK protein.


Subject(s)
Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Focal Adhesion Protein-Tyrosine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Triterpenes/pharmacology , Tumor Suppressor Protein p53/metabolism , Bile Duct Neoplasms/diet therapy , Cell Line, Tumor , Cholangiocarcinoma/drug therapy , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression/drug effects , Humans , Molecular Targeted Therapy , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Triterpenes/therapeutic use
20.
J Med Chem ; 63(20): 11725-11755, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32931277

ABSTRACT

Mutated or amplified Her2 serves as a driver of non-small cell lung cancer or mediates resistance toward the inhibition of its family member epidermal growth factor receptor with small-molecule inhibitors. To date, small-molecule inhibitors targeting Her2 which can be used in clinical routine are lacking, and therefore, the development of novel inhibitors was undertaken. In this study, the well-established pyrrolopyrimidine scaffold was modified with structural motifs identified from a screening campaign with more than 1600 compounds, which were applied against wild-type Her2 and its mutant variant Her2-A775_G776insYVMA. The resulting inhibitors were designed to covalently target a reactive cysteine in the binding site of Her2 and were further optimized by means of structure-based drug design utilizing a set of obtained complex crystal structures. In addition, the analysis of binding kinetics and absorption, distribution, metabolism, and excretion parameters as well as mass spectrometry experiments and western blot analysis substantiated our approach.


Subject(s)
Drug Design , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Humans , Kinetics , Models, Molecular , Molecular Structure , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/chemistry , Pyrimidines/chemical synthesis , Pyrimidines/chemistry , Pyrroles/chemical synthesis , Pyrroles/chemistry , Receptor, ErbB-2/genetics , Receptor, ErbB-2/isolation & purification , Structure-Activity Relationship , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL