Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters











Publication year range
1.
Pharm Dev Technol ; 26(1): 21-29, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33070673

ABSTRACT

Multidrug resistance (MDR) is a serious challenge in chemotherapy and also a major threat to breast cancer treatment. As an intracellular energy factory, mitochondria provide energy for drug efflux and are deeply involved in multidrug resistance. Mitochondrial targeted delivery of doxorubicin can overcome multidrug resistance by disrupting mitochondrial function. By incorporating a reactive oxygen species (ROS)-responsive hydrophobic group into the backbone structure of hyaluronic acid - a natural ligand for the highly expressed CD44 receptor on tumor surfaces, a novel ROS-responsive and CD44-targeting nano-carriers was constructed. In this study, mitochondria-targeted triphenylphosphine modified-doxorubicin (TPP-DOX) and amphipathic ROS-responsive hyaluronic acid derivatives (HA-PBPE) were synthesized and confirmed by 1H NMR. The nanocarriers TPP-DOX @ HA-PBPE was prepared in a regular shape and particle size of approximately 200 nm. Compared to free DOX, its antitumor activity in vitro and tumor passive targeting in vivo has been enhanced. The ROS-responsive TPP-DOX@HA-PBPE nanocarriers system provide a promising strategy for the reverse of MDR and efficient delivery of doxorubicin derivatives into drug-resistant cancer cells.


Subject(s)
Antineoplastic Agents/metabolism , Breast Neoplasms/metabolism , Doxorubicin/metabolism , Drug Resistance, Multiple/drug effects , Nanoparticles/metabolism , Reactive Oxygen Species/metabolism , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/chemistry , Breast Neoplasms/drug therapy , Dose-Response Relationship, Drug , Doxorubicin/administration & dosage , Doxorubicin/chemistry , Drug Carriers/administration & dosage , Drug Carriers/chemistry , Drug Carriers/metabolism , Drug Delivery Systems/methods , Drug Resistance, Multiple/physiology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Female , Humans , MCF-7 Cells , Mice , Mice, Nude , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Reactive Oxygen Species/chemistry
3.
Aging Dis ; 11(1): 44-59, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32010480

ABSTRACT

Many oncogenes are involved in the progression from low-grade squamous intraepithelial lesions (LSILs) to high-grade squamous intraepithelial lesions (HSILs); which greatly increases the risk of cervical cancer (CC). Thus, a reliable biomarker for risk classification of LSILs is urgently needed. The prolyl isomerase Pin1 is overexpressed in many cancers and contributes significantly to tumour initiation and progression. Therefore, it is important to assess the effects of cancer therapies that target Pin1. In our study, we demonstrated that Pin1 may serve as a biomarker for LSIL disease progression and may constitute a novel therapeutic target for CC. We used a the novel Pin1 inhibitor KPT-6566, which is able to covalently bind to Pin1 and selectively target it for degradation. The results of our investigation revealed that the downregulation of Pin1 by shRNA or KPT-6566 inhibited the growth of human cervical cancer cells (CCCs). We also discovered that the use of KPT-6566 is a novel approach to enhance the therapeutic efficacy of cisplatin (DDP) against CCCs in vitro and in vivo. We showed that KPT-6566-mediated inhibition of Pin1 blocked multiple cancer-driving pathways simultaneously in CCCs. Furthermore, targeted Pin1 treatment suppressed the metastasis and invasion of human CCCs, and downregulation of Pin1 reversed the epithelial-mesenchymal transition (EMT) of CCCs via the c-Jun/slug pathway. Collectively, we showed that Pin1 may be a marker for the risk of progression to HSIL and that inhibition of Pin1 has anticancer effects against CC.

4.
Biomater Sci ; 8(1): 118-124, 2020 Jan 01.
Article in English | MEDLINE | ID: mdl-31777865

ABSTRACT

On account of the biological significance of self-assembling peptides in blocking the cellular mass exchange as well as impeding the formation for actin filaments resulting in program cell death, stimuli-responsive polypeptide nanoparticles have attracted more and more attention. In this work, we successfully fabricated doxorubicin-loaded polyethylene glycol-block-peptide (FFKY)-block-tetraphenylethylene (PEG-Pep-TPE/DOX) nanoparticles, where the aggregation-induced emission luminogens (AIEgen, TPE-CHO) can become a fluorescence resonance energy transfer (FRET) pair with the entrapped antitumor drug DOX to detect the release of drugs dynamically. This is the first successful attempt to detect and quantify the change of FRET signals in A549 cells via three methods to monitor the cellular uptake of nanoprobes and intracellular drug molecule release intuitively. As we proposed here, the combination of free DOX and the self-assembling peptide could achieve the synergistic anticancer efficacy. The multifunctional PEG-Pep-TPE/DOX nanoparticles may provide a new opportunity for combination cancer therapy and real-time detection of the drug release from stimuli-responsive nanomedicine.


Subject(s)
Antineoplastic Agents/chemistry , Doxorubicin/chemistry , Fluorescence Resonance Energy Transfer/methods , Nanoparticles/chemistry , Peptides/chemistry , Polyethylene Glycols/chemistry , Stilbenes/chemistry , A549 Cells , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacology , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cell Survival/drug effects , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Liberation , Glutathione/chemistry , Humans , Hydrogen-Ion Concentration , Luminescent Agents/chemistry , Nanoparticles/toxicity
5.
Drug Dev Ind Pharm ; 45(9): 1556-1564, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31271317

ABSTRACT

Objective: This study was aimed to develop DOX-TPP loaded acetal-PEG-PCCL micelles to improve the clinical efficacy of drug resistance tumor. Significance: Chemotherapy is one of the main treatments for breast cancer but is plagued by multidrug resistance (MDR). DOX-TPP-loaded micelles can enhance the specific concentration of drugs in the tumor and improve the efficacy and overcome MDR. Methods: In this study, DOX-TPP-loaded micelles based on acetal-PEG-PCCL were prepared and their physicochemical properties were characterized. The cellular uptake and ability to induce apoptosis of the micelles was confirmed by flow cytometry in MCF-7/ADR cells. In addition, cytotoxicity of the micelles was studied in MCF-7 cells and MCF-7/ADR cells. Confocal is used to study the subcellular distribution of DOX. Free DOX-TPP or DOX-TPP-loaded acetal-PEG-PCCL micelles were administered via intravenous injection in the tail vain for the biodistribution study in vivo. Results: The diameter of micelles was about 102.4 nm and their drug-loading efficiency is 61.8%. The structural characterization was confirmed by 1H NMR. The micelles exhibited better antitumor efficacy compared to free doxorubicin in MCF-7/ADR cells by MTT assay. The apoptotic rate and the cellular uptake of micelles were significantly higher than free DOX and DOX-TPP. Micelles can efficiently deliver mitochondria-targeting DOX-TPP to tumor cells. The result of bio-distribution showed that the micelles had stronger tumor infiltration ability than free drugs. Conclusions: In this study, mitochondriotropic DOX-TPP was conjugated to the nanocarrier acetal-PEG-PCCL via ionic interaction to form a polymer, which spontaneously formed spherical micelles. The cytotoxicity and cellular uptake of the micelles are superior to free DOX and exhibit mitochondrial targeting and passive tumor targeting, indicating that they have potential prospects.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Nanoconjugates/chemistry , Organophosphorus Compounds/administration & dosage , Acetals/chemistry , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Breast Neoplasms/pathology , Doxorubicin/analogs & derivatives , Doxorubicin/pharmacokinetics , Drug Compounding , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Female , Humans , MCF-7 Cells , Micelles , Mitochondria/drug effects , Mitochondria/pathology , Organophosphorus Compounds/chemistry , Organophosphorus Compounds/pharmacokinetics , Polyesters/chemistry , Polyethylene Glycols/chemistry , Tissue Distribution
6.
Eur J Pharm Biopharm ; 137: 122-130, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30776412

ABSTRACT

The tumor microenvironment is the cellular environment that is also described as the "soil" for supporting tumor growth, proliferation, invasion and metastasis, as well as protecting tumor cells from immunological recognition. Notably, tumor cells can grow much faster than other normal organs and invade surrounding tissues more easily, which results in abnormal expression of enzymes in the tumor microenvironment, including matrix metalloproteinases, cathepsins, phospholipases, oxidoreductases, etc. In opposite, due to the high selectivity and catalytic activity, these enzymes can promote nanoparticles to recognize tumor tissues more accurately, and the more accumulation of drugs at primal tumor sites will enhance therapeutic efficacy with lower systemic toxicity. Therefore, one promising antitumor strategy is to design stimulus-responsive nanoscale delivery systems triggered by the enzymes with the support of various nanocarriers, such as liposomes, micelles and inorganic nanoparticles, etc. In this review, numerous facts were cited to summarize and discuss the typical types of enzyme-stimulus responsive nanoscale delivery systems. More importantly, we also focused on their recent advancements in antitumor therapy, and offered the direction for further studies.


Subject(s)
Drug Delivery Systems , Neoplasms/drug therapy , Tumor Microenvironment/drug effects , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Enzymes/metabolism , Humans , Liposomes , Micelles , Nanoparticles , Neoplasms/enzymology
7.
Acta Pharmacol Sin ; 39(10): 1681-1692, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29849132

ABSTRACT

Breast cancer is the leading cause of cancer-related death for women, and multidrug resistance (MDR) is the major obstacle faced by chemotherapy for breast cancer. We have previously synthesized a doxorubicin (DOX) derivative by conjugating DOX with triphenylphosphonium (TPP) to achieve mitochondrial delivery, which induced higher cytotoxicity in drug-resistant breast cancer cells than DOX itself. Due to its amphiphilicity, TPP-DOX is difficult to physically entrap in nanocarriers. Thus, we linked it to hyaluronic acid (HA) by a novel ionic bond utilizing the specific bromide ion of TPP to form supra-molecular self-assembled structures (HA-ionic-TPP-DOX). The product was analyzed uisng 1H-NMR, 13C-NMR and mass spectrometry. The HA nanocarriers (HA-ionic-TPP-DOX) were shown to self-assemble into spherical nanoparticles, and sensitive to acidic pH in terms of morphology and drug release. Compared with free DOX, HA-ionic-TPP-DOX produced much greater intracellular DOX accumulation and mitochondrial localization, leading to increased ROS production, slightly decreased mitochondrial membrane potential, increased cytotoxicity in MCF-7/ADR cells and enhanced tumor targeting in vivo. In xenotransplant zebrafish model with the MCF-7/ADR cell line, both TPP-DOX and HA-ionic-TPP-DOX inhibited tumor cell proliferation without inducing significant side effects compared with free DOX. In addition, we observed a better anti-tumor effect of HA-ionic-TPP-DOX on MCF-7/ADR cells in zebrafish than that of TPP-DOX treatment. Furthermore, HA-ionic-DOX-TPP exhibited favorable biocompatibility and anti-tumor effects in MCF-7/ADR tumor-bearing nude mice in comparison with the effects of TPP-DOX and DOX, suggesting the potential of HA-ionic-TPP-DOX for the targeted delivery and controlled release of TPP-DOX, which can lead to the sensitization of resistant breast tumors.


Subject(s)
Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Hyaluronic Acid/chemistry , Mitochondria/metabolism , Animals , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Doxorubicin/chemistry , Drug Liberation , Drug Resistance, Neoplasm/drug effects , Female , Humans , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mice, Inbred BALB C , Mice, Nude , Onium Compounds/chemistry , Organophosphorus Compounds/chemistry , Zebrafish
8.
Mol Pharm ; 15(3): 882-891, 2018 03 05.
Article in English | MEDLINE | ID: mdl-29357260

ABSTRACT

Multidrug resistance (MDR) is the major obstacle for chemotherapy. In a previous study, we have successfully synthesized a novel doxorubicin (DOX) derivative modified by triphenylphosphonium (TPP) to realize mitochondrial delivery of DOX and showed the potential of this compound to overcome DOX resistance in MDA-MB-435/DOX cells. (1) To introduce specificity for DOX-TPP to cancer cells, here we report on the conjugation of DOX-TPP to hyaluronic acid (HA) by hydrazone bond with adipic acid dihydrazide (ADH) as the acid-responsive linker, producing HA- hydra-DOX-TPP nanoparticles. Hyaluronic acid (HA) is a natural water-soluble linear glycosaminoglycan, which was hypothesized to increase the accumulation of nanoparticles containing DOX-TPP in the mitochondria of tumor cells upon systemic administration, overcoming DOX resistance, in vivo. Our results showed HA- hydra-DOX-TPP to self-assemble to core/shell nanoparticles of good dispersibility and effective release of DOX-TPP from the HA- hydra-DOX-TPP conjugate in cancer cells, which was followed by enhanced DOX mitochondria accumulation. The HA- hydra-DOX-TPP nanoparticles also showed improved anticancer effects, better tumor cell apoptosis, and better safety profile compared to free DOX in MCF-7/ADR bearing mice.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Breast Neoplasms/drug therapy , Doxorubicin/administration & dosage , Mitochondria/metabolism , Nanoconjugates/chemistry , Animals , Antibiotics, Antineoplastic/chemistry , Apoptosis/drug effects , Breast Neoplasms/pathology , Doxorubicin/chemistry , Drug Liberation , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Female , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/chemistry , Hydrogen-Ion Concentration , Lysosomes/drug effects , Lysosomes/metabolism , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Mitochondria/drug effects , Organophosphorus Compounds/administration & dosage , Organophosphorus Compounds/chemistry , Tissue Distribution , Xenograft Model Antitumor Assays
9.
Discov Med ; 22(119): 7-17, 2016 08.
Article in English | MEDLINE | ID: mdl-27585226

ABSTRACT

Multidrug resistance (MDR) to Doxorubicin (DOX) remains a major obstacle to successful cancer treatment. The present study sought to overcome the MDR of lung cancer cells and achieve radiosensitization by developing a composite DOX-loaded micelle (M-DOX). M-DOX containing PEG-PCL/Pluronic P105 was prepared by the solvent evaporation method. Lung cancer cell line A549 was adopted in this study. In vitro cytotoxicity, cellular uptake behavior, subcellular distribution, and radiosensitivity were evaluated by the treatment with M-DOX, and free DOX was used as a control. A549 cells treated with M-DOX as opposed to free DOX showed greater cellular uptake as well as greater cytotoxicity. Furthermore, M-DOX reached the mitochondria and lysosome effectively after cellular uptake, and fluorescence used to track M-DOX was found to be surrounding the nucleus. Finally, colony-forming assays demonstrated that M-DOX treatment improved radiosensitization when compared to free DOX. Based on the increased cytotoxicity and radiosensitization, M-DOX could be considered as a promising drug delivery system to overcome MDR in lung cancer therapy.


Subject(s)
Micelles , Polymers/pharmacology , A549 Cells , Antibiotics, Antineoplastic/pharmacology , Chromatography, High Pressure Liquid , Doxorubicin/pharmacology , Drug Resistance, Neoplasm/drug effects , Flow Cytometry , Humans , Lung Neoplasms/metabolism , Microscopy, Electron, Transmission , Polymers/chemistry , Radiation Tolerance/drug effects
10.
Oncol Lett ; 12(6): 5363-5369, 2016 Dec.
Article in English | MEDLINE | ID: mdl-28105244

ABSTRACT

Gefitinib is an epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKI) that has been demonstrated to be clinically useful for the treatment of patients with non-small cell lung cancer (NSCLC). However, ~50% of patients do not respond to EGFR TKI treatment through the emergence of mutations, such as T790M. Therefore, it is important to determine which patients are eligible for treatment with gefitinib. As a preferred dimerization partner for EGFR, the role of EGFR 2 (HER2) in mediating sensitivity to gefitinib is poorly understood. In the present study, full-length human HER2 cDNA was introduced to the NSCLC cell lines H1975 and H1299, which have a low endogenous expression level of HER2. In addition, it was observed in the present study that the H1975 cell line harbored the L858R and T790M mutations in the EGFR kinase domain. Western blot analysis and MTT assay were used to evaluate the TKI sensitivity of HER2 expression status, and the activation of HER3 and HER2 downstream effectors. The results indicated that the sensitivity of H1975 cells to gefitinib was restored by the overexpression of HER2, which stimulated HER2-driven signaling cascades accompanied by the activation of protein kinase B. By contrast, ectopic HER2 overexpression in H1299 cells did not significantly alter the sensitivity to gefitinib treatment. In conclusion, the current study results suggested that the relatively resistance of the H1975 cell line to gefitinib could be reversed by the overexpression of HER2. Therefore, the expression of HER2 could also be considered when evaluate the patients' potential response to gefitinib, particularly in the subgroup of lung cancer patients who harbor an EGFR mutation.

11.
Int J Nanomedicine ; 7: 2661-71, 2012.
Article in English | MEDLINE | ID: mdl-22679376

ABSTRACT

BACKGROUND: The purpose of this study is to evaluate the efficacy of composite doxorubicinloaded micelles for enhancing doxorubicin radiosensitivity in multicellular spheroids from a non-small cell lung cancer cell line. METHODS: A novel composite doxorubicin-loaded micelle consisting of polyethylene glycolpolycaprolactone/Pluronic P105 was developed, and carrier-mediated doxorubicin accumulation and release from multicellular spheroids was evaluated. We used confocal laser scanning microscopy and flow cytometry to study the accumulation and efflux of doxorubicin from A549 multicellular spheroids. Doxorubicin radiosensitization and the combined effects of irradiation and doxorubicin on cell migration and proliferation were compared for the different doxorubicin delivery systems. RESULTS: Confocal laser scanning microscopy and quantitative flow cytometry studies both verified that, for equivalent doxorubicin concentrations, composite doxorubicin-loaded micelles significantly enhanced cellular doxorubicin accumulation and inhibited doxorubicin release. Colony-forming assays demonstrated that composite doxorubicin-loaded micelles are radiosensitive, as shown by significantly reduced survival of cells treated by radiation + composite micelles compared with those treated with radiation + free doxorubicin or radiation alone. The multicellular spheroid migration area and growth ability verified higher radiosensitivity for the composite micelles loaded with doxorubicin than for free doxorubicin. CONCLUSION: Our composite doxorubicin-loaded micelle was demonstrated to have radiosensitization. Doxorubicin loading in the composite micelles significantly increased its cellular uptake, improved drug retention, and enhanced its antitumor effect relative to free doxorubicin, thereby providing a novel approach for treatment of cancer.


Subject(s)
Doxorubicin/pharmacology , Lung Neoplasms/drug therapy , Lung Neoplasms/radiotherapy , Micelles , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Doxorubicin/chemistry , Doxorubicin/pharmacokinetics , Drug Carriers/chemistry , Flow Cytometry , Humans , Lung Neoplasms/pathology , Microscopy, Confocal , Nanocapsules/chemistry , Polyethylene Glycols/chemistry , Radiation Tolerance/drug effects , Spheroids, Cellular/drug effects , Spheroids, Cellular/radiation effects , Tumor Cells, Cultured
12.
J Zhejiang Univ Sci B ; 7(3): 235-40, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16502512

ABSTRACT

OBJECTIVES: To detect the serum proteomic patterns by using SELDI-TOF-MS (surface enhanced laser desorption/ ionization-time of flight-mass spectrometry) technology and CM10 ProteinChip in colorectal cancer (CRC) patients, and to evaluate the significance of the proteomic patterns in the tumour staging of colorectal cancer. METHODS: SELDI-TOF-MS and CM10 ProteinChip were used to detect the serum proteomic patterns of 76 patients with colorectal cancer, among them, 10 Stage I, 19 Stage II, 16 Stage III and 31 Stage IV samples. Different stage models were developed and validated by support vector machines, discriminant analysis and time-sequence analysis. RESULTS: The Model I formed by 6 protein peaks (m/z: 2759.58, 2964.66, 2048.01, 4795.90, 4139.77 and 37761.60) could be used to distinguish local CRC patients (Stage I and Stage II) from regional CRC patients (Stage III) with an accuracy of 86.67% (39/45). The Model II formed by 3 protein peaks (m/z: 6885.30, 2058.32 and 8567.75) could be used to distinguish locoregional CRC patients (Stage I, Stage II and Stage III) from systematic CRC patients (Stage IV) with an accuracy of 75.00% (57/76). The Model III could distinguish Stage I from Stage II with an accuracy of 86.21% (25/29). The Model IV could distinguish Stage I from Stage III with accuracy of 84.62% (22/26). The Model V could distinguish Stage II from Stage III with accuracy of 85.71% (30/35). The Model VI could distinguish Stage II from Stage IV with accuracy of 80.00% (40/50). The Model VII could distinguish Stage III from Stage IV with accuracy of 78.72% (37/47). Different stage groups could be distinguished by the two-dimensional scattered spots figure obviously. CONCLUSION: This method showed great success in preoperatively determining the colorectal cancer stage of patients.


Subject(s)
Biomarkers, Tumor/blood , Colorectal Neoplasms/blood , Colorectal Neoplasms/diagnosis , Gene Expression Profiling/methods , Neoplasm Proteins/blood , Preoperative Care/methods , Protein Array Analysis/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adolescent , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Female , Humans , Male , Middle Aged , Neoplasm Staging , Reproducibility of Results , Sensitivity and Specificity
13.
Zhonghua Zhong Liu Za Zhi ; 28(10): 753-7, 2006 Oct.
Article in Chinese | MEDLINE | ID: mdl-17366787

ABSTRACT

OBJECTIVE: To detect the serum proteomic patterns by using SELDI-TOF-MS and CM10 ProteinChip techniques in colorectal cancer (CRC) patients, and to evaluate the significance of the proteomic patterns in colorectal cancer staging. METHODS: A total of 76 serum samples were obtained from CRC patients at different clinical stages, including Dukes A (n = 10), Dukes B (n = 19), Dukes C (n = 16) and Dukes D (n = 31). Different stage models were developed and validated by bioinformatics methods of support vector machines, discriminant analysis and time-sequence analysis. RESULTS: The model I formed by six proteins of peaks at m/z 2759.6, 2964.7, 2048.0, 4795.9, 4139.8 and 37 761.6 could do the best as potential biomarkers to distinguish local CRC patients (Dukes A and Dukes B) from regional CRC patients (Dukes C ) with an accuracy of 86.7%. The model II formed by 3 proteins of peaks at m/z 6885.3, 2058.3 and 8567.8 could do the best to distinguish locoregional CRC patients (Dukes A, B and C) from systematic CRC patients (Dukes D) with an accuracy of 75.0%. The mode III could distinguish Dukes A from Dukes B with an accuracy of 86.2% (25/29). The model IV could distinguish Dukes A from Dukes C with an accuracy of 84.6% (22/26). The model V could distinguish Dukes B from Dukes C with an accuracy of 85.7% (30/35). The model VI could distinguish Dukes B from Dukes D with an accuracy of 80.0% (40/50). The model VII could distinguish Dukes C from Dukes D with an accuracy of 78.7% (37/47). Different stage groups could be distinguished by the two-dimensional scattered spots figure obviously. CONCLUSION: Our findings indicate that this method can well be used in preoperative staging of colorectal cancers and the screened tumor markers may serve for guidance of integrating treatment of colorectal cancers.


Subject(s)
Colorectal Neoplasms/pathology , Neoplasm Staging/methods , Protein Array Analysis/methods , Proteomics/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Adolescent , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Colorectal Neoplasms/blood , Female , Humans , Male , Middle Aged , Neoplasm Proteins/blood , Preoperative Care , Reproducibility of Results
SELECTION OF CITATIONS
SEARCH DETAIL