scholarly journals TAGUCHI-BASED OPTIMIZATION OF HEAD AND NECK CT ANGIOGRAPHY: IN-VIVO ENHANCED TRIGGERED TIMING FOR 600 PATIENTS

Author(s):  
CHIH-SHENG LIN ◽  
YUNG-FU CHEN ◽  
JIE DENG ◽  
DENG-HO YANG ◽  
MING-HSIANG CHEN ◽  
...  

The Taguchi-based optimization of head and neck CT angiography (CTA) using in-vivo enhanced triggered timing for 600 patients was accomplished in this study. A total of 600 patients were categorized into three (test, verification, and implementation groups with 360, 120, and 120 patients, respectively). The patients in the test group were randomly assigned into 18 sub-groups according to Taguchi L[Formula: see text](2[Formula: see text]) orthogonal array for optimizing factor combinations of head and neck CTA. The signal-to-noise ratio was defined as a high LRA/US ratio (both left and right arterial pressure divided by upper sinuses’ one) and low stdev. The seven factors of CTA were: (A) left- or right-hand injection; (B) tube current, mA; (C) kilovoltage peak, kVp; (D) contrast media concentration; (E) FOV; (F) flow rate of contrast media; and (G) rotation time for one CTA loop. With the Taguchi unique organization, 18 sub-groups (each containing 20 patients’ real LRA/US ratios) could cover 1458 cases and provide the appropriate reliability. The optimal factor combination was identified as follows: (A) left-hand injection, (B) tube current of 280[Formula: see text]mA, (C) 100[Formula: see text]kVp, (D) contrast media concentration of 60%, (E) FOV of 250[Formula: see text]mm, (F) contrast media flow rate of 4.0[Formula: see text]ml/s, and (G) rotation time for one CTA loop of 0.9[Formula: see text]s. The optimal suggestion was validated in the verification group with 120 patients and applied to the implementation group with 120 patients. The successful factor combination is instrumental in defining a standard protocol for medical staff in deriving the maximal LRA/US value ([Formula: see text]) of head and neck CTA.

2012 ◽  
Vol 53 (8) ◽  
pp. 929-934 ◽  
Author(s):  
Birgitta Ramgren ◽  
Isabella M Björkman-Burtscher ◽  
Stig Holtås ◽  
Roger Siemund

1977 ◽  
Author(s):  
W. H. Krause ◽  
A. Lang

It is known, that angiographic contrast media have an anticoagulant activity in vitro. The purpose of the present study was, to investigate this effect in vivo. The catheter was introduced percutaneously according to Seldinger into the femoral artery. The prothrombin time, activated thromboplastin time (APTT), thrombin time, reptilase time, fibrinogen, plasminogen, antithrombin III, platelets, fibrin/fibrinogen degradation products (FDP), haematocrit and contrast media concentration were studied in a series of 50 patients before and following abdominal aorto-arteriographic procedures up to 6 hours. Thrombin time and reptilase time were prolonged significantly 30 and 60 minutes after angiography. There was a correlation between clotting tiies and contrast media concentrations. Prothrombin time, APTT, and platelet counts remained unchanged. Fibrinogen, plasminogen,and antithrombin III levels showed a significant reduction after 30 minutes. FDP concentrations were increased significantly up to 6 hours, there was no correlation between contrast media concentrations and split products. The results were corrected for contrast media dilution according to the haematocrit. No thromboembolic complication was observed. The results suggest that angiographic procedure may initiate an intravascular coagulation with an activation of the fibrinolytic system. In addition the contrast media showed an inhibition of fibrin polymerization in vivo.


2021 ◽  
Vol 9 (Suppl 3) ◽  
pp. A915-A916
Author(s):  
Srinivas Reddy Boreddy ◽  
Reshmi Nair ◽  
Arindam Banerjee ◽  
Anshu Kuriakose ◽  
Prashant Kumar Pandey ◽  
...  

BackgroundGiven the pleiotropic functions of transforming growth factor-beta (TGFβ), current approaches to targeting systemic TGFβ will likely lead to suboptimal clinical activity and/or undesirable effects. Epidermal growth factor receptor (EGFR) is one of the most extensively validated tumor-associated antigens. Bicara Therapeutics has developed a novel bifunctional fusion protein, composed of a monoclonal antibody against EGFR and an extracellular domain of human TGFβ receptor II (TGFβRII). We demonstrate BCA101 has the potential to improve anti-tumor response by leveraging the cooperativity between EGFR and TGFβ signaling pathways while restricting TGFβ neutralization to EGFR-expressing tissues.MethodsFunctional neutralization of TGFβ by BCA101 was demonstrated by several in vitro assays which assessed TGFβ-dependent epithelial to mesenchymal transition (EMT), cell invasion, inducible Treg differentiation, as well as allogeneic immune responses in tumor cell/immune cell coculture assays. In vivo, the anti-tumor efficacy of BCA101 was determined in tumor xenograft mouse models, using either human tumor cell lines or patient-derived tumor cells (PDX), as well as in a humanized mouse model.ResultsIn vitro, we showed BCA101 is capable of simultaneously binding EGFR and TGFβ1 with a significantly higher affinity for EGFR. The incorporation of the TGFβRII ”trap” did not sterically interfere with the ability of BCA101 to bind EGFR, inhibit cell proliferation or mediate antibody-dependent cellular cytotoxicity (ADCC). Relative to cetuximab, BCA101 showed improved ability to reverse EMT and preserve ADCC activity. In tumor cell/immune cell co-culture assays, BCA101 increased production of proinflammatory cytokines associated with T and NK cell activation and suppressed VEGF release. Further, BCA101 inhibited differentiation of inducible Treg and displayed an immuno-potentiating profile in the BioMAP® TME model. In vivo, biodistribution studies showed that BCA101 localized to tumor tissues in xenograft mouse models, with comparable kinetics as cetuximab. TGFβ in tissues was neutralized to about 90% at 10 mg/kg of BCA101 while equimolar doses of TGFβRII receptor inhibited TGFβ in tumors by around 50%, confirming improved tumor localization with BCA101. In PDX models derived from head and neck cancer squamous cell carcinoma patients, BCA101 exerted sustained antitumor effect and delayed tumor growth compared to cetuximab. Finally, BCA101 improved the anti-tumor activity of PD1 blockade therapy in humanized HuNOG-EXL mice bearing PC-3 xenografts (figure 1).Abstract 874 Figure 1BCA101 shows superiority over cetuximab in animal models. (A) & (B). Patient derived xenograft (PDX) models. Patient derived tumors were engrafted into female NOG mice. Once tumor reached about 130 mm3, mice were randomized into control and test groups. Test group mice were treated with either BCA101 (A) or cetuximab (B), thrice a week (i.p), whereas control animals received placebo alone. Mice were treated for 27 days followed by a treatment-free phase until Day 79. Tumor volumes and mice weight were recorded twice a week. (C). BCA101 inhibits FaDu tumor xenograft growth (CDX) in vivo. Nude mice were implanted with FaDu cells on flanks. Once tumor reached about 100 mm3, mice were randomized (n=7) and treated with six doses of test compounds, BCA101 and cetuximab. Tumor volume and mice weight were recorded twice a week. (D). BCA101 and anti-PD1 combination studies in hu-NOG-EXL humanized animal model. PC-3 cells were implanted into flank of Hu-NOG-EXL humanized mice and randomized into control and test groups once tumors reached about 120 mm3. Test group mice were treated with cetuximab or BCA101, intraperitoneally for 6 doses. Anti-PD1 antibody (pembrolizumab) was administered intraperitoneally at a dose of 10 mg/kg with a dosage schedule of every fifth-day for 5 doses (Q5Dx5). Statistical analysis for panel (C) & (D) was performed using repeated measures two-way ANOVA followed by Bonferroni’s multiple comparison test. Significance was indicated by * = p value ≤0.05, ** = p value ≤0.01 and *** = p value ≤0.001. Tumor volumes are presented as Mean ± SEM.ConclusionsThese results support the clinical development of BCA101 as a targeted immunotherapy with the potential to induce improved anti-tumor response with a wider therapeutic window, either as a monotherapy or in combination with immune checkpoint blockade therapy.AcknowledgementsWe acknowledge Mazumdar Shaw Center for Translational Research for providing the human tissues used for the PDX studies. We thank Syngene International for conducting the PDX and humanized mice studies at their vivarium. We thank Dr. Sreesha Srinivasa for providing suggestions and feedback at the early stages of this project.ReferencesBedi A, Chang X, Noonan K , Pham V, Bedi R, Fertig EJ, Considine M, Califano JA, Borrello I, Chung CH, Sidransky D, Ravi R. Inhibition of TGF-b enhances the in vivo antitumor efficacy of EGF receptor–targeted therapy. Mol Cancer Ther 2012;11:1–11.Yegodayev KM, Novoplansky O, Golden A, Prasad M, Levin L, Jagadeeshan S, Zorea J, Dimitstein O, Joshua B-Z, Cohen L, Khrameeva E, Elkabets M. TGF-Beta-activated cancer-associated fibroblasts limit cetuximab efficacy in preclinical models of head and neck cancer. Cancers 2020;12:1–17.Ethics ApprovalMice were maintained as per the regulations of Committee for the Purpose of Control and Supervision of Experiments on Animals (CPCSEA), Government of India and Association for Assessment and Accreditation of Laboratory Animal Care (AAALAC) guidelines. All animal experiments were approved by institutional ethical committee and performed under approved protocols. For PDX model, head and neck cancer patient samples were obtained from Mazumdar Shaw Medical Foundation, Bengaluru, India after appropriate approvals were obtained from institutional ethical committee: NHH/MEC-RC2016-404


1994 ◽  
Vol 111 (3) ◽  
pp. 189-196 ◽  
Author(s):  
C SNYDERMAN ◽  
I KLAPAN ◽  
M MILANOVICH ◽  
D HEO ◽  
R WAGNER ◽  
...  

2017 ◽  
Vol 68 (8) ◽  
pp. 1944-1945
Author(s):  
Catalin Stefan ◽  
Gabriel Lostun ◽  
Alexandra Lostun

Head and neck cancer represents 3% of malignancies, and it is associated with high mortality due to advanced stage diagnosis. In early stages, the symptomatology can either be absent or be very common, misleading the patient who often ignores it. Early diagnosis of head and neck neoplasia is essential for a favorable long-term outcome. Lately new in vivo examination techniques were developed, and older ones have been improved. Today, in vivo staining techniques are an important tool in the diagnostic of head and neck cancer. Lugol iodine staining method provides valuable information concerning the tumor, allowing the surgeon to differentiate premalignant and malignant lesions.


Cancers ◽  
2021 ◽  
Vol 13 (10) ◽  
pp. 2396
Author(s):  
Nina Schoenwaelder ◽  
Inken Salewski ◽  
Nadja Engel ◽  
Mareike Krause ◽  
Björn Schneider ◽  
...  

Cyclin-dependent kinase inhibitors (CDKi´s) display cytotoxic activity against different malignancies, including head and neck squamous cell carcinomas (HNSCC). By coordinating the DNA damage response, these substances may be combined with cytostatics to enhance cytotoxicity. Here, we investigated the influence of different CDKi´s (palbociclib, dinaciclib, THZ1) on two HNSCC cell lines in monotherapy and combination therapy with clinically-approved drugs (5-FU, Cisplatin, cetuximab). Apoptosis/necrosis, cell cycle, invasiveness, senescence, radiation-induced γ-H2AX DNA double-strand breaks, and effects on the actin filament were studied. Furthermore, the potential to increase tumor immunogenicity was assessed by analyzing Calreticulin translocation and immune relevant surface markers. Finally, an in vivo mouse model was used to analyze the effect of dinaciclib and Cisplatin combination therapy. Dinaciclib, palbociclib, and THZ1 displayed anti-neoplastic activity after low-dose treatment, while the two latter substances slightly enhanced radiosensitivity. Dinaciclib decelerated wound healing, decreased invasiveness, and induced MHC-I, accompanied by high amounts of surface-bound Calreticulin. Numbers of early and late apoptotic cells increased initially (24 h), while necrosis dominated afterward. Antitumoral effects of the selective CDKi palbociclib were weaker, but combinations with 5-FU potentiated effects of the monotherapy. Additionally, CDKi and CDKi/chemotherapy combinations induced MHC I, indicative of enhanced immunogenicity. The in vivo studies revealed a cell line-specific response with best tumor growth control in the combination approach. Global acting CDKi’s should be further investigated as targeting agents for HNSCC, either individually or in combination with selected drugs. The ability of dinaciclib to increase the immunogenicity of tumor cells renders this substance a particularly interesting candidate for immune-based oncological treatment regimens.


2021 ◽  
Vol 13 (1) ◽  
Author(s):  
Xiaochen Wang ◽  
Kang Li ◽  
Yuehan Wan ◽  
Fangfang Chen ◽  
Maosheng Cheng ◽  
...  

AbstractMethyltransferase like 13 (METTL13), a kind of methyltransferase, is implicated in protein binding and synthesis. The upregulation of METTL13 has been reported in a variety of tumors. However, little was known about its potential function in head and neck squamous cell carcinoma (HNSCC) so far. In this study, we found that METTL13 was significantly upregulated in HNSCC at both mRNA and protein level. Increased METTL13 was negatively associated with clinical prognosis. And METTL13 markedly affected HNSCC cellular phenotypes in vivo and vitro. Further mechanism study revealed that METTL13 could regulate EMT signaling pathway by mediating enhancing translation efficiency of Snail, the key transcription factor in EMT, hence regulating the progression of EMT. Furthermore, Snail was verified to mediate METTL13-induced HNSCC cell malignant phenotypes. Altogether, our study had revealed the oncogenic role of METTL13 in HNSCC, and provided a potential therapeutic strategy.


2021 ◽  
Vol 21 (1) ◽  
Author(s):  
Chao Jing ◽  
Dandan Liu ◽  
Qingchuan Lai ◽  
Linqi Li ◽  
Mengqian Zhou ◽  
...  

Abstract Background Deubiquitinating enzymes (DUBs) play critical roles in various cancers by modulating functional proteins post-translationally. Previous studies have demonstrated that DUB Josephin Domain Containing 1 (JOSD1) is implicated in tumor progression, however, the role and mechanism of JOSD1 in head and neck squamous cell carcinoma (HNSCC) remain to be explored. In this study, we aimed to identify the clinical significance and function of JOSD1 in HNSCC. Methods The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases were analyzed to find novel DUBs in HNSCC. Immunohistochemistry assay was performed to determine the expression of JOSD1 in our cohort of 42 patients suffered with HNSCC. Kaplan–Meier analysis was used to identify the correlation between JOSD1 and the prognosis of HNSCC patients. The regulation of BRD4 on JOSD1 was determined by using pharmacological inhibition and gene depletion. The in vitro and in vivo experiments were conducted to elucidate the role of JOSD1 in HNSCC. Results The results of IHC showed that JOSD1 was aberrantly expressed in HNSCC specimens, especially in the chemoresistant ones. The overexpression of JOSD1 indicated poor clinical outcome of HNSCC patients. Moreover, JOSD1 depletion dramatically impaired cell proliferation and colony formation, and promoted cisplatin-induced apoptosis of HNSCC cells in vitro. Additionally, JOSD1 suppression inhibited the tumor growth and improved chemosensitivity in vivo. The epigenetic regulator BRD4 contributed to the upregulation of JOSD1 in HNSCC. Conclusions These results demonstrate that JOSD1 functions as an oncogene in HNSCC progression, and provide a promising target for clinical diagnosis and therapy of HNSCC.


Cancers ◽  
2021 ◽  
Vol 13 (7) ◽  
pp. 1742
Author(s):  
Melysa Fitriana ◽  
Wei-Lun Hwang ◽  
Pak-Yue Chan ◽  
Tai-Yuan Hsueh ◽  
Tsai-Tsen Liao

Head and neck squamous cell carcinomas (HNSCCs) are epithelial malignancies with 5-year overall survival rates of approximately 40–50%. Emerging evidence indicates that a small population of cells in HNSCC patients, named cancer stem cells (CSCs), play vital roles in the processes of tumor initiation, progression, metastasis, immune evasion, chemo-/radioresistance, and recurrence. The acquisition of stem-like properties of cancer cells further provides cellular plasticity for stress adaptation and contributes to therapeutic resistance, resulting in a worse clinical outcome. Thus, targeting cancer stemness is fundamental for cancer treatment. MicroRNAs (miRNAs) are known to regulate stem cell features in the development and tissue regeneration through a miRNA–target interactive network. In HNSCCs, miRNAs act as tumor suppressors and/or oncogenes to modulate cancer stemness and therapeutic efficacy by regulating the CSC-specific tumor microenvironment (TME) and signaling pathways, such as epithelial-to-mesenchymal transition (EMT), Wnt/β-catenin signaling, and epidermal growth factor receptor (EGFR) or insulin-like growth factor 1 receptor (IGF1R) signaling pathways. Owing to a deeper understanding of disease-relevant miRNAs and advances in in vivo delivery systems, the administration of miRNA-based therapeutics is feasible and safe in humans, with encouraging efficacy results in early-phase clinical trials. In this review, we summarize the present findings to better understand the mechanical actions of miRNAs in maintaining CSCs and acquiring the stem-like features of cancer cells during HNSCC pathogenesis.


Sign in / Sign up

Export Citation Format

Share Document