Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Clin Transl Med ; 14(5): e1675, 2024 May.
Article in English | MEDLINE | ID: mdl-38689424

ABSTRACT

INTRODUCTION: Intrahepatic cholangiocarcinoma (ICC) is characterized by a dismal prognosis with limited therapeutic alternatives. To explore phosphatase and tension homolog (PTEN) as a biomarker for proteasome inhibition in ICC, we conducted a phase II trial to assess the second-line efficacy of bortezomib in PTEN-deficient advanced ICC patients. METHODS: A total of 130 patients with advanced ICC in our centre were screened by PTEN immunohistochemical staining between 1 July 2017, and 31 December 2021, and 16 patients were ultimately enrolled and treated with single-agent bortezomib 1.3 mg/m2 on days 1, 4, 8 and 11 of a 21-day cycle. The primary endpoint was the objective response rate (ORR) according to Response Evaluation Criteria in Solid Tumors v1.1. RESULTS: The median follow-up was 6.55 months (95% confidence interval [CI]: 0.7-19.9 months). Among the 16 enrolled patients, the ORR was 18.75% (3/16) and the disease control rate was 43.75% (7/16). The median progress-free survival was 2.95 months (95% CI: 2.1-5.1 months) and the median overall survival (mOS) was 7.2 months (95% CI: 0.7-21.6 months) in the intent-to-treat-patients. Treatment-related adverse events of any grade were reported in 16 patients, with thrombopenia being the most common toxicity. Patients with PTEN staining scores of 0 were more likely to benefit from bortezomib than those with staining scores > 0. CONCLUSIONS: Bortezomib yielded an encouraging objective response and a favourable OS as a second-line agent in PTEN-deficient ICC patients. Our findings suggest bortezomib as a promising therapeutic option for patients with PTEN-deficient ICC. HIGHLIGHTS: There is a limited strategy for the second-line option of intrahepatic cholangiocarcinoma (ICC). This investigator-initiated phase 2 study evaluated bortezomib in ICC patients with phosphatase and tension homology deficiency. The overall response rate was 18.75% and the overall survival was 7.2 months in the intent-to-treat cohort. These results justify further developing bortezomib in ICC patients with PTEN deficiency.


Subject(s)
Bile Duct Neoplasms , Bortezomib , Cholangiocarcinoma , PTEN Phosphohydrolase , Humans , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Bortezomib/therapeutic use , Bortezomib/pharmacology , Male , Female , Middle Aged , Aged , Prospective Studies , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Adult , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology
2.
Liver Cancer ; 12(3): 281-288, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37593364

ABSTRACT

Introduction: Intrahepatic cholangiocarcinoma (ICC) is a highly aggressive liver malignancy with poor prognosis. Recently, the development of immune checkpoint inhibitors (ICIs), such as programmed cell death 1 (PD-1) inhibitors, has emerged as a promising strategy in multiple tumor types, including ICC. Microsatellite instability-high (MSI-H) is an important biomarker for ICIs in solid tumors. The response rate in patients with MSI-H is significantly higher than in those with microsatellite stability/microsatellite instability-low. And approximately 80-90% of the patients with MSI-H could maintain sustained clinical benefits once they had an initial response. However, some patients could have primary resistance at the beginning, and some might have acquired resistance after long-term treatment. Case Presentation: We present the case of an ICC patient with MSI-H who suffered rapid progression after a short-term remission with camrelizumab, a kind of PD-1 inhibitor, as second-line treatment. The patient's genomic and immune features were analyzed by next-generation sequencing and multiplex immunofluorescence staining to explore the possible mechanisms of the rapidly acquired resistance of ICIs in this MSI-H case. Conclusion: The genomic and immunohistochemical analysis showed that TGFBR2 mutation, loss of HLA B44 supertype, carrying B62 supertype, and increased PD-L1+ cells, macrophages, and Tregs in the tumor microenvironment might be related to the nonsustain benefit of ICIs in this MSI-H patient.

3.
Sci Transl Med ; 15(704): eadd7464, 2023 07 12.
Article in English | MEDLINE | ID: mdl-37437018

ABSTRACT

Gemcitabine is a nucleoside analog that has been successfully used in the treatment of multiple cancers. However, intrinsic or acquired resistance reduces the chemotherapeutic potential of gemcitabine. Here, we revealed a previously unappreciated mechanism by which phosphatase and tensin homolog (PTEN), one of the most frequently mutated genes in human cancers, dominates the decision-making process that is central to the regulation of gemcitabine efficacy in cholangiocarcinoma (CCA). By investigating a gemcitabine-treated CCA cohort, we found that PTEN deficiency was correlated with the improved efficacy of gemcitabine-based chemotherapy. Using cell-based drug sensitivity assays, cell line-derived xenograft, and patient-derived xenograft models, we further confirmed that PTEN deficiency or genetic-engineering down-regulation of PTEN facilitated gemcitabine efficacy both in vitro and in vivo. Mechanistically, PTEN directly binds to and dephosphorylates the C terminus of the catalytic subunit of protein phosphatase 2A (PP2Ac) to increase its enzymatic activity, which further dephosphorylates deoxycytidine kinase (DCK) at Ser74 to diminish gemcitabine efficacy. Therefore, PTEN deficiency and high phosphorylation of DCK predict a better response to gemcitabine-based chemotherapy in CCA. We speculate that the combination of PP2A inhibitor and gemcitabine in PTEN-positive tumors could avoid the resistance of gemcitabine, which would benefit a large population of patients with cancer receiving gemcitabine or other nucleoside analogs.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Humans , Phosphorylation , Gemcitabine , Nucleosides , Bile Ducts, Intrahepatic , PTEN Phosphohydrolase
4.
Nat Commun ; 14(1): 1340, 2023 03 11.
Article in English | MEDLINE | ID: mdl-36906670

ABSTRACT

The prognosis of biliary tract cancer (BTC) remains unsatisfactory. This single-arm, phase II clinical trial (ChiCTR2000036652) investigated the efficacy, safety, and predictive biomarkers of sintilimab plus gemcitabine and cisplatin as the first-line treatment for patients with advanced BTCs. The primary endpoint was overall survival (OS). Secondary endpoints included toxicities, progression-free survival (PFS), and objective response rate (ORR); multi-omics biomarkers were assessed as exploratory objective. Thirty patients were enrolled and received treatment, the median OS and PFS were 15.9 months and 5.1 months, the ORR was 36.7%. The most common grade 3 or 4 treatment-related adverse events were thrombocytopenia (33.3%), with no reported deaths nor unexpected safety events. Predefined biomarker analysis indicated that patients with homologous recombination repair pathway gene alterations or loss-of-function mutations in chromatin remodeling genes presented better tumor response and survival outcomes. Furthermore, transcriptome analysis revealed a markedly longer PFS and tumor response were associated with higher expression of a 3-gene effector T cell signature or an 18-gene inflamed T cell signature. Sintilimab plus gemcitabine and cisplatin meets pre-specified endpoints and displays acceptable safety profile, multiomics potential predictive biomarkers are identified and warrant further verification.


Subject(s)
Bile Duct Neoplasms , Biliary Tract Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biliary Tract Neoplasms/drug therapy , Biliary Tract Neoplasms/pathology , Cisplatin/therapeutic use , Deoxycytidine/therapeutic use , Gemcitabine
5.
Front Immunol ; 13: 943066, 2022.
Article in English | MEDLINE | ID: mdl-36159865

ABSTRACT

Background: Immune checkpoint inhibitor (ICI)-combined chemotherapy in advanced intrahepatic cholangiocarcinoma has been proved to have more efficacy in a series of clinical trials. However, whether the tumor microenvironment (TME) plays a vital role in immune-combined therapy has not been rigorously evaluated. Methods: Firstly, we assayed the immunogenic properties of GEM-based chemotherapy. Then, 12 ICC patients treated with PD-1 inhibitor (sintilimab) combined with gemcitabine and cisplatin (GemCis) from a phase 2 clinical trial (ChiCTR2000036652) were included and their immune-related gene expression profiles were analyzed using RNA from baseline tumor samples. Immune-related signature correlating with clinical outcome was identified according to the 12 ICC patients, and its predictive value was validated in an ICC cohort with 26 patients. Multiplexed immunofluorescence (mIF) and flow cytometry (FCM) analysis were performed to evaluate the immune-related molecules with therapeutic outcomes. Results: GEM-based chemotherapy induced immunogenic cell death of cholangiocarcinoma cells, together with increased CD274 expression. In an ICC cohort, we found that upregulation of immune-checkpoint molecules and immune response-related pathways were significantly related to better clinical outcome. On the contrary, baseline immune-cell proportions in tumor tissues did not show any correlation with clinical benefit between responders and non-responders. Immune-related signature (including six genes) correlating with clinical outcome was identified according to the 12 ICC patients, and its predictive value was validated in a small ICC cohort with 26 patients. Conclusion: Immune-related RNA signature predicts the outcome of PD-1 inhibitor-combined GEMCIS therapy in advanced intrahepatic cholangiocarcinoma, which could be tested as a biomarker for immune-chemotherapy in the future.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/drug therapy , Bile Duct Neoplasms/genetics , Bile Ducts, Intrahepatic/metabolism , Cholangiocarcinoma/drug therapy , Cholangiocarcinoma/genetics , Cholangiocarcinoma/metabolism , Cisplatin/therapeutic use , Clinical Trials, Phase II as Topic , Humans , Immune Checkpoint Inhibitors/therapeutic use , RNA , Tumor Microenvironment
6.
Hepatobiliary Pancreat Dis Int ; 21(5): 440-449, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36115807

ABSTRACT

BACKGROUND: Biliary tract cancers (BTCs) comprise a heterogeneous group of aggressive malignancies with unfavorable prognoses. The benefit of chemotherapy seems to have reached a bottleneck and, therefore, new effective therapeutic strategies for advanced BTCs are needed. Molecularly targeted therapies in selected patients are rapidly changing the situation. However, the low frequency of specific driver alterations in BTCs limits their wide application. Recently, immunotherapeutic approaches are also under active investigation in BTCs, but the role of immunotherapy in BTCs remains controversial. DATA SOURCES: PubMed, Web of Science, and meeting resources were searched for relevant articles published from January 2017 to May 2022. The search aimed to identify current and emerging immunotherapeutic approaches for BTCs. Information on clinical trials was obtained from https://clinicaltrials.gov/ and http://www.chictr.org.cn/. RESULTS: Immunotherapy in BTC patients is currently under investigation, and most of the investigations focused on the application of immune checkpoint inhibitors (ICIs). However, only a subgroup of BTCs with microsatellite-instability high (MSI-H)/DNA mismatch repair-deficient (dMMR) or tumor mutational burden-high (TMB-H) benefit from monotherapy of ICIs, and limited activity was observed in the second or subsequent settings. Nevertheless, promising results come from studies of ICIs in combination with other therapeutic approaches, including chemotherapy, in advanced BTCs, with a moderate toxicity profile. Recent studies demonstrated that compared to GEMCIS alone, durvalumab plus GEMCIS significantly improved patient survival (TOPAZ-1 trial) and that ICIs-combined chemoimmunotherapy is poised to become a new frontline therapy option, regardless of TMB and MMR/MSI status. Adoptive cell therapy and peptide- or dendritic-based cancer vaccines are other immunotherapeutic options that are being studied in BTCs. Numerous biomarkers have been investigated to define their predictive role in response to ICIs, but no predictive biomarker has been validated, except MSI-H/dMMR. CONCLUSIONS: The role of immunotherapy in BTCs is currently under investigation and the results of ongoing studies are eagerly anticipated. Several studies have demonstrated the safety and efficacy of ICIs in combination with chemotherapy in treatment-naive patients, such as the phase III TOPAZ-1 trial, which will change the standard care of first-line chemotherapy for advanced BTCs. However, further research is needed to understand the best combination with immunotherapy and to discover more predictive biomarkers to guide clinical practice.


Subject(s)
Bile Duct Neoplasms , Biliary Tract Neoplasms , Cancer Vaccines , Biliary Tract Neoplasms/therapy , Cancer Vaccines/adverse effects , Humans , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/methods , Microsatellite Instability
7.
J Clin Lab Anal ; 36(3): e24244, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35040184

ABSTRACT

BACKGROUND: Long non-coding RNAs (lncRNAs) play crucial roles in immune regulation and, therefore, may be closely related to the tumor microenvironment (TME). However, there are few studies regarding the relationship between the lncRNAs and the TME in liver cancer. METHODS: Firstly, we constructed a lncRNA signature based on the top 10 immune-inversely related lncRNAs obtained from the ImmLnc database and performed disease-free survival (DFS) and overall survival (OS) analyses for the patients included in the Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) stratified by the lncRNA signature. Then, we explored the relationship between the lncRNA signature with distinct mutation profiles and the tumor microenvironment (TME). RESULTS: The lncRNA signature was successfully constructed and verified by survival analysis. The high lncRNA signature was correlated with a decreased DFS and OS in liver cancer and other two gastrointestinal cancers. The mutation profiles showed that the Lnc_high group had a higher number of mutations on many genes, mostly enriched in p53 and WNT pathways. The TME results showed that the Lnc_high group had the highest proportion (51%) of lymphocyte depletion-characterized immune subtype, and a higher expression of immune checkpoint molecules such as LAG3, PD-L1, CTLA4. On the contrary, in the Lnc_low group, infiltrating immune-cell proportions were significantly higher, and a significant enhancement of four axes of the cancer immunity cycle immunogram was observed in this group. CONCLUSIONS: The lncRNA signature we constructed identified an immune-excluded subtype of liver cancer with unfavorable clinic outcomes, which could be tested as a biomarker for immunotherapy in the future.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , RNA, Long Noncoding , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/pathology , Gene Expression Profiling/methods , Humans , Liver Neoplasms/pathology , RNA, Long Noncoding/metabolism , Tumor Microenvironment/genetics
8.
Altern Ther Health Med ; 28(2): 24-31, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34653023

ABSTRACT

OBJECTIVE: Prognosis of cholangiocarcinoma is poor, and palliative treatment options are limited in China. This study aimed to analyze prognostic factors affecting survival in patients with advanced cholangiocarcinoma. METHODS: Clinical data on 201 consecutive patients with cholangiocarcinoma who received treatment at a single center from May 2014 to December 2018 were analyzed retrospectively. Survival curves were plotted using the Kaplan-Meier method. Survival analyses were performed using a log-rank test. RESULTS: For first-line therapy, the disease control rate was 56% (85/152) and the overall response rate was 16% (24/152). The total disease control rate was 34% (23/67) for second-line therapy. The median progression-free survival was 7 months, and the median overall survival was 17 months. Next-generation sequencing was performed for 59 patients. The most frequently mutated genes were TP53 and PI3KCA. No significant association was found between gene mutations and treatment response or survival. Of 5 patients with high levels of microsatellite instability, 4 (80%) were sensitive to anti-programmed death 1 antibodies and remained in partial remission at last follow-up. CONCLUSIONS: Macroscopic tumor characteristics, rather than gene mutations, determine the prognosis of advanced cholangiocarcinoma. High microsatellite instability may be a favorable predictor of response to immunotherapy for cholangiocarcinoma.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Bile Duct Neoplasms/therapy , Bile Ducts, Intrahepatic , Cholangiocarcinoma/therapy , Humans , Palliative Care , Prognosis , Retrospective Studies
9.
Front Immunol ; 12: 799822, 2021.
Article in English | MEDLINE | ID: mdl-35003124

ABSTRACT

Intrahepatic cholangiocarcinoma (iCCA) is the second most common primary liver cancer with a poor prognosis. Recently, an immunotherapy strategy represented by programmed cell death 1 (PD-1) inhibitors has been applied to the systemic treatment of advanced iCCA. However, immunotherapy combined with chemotherapy as first-line maintenance therapy was rarely reported. Our report presented an advanced iCCA patient who had a dramatic response to the PD-1 inhibitor sintilimab combined with gemcitabine plus cisplatin as the first-line therapy and sintilimab combined with capecitabine as maintenance therapy, yielding an ongoing progression-free survival of 16 months.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Bile Duct Neoplasms/drug therapy , Capecitabine/administration & dosage , Cholangiocarcinoma/drug therapy , Immune Checkpoint Inhibitors/administration & dosage , Maintenance Chemotherapy/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Humans , Male , Middle Aged
10.
Sci Transl Med ; 12(562)2020 09 23.
Article in English | MEDLINE | ID: mdl-32967970

ABSTRACT

Patient-derived xenografts (PDXs) and PDX-derived cells (PDCs) are useful in preclinical research. We performed a drug screening assay using PDCs and identified proteasome inhibitors as promising drugs for cholangiocarcinoma (CCA) treatment. Furthermore, we determined that phosphate and tensin homology deleted on chromosome ten (PTEN) deficiency promotes protein synthesis and proteasome subunit expression and proteolytic activity, creating a dependency on the proteasome for cancer cell growth and survival. Thus, targeting the proteasome machinery with the inhibitor bortezomib inhibited the proliferation and survival of CCA cells lacking functional PTEN. Therapeutic evaluation of PDXs, autochthonous mouse models, and patients confirmed this dependency on the proteasome. Mechanistically, we found that PTEN promoted the nuclear translocation of FOXO1, resulting in the increased expression of BACH1 and MAFF BACH1 and MAFF are transcriptional regulators that recognize the antioxidant response element, which is present in genes encoding proteasome subunits. PTEN induced the accumulation and nuclear translocation of these proteins, which directly repressed the transcription of genes encoding proteasome subunits. We revealed that the PTEN-proteasome axis is a potential target for therapy in PTEN-deficient CCA and other PTEN-deficient cancers.


Subject(s)
Bile Duct Neoplasms , Cholangiocarcinoma , Animals , Bile Duct Neoplasms/drug therapy , Bile Ducts, Intrahepatic , Bortezomib/pharmacology , Bortezomib/therapeutic use , Cell Line, Tumor , Cholangiocarcinoma/drug therapy , Humans , Mice , PTEN Phosphohydrolase/genetics , Proteasome Endopeptidase Complex
11.
J Cancer Res Clin Oncol ; 146(10): 2651-2657, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32623573

ABSTRACT

PURPOSE: Patients with DNA mismatch repair-deficient (dMMR)/microsatellite instability-high (MSI-H) cancers are prone to response to programmed cell death-1 (PD-1) checkpoint inhibitors. Therefore, we explored the efficacy and safety of a PD-1 checkpoint inhibitor camrelizumab in advanced or metastatic solid tumour with dMMR/MSI-H. METHODS: Patients with dMMR/MSI-H advanced or metastatic solid tumours who had received at least one line of prior systemic chemotherapy were recruited. Camrelizumab was given intravenously 200 mg every 2-week treatment cycle. The primary endpoint was objective response rate according to Response Evaluation Criteria in Solid Tumours v1.1. RESULTS: Twelve patients were enrolled. As data cutoff, eight patients (66.7%, 95% CI 34.9-90.1) achieved objective response. Disease control rate reached 100% (95% CI 73.5-100). Progression-free survival rate at 12 months was 83.3% (95% CI 48.2-95.6), and overall survival rate at 12 months was 90% (95% CI 47.3-98.5). The most common treatment-related adverse events were reactive cutaneous capillary endothelial proliferation (100%), increased alanine aminotransferase (41.7%), and increased aspartate aminotransferase (41.7%). CONCLUSIONS: Camrelizumab provided durable objective response and disease control in pre-treated patients with dMMR/MSI-H advanced or metastatic solid tumour, being a promising treatment option for these patients.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Agents, Immunological/adverse effects , Antineoplastic Agents, Immunological/therapeutic use , DNA Mismatch Repair , Female , Humans , Male , Microsatellite Instability , Middle Aged , Neoplasm Metastasis , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Progression-Free Survival , Prospective Studies , Treatment Outcome
12.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-793104

ABSTRACT

@# Objective: :To detect the gene mutation in cholangiocarcinoma patients using the next generation sequencing (NGS) technology, and to analyze its correlation to the prognosis of the patients. Methods: From June 2016 to June 2018, 40 patients diagnosed with cholangiocarcinoma received NGS examination to screen the possible mutations (single base mutation, structural variation, copy number variation and gene fusion, etc.). The disease control rates (DCR), progression-free survival (PFS) and overall survival (OS) of the patients, who received the first line therapy, were retrospectively reviewed to analyze the relationship between signaling pathway as well as its genetic variation and the prognosis of cholangiocarcinoma patients. Results: The median PFS of patients with and without TP53 mutation was 11.0 and 8.3 months, respectively (P=0.332), while OS was 14.3 and 32.9 months, respectively (P=0.041). The median PFS of patients with and without PI3K mutations was 8.3 and 11.0 months, respectively (P=0.285), while OS was 14.3 and 37.0 months, respectively (P=0.020). The median PFS of patients with and without mTOR pathway mutations was 6.3 and 10.3 months, respectively (P=0.020), while OS was 15.6 and 19.6 months, respectively (P=0.892). There was no significant effect of pathway-related gene mutations on patients’survival. Conclusion: The prognosis of cholangiocarcinoma patients with TP53 and PI3K pathway activation had obviously poor prognosis than those without. No significant difference was observed between the patients with and without mTOR pathway activation and IDH mutation.

13.
Article in English | MEDLINE | ID: mdl-27336703

ABSTRACT

Multiple myeloma (MM), a malignant neoplastic serum-cell disorder, has been a serious threat to human health. The determination of 6 commonly used drug concentrations, including thalidomide, lenalidomide, cyclophosphamide, bortezomib, dexamethasone and adriamycin, in MM patients was of great clinical interest. Herein, we reported a method for the rapid and simultaneous measurement of the above therapeutics by liquid chromatography-tandem mass spectroscopy (LC-MS/MS) method with solid phase extraction. Analysis was performed on a Waters XBridge(®) BEH C18 column (2.5µm, 2.1 mm×50mm), with formic acid aqueous solution and acetonitrile as the mobile phase at flow rate 0.3mL/min. All analytes showed good correlation coefficients (r>0.996), and LLOQ of thalidomide, lenalidomide, cyclophosphamide, bortezomib, dexamethasone and adriamycin were 4, 2, 2, 2, 2 and 2ng/mL, respectively. The inter- and intra-day precisions and stability were expressed as variation coefficients within 15% and relative error less than 15%. Dilution effect, carryover and incurred sample reanalysis were investigated according to the 2015 edition Chinese Pharmacopoeia guidelines, as US FDA (2013, revision 1) required. The LC-MS/MS based assay described in this article may improve future clinical studies evaluating common therapeutics for MM treatment.


Subject(s)
Antineoplastic Agents/blood , Bortezomib/blood , Cyclophosphamide/blood , Dexamethasone/blood , Doxorubicin/blood , Thalidomide/analogs & derivatives , Thalidomide/blood , Angiogenesis Inhibitors/blood , Chromatography, Liquid/methods , Drug Monitoring/methods , Humans , Immunosuppressive Agents/blood , Lenalidomide , Limit of Detection , Multiple Myeloma/blood , Multiple Myeloma/drug therapy , Tandem Mass Spectrometry/methods
14.
Zhonghua Xue Ye Xue Za Zhi ; 37(4): 288-91, 2016 Apr.
Article in Chinese | MEDLINE | ID: mdl-27093990

ABSTRACT

OBJECTIVE: To investigate the significance of serum IgD quantitation in evaluation of clinical efficacy in IgD myeloma. METHODS: Serum IgD and free light chain (sFLC) levels were determined by immune scatter turbidimetry with SPA plus analysis machine in 29 patients with IgD multiple myeloma (MM) achieving VGPR or better response following previous treatments. The concurrent immunofixation electrophoresis (IFE) results were also incorporated and analyzed. RESULTS: Increased IgD levels were detected in 1 of 12 patients achieving sCR, 2 of 5 patients achieving CR and 4 of 12 patients achieving VGPR, respectively. The median progression-free survival (PFS) was 38.5 months, 34.1 months and 15.5 months for patients achieving sCR, CR and VGPR, respectively, with a significant difference between sCR and VGPR groups (P=0.022), and between CR and VGPR groups (P=0.018). There was no difference in overall survival (OS) among sCR, CR and VGPR groups (P>0.05). The median PFS were 7.8, 33.7 and 43.9 months, respectively for the patients with both abnormal sFLC ratios and IgD levels (6 cases, Group A), with either abnormal sFLC ratios or increased IgD levels (10 cases, Group B) or with normal sFLC ratios and IgD levels (13 cases, Group C). A significant PFS benefit of Group A over Group C was found (P=0.033), and no differences in terms of OS among three groups (P>0.05). CONCLUSION: IgD levels may remain abnormal in IgD MM patients who have achieved VGPR or better response, and IgD quantitation represented a useful assay complementary to the current lab examinations. IgD quantitation assay was of significance in clinical efficacy evaluation and survival judgement, and should be incorporated into the evaluation parameters used for IgD MM in addition to sFLC and IFE assays.


Subject(s)
Immunoglobulin D/blood , Multiple Myeloma/blood , Disease-Free Survival , Humans , Immunoglobulin Light Chains/blood , Multiple Myeloma/diagnosis , Nephelometry and Turbidimetry , Remission Induction , Treatment Outcome
15.
Zhonghua Xue Ye Xue Za Zhi ; 37(1): 14-9, 2016 Jan.
Article in Chinese | MEDLINE | ID: mdl-26876247

ABSTRACT

OBJECTIVE: To explore the efficacy and prognostic factors of induction therapy combined with autogenetic peripheral blood stem cells transplantation (APBSCT)in patients with multiple myeloma (MM). METHODS: From January 1998 to May 2015, 201 patients with MM were enrolled. All patients received APBSCT after induction therapy. With the follow up to 20 June 2015, the overall survival (OS), progression free survival (PFS)and prognostic factor were analyzed. RESULTS: ① With a media follow up of 36.67 months, the median PFS and OS were 22.87 (17.48- 28.26)and 69.63 (63.57- 75.69)months, 5-year PFS and OS were 17% and 49%, respectively. ②After APBSCT, when the subgroup (n= 112) achieved complete response (CR)compared with the subgroup (n=89) not achieved CR, the median PFS were 32.93 (21.03-44.83) and 18.13 (14.46-21.80) months (P<0.001), respectively; And the media OS were 96.77 (71.79- 121.75)and 54.70 (49.53- 59.87) months (P=0.004), respectively. The risks for disease progression and death declined in CR subgroup. ③ Two subgroups included or not included bortezomib/thalidomide at induction therapy (123 patientsvs 21 patients), the media PFS were 31.67 (24.36- 38.98)and 15.20 (10.11- 20.29) months (P=0.013), respectively; And the media OS were 76.30 (55.44- 97.15)and 52.03 (33.76- 70.30) months (P=0.014), respectively. ④According to the ISS stage, the media OS of stageⅠ, Ⅱ, Ⅲ were 99.47 (59.58-139.36), 66.77 (52.17-81.37), 53.97 (28.71-79.23) (P< 0.001), respectively. The risk for death of stage Ⅱ, Ⅲ were 2.16 and 3.04 times higher than stage Ⅰ, with no difference in terms of PFS. ⑤ The media PFS in IgD (n=22) and IgG (n=101) type MM were 11.17 (10.27- 13.13)and 35.43 (22.69- 48.17)months (P=0.007) , respectively; The media OS were 30.83 (0.24-61.42)and 70.70 (53.52-87.88) months (P=0.039), respectively. The risk for disease progression of IgD type was 2.47 times higher than IgG type. ⑥ Patients received 1 line induction therapy (n=132) compared with more than 1 line (n=69), the media PFS were 25.43 (16.09- 34.77)and 20.27 (15.04- 25.50) months (P=0.042). ⑦Cox analysis showed that CR after APBSCT and ISS stage were independent prognostic factors for OS. IgD type MM and CR after APBSCT were independent prognosis factor for PFS. CONCLUSION: CR after APBSCT and ISS stage were independent prognostic factors for OS in MM. CR after APBSCT was independent prognostic factor for PFS in MM. However, disease progression more likely occurred in IgD type MM, which was independent negative prognostic factor for PFS in MM.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Multiple Myeloma/therapy , Peripheral Blood Stem Cell Transplantation , Bortezomib/therapeutic use , Disease-Free Survival , Humans , Multiple Myeloma/diagnosis , Neoadjuvant Therapy , Prognosis , Remission Induction , Survival Rate , Thalidomide/therapeutic use , Transplantation, Autologous , Treatment Outcome
16.
Nanotechnology ; 26(38): 385702, 2015 Sep 25.
Article in English | MEDLINE | ID: mdl-26335515

ABSTRACT

A simple strategy of Ce(3+) doping is proposed to realize multicolor tuning and predominant red emission in BaLnF5:Yb(3+)/Ho(3+) (Ln(3+) = Gd(3+), Y(3+), Yb(3+)) systems. A tunable upconversion (UC) multicolor output from green/yellow to red can be readily achieved in a fixed Yb(3+)/Ho(3+) composition by doping Ce(3+), providing an effective route for multicolor tuning widely used for various optical components. Moreover, compared with Ce(3+)-free UC nanoparticles (UCNPs), a remarkable enhancement of the red-to-green (R/G) ratio is observed by doping 30% Ce(3+), arising from the two largely promoted cross-relaxation (CR) processes between Ce(3+) and Ho(3+). UCNPs with pure red emission are selected as in vivo UC bioimaging agents, demonstrating the merits of deep penetration depth, the absence of autofluorescence and high contrast in small animal bioimaging. Moreover, such fluorescence imaging nanoprobes can also be used as contrast agents for three-dimensional (3D) x-ray bioimaging by taking advantage of the high K-edge values and x-ray absorption coefficients of Ba(2+), Gd(3+), and Ce(3+) in our designed nanoprobes. Thus, the simultaneous realization of multicolor output, highly enhanced R/G ratio, and predominant red emission makes the Ce(3+)-doped UCNPs very useful for widespread applications in optical components and bioimaging.


Subject(s)
Cerium/chemistry , Molecular Imaging/methods , Nanoparticles/analysis , Nanoparticles/chemistry , Optical Imaging/methods , Animals , Color , Gadolinium/chemistry , Holmium/chemistry , Mice, Inbred Strains , Molecular Imaging/instrumentation , Nanoparticles/administration & dosage , Optical Devices , Optical Imaging/instrumentation , Tissue Distribution , X-Rays , Ytterbium/chemistry , Yttrium/chemistry
17.
Zhonghua Yi Xue Za Zhi ; 95(10): 731-5, 2015 Mar 17.
Article in Chinese | MEDLINE | ID: mdl-26080842

ABSTRACT

OBJECTIVE: To evaluate the impact of cytogenetic grouping and autologous stem cell transplantation on the prognosis of patients with multiple myeloma (MM) induced by PAD (velcade+epirubicin+dexamethasone) and TAD (thalidomide+epirubicin+dexamethasone). METHODS: A total of 191 patients with a definite diagnosis of MM were enrolled from May 2008 to December 2013 into this prospective study. They were non-randomly induced by PAD (n = 132) or TAD (n = 59) plus autologous stem cell transplantation or chemotherapy. Response and survival rates were also analyzed between two groups. RESULTS: The overall response rates of PAD and TAD groups were 84.4% (108/128) and 69.5% (41/59) (P = 0.011) respectively. The very good partial remission (VGPR) rates were 70.3% (90/128) and 32.2% (19/59) (P < 0.001) and near complete remission/complete remission (nCR/CR) rates 68.0% (87/128) and 25.4% (15/59) respectively (P < 0.001). Both progression-free survival (PFS) and overall survival (OS) showed no significant inter-group difference (P = 0.223, 0.989). The survival analysis of PAD group showed that FISH high-risk group had shorter PFS and OS than FISH low-risk group (15.2 vs 19.1 months for PFS, P = 0.098; 41.2 months vs non-attaining for OS, P = 0.017). In FISH high-risk group, patients consolidated with autologous stem cell transplantation showed longer PFS than those with chemotherapy (17.8 vs 14.6 months, P = 0.029) while the OS showed no difference (P = 0.840). In FISH low-risk group, no difference were observed in PFS and OS between patients with consolidation therapies alone (P = 0.131, 0.294). CONCLUSIONS: The response rates are higher in patients induced by PAD than by TAD. After PAD induction, cytogenetic grouping may further distinguish the prognosis of MM patients. For FISH high-risk patients, their PFS is prolonged by autologous stem cell transplantation.


Subject(s)
Multiple Myeloma , Antineoplastic Combined Chemotherapy Protocols , Disease-Free Survival , Hematopoietic Stem Cell Transplantation , Humans , Prognosis , Prospective Studies , Remission Induction , Survival Rate , Transplantation, Autologous
18.
Stem Cells Int ; 2015: 613045, 2015.
Article in English | MEDLINE | ID: mdl-25688273

ABSTRACT

Aim. To evaluate whether patients with multiple myeloma at various risks can still benefit the same from autologous stem cell transplantation consolidation in the era of novel agents. We retrospectively analyzed 67 consecutive myeloma patients receiving autologous stem cell transplantation after bortezomib and/or thalidomide based inductions. Totally 17 high-risk, 24 intermediate-risk, and 26 low-risk patients were enrolled, based on fluorescence in situ hybridization and ISS stage. Meanwhile, another 67 risk-, response depth-, and age-matched patients not proceeding to autologous stem cell transplantation were chosen as controls. Our preliminary data indicated that, in the high-risk subgroup, progression-free survival and overall survival were both significantly prolonged after autologous stem cell transplantation (P < 0.001 and P = 0.015) while, in the intermediate-risk subgroup, neither progression-free survival nor overall survival was prolonged significantly after autologous stem cell transplantation (P > 0.05), and in the low-risk subgroup, only progression-free survival was extended significantly (P = 0.012) after autologous stem cell transplantation. Multiple variables analysis further indicated that autologous stem cell transplantation and risk stratification were two independent prognostic factors for overall survival. Our results indicated that myeloma patients at different risks all benefit from autologous stem cell transplantation consolidation even in the era of novel agents.

19.
Clin Exp Med ; 15(1): 85-96, 2015 Feb.
Article in English | MEDLINE | ID: mdl-24356728

ABSTRACT

The unfolded protein response (UPR) is an essential pathway for both normal and malignant plasma cells to maintain endoplasmic reticulum (ER) homeostasis in response to the large amount of immunoglobulin (Ig) output. The inositol-requiring enzyme 1-X-box binding protein-1 (IRE1-XBP-1) arm of the UPR pathway has been shown to play crucial roles not only in relieving the ER stress by up-regulating a series of genes favoring ER-associated protein degradation and protein folding, but in mediating terminal plasmacytic differentiation and maturation. Myeloma cells comprise various subsets arrested in diverse differentiated phases, and the immaturity of myeloma cells has been taken as a marker for poor prognosis, suggesting that differentiation induction would be a promising therapeutic strategy for myeloma. Herein, we used low-dose pharmacological UPR inducers such as tunicamycin (TM) and dithiothreitol (DTT) to efficiently activate the IRE1-XBP-1 pathway in myeloma cells characterized by transcriptional expression increase in spliced XBP-1 and molecular chaperons, accompanied by significant differentiation and maturation of these myeloma cells, without concomitant cytotoxicity. These differentiated myeloma cells exhibited a more mature appearance with well-developed cytoplasm and a reduced nucleocytoplasmic ratio, and a further differentiated phenotype with markedly increased expression of CD49e together with significantly elevated cellular secretion of Ig light chain as shown by flow cytometry and ELISA, in contrast to the control myeloma cells without exposed to TM or DTT. Moreover, siRNA knockdown of XBP-1 disrupted TM- or DTT-induced myeloma cell differentiation and maturation. Our study, for the first time, validated that the modest activation of the UPR pathway enables myeloma cells to further differentiate, and identified that XBP-1 plays an indispensable role in UPR-mediated myeloma cell differentiation and maturation. Thus, we provided the rationale and feasibility for the exploration of the novel therapeutic strategy of differentiation induction for plasmacytic malignancies.


Subject(s)
DNA-Binding Proteins/genetics , Gene Expression Regulation, Neoplastic , Multiple Myeloma/genetics , Transcription Factors/genetics , Adult , Aged , Cell Differentiation , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Dithiothreitol/pharmacology , Female , Humans , Immunoglobulin Light Chains/genetics , Immunoglobulin Light Chains/metabolism , Integrin alpha5/genetics , Integrin alpha5/metabolism , Male , Middle Aged , Multiple Myeloma/metabolism , Multiple Myeloma/pathology , Plasma Cells/drug effects , Plasma Cells/metabolism , Plasma Cells/pathology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Regulatory Factor X Transcription Factors , Signal Transduction , Transcription Factors/antagonists & inhibitors , Transcription Factors/metabolism , Tunicamycin/pharmacology , Unfolded Protein Response/drug effects , Unfolded Protein Response/genetics , X-Box Binding Protein 1
20.
Biomaterials ; 35(36): 9689-97, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25176069

ABSTRACT

Simultaneous in vivo luminescence and X-ray bioimaging in a tissue or animal integrates the advantages of each single-modal imaging technology, and will find widespread application in biological and clinical fields. However, synergistic dual-modal bioimaging that utilizes a new generation of upconversion nanoprobes is still limited. In addition, investigations concentrated on in vivo biodistribution of these nanoprobes may contribute to diagnosis and treatment, but long-term in vivo tracking based on these nanoprobes is rarely reported. In this work, water-soluble NaLuF4: Yb/Er nanophosphors were prepared through modified one-pot simultaneous synthesis and surface modification method. Owing to the outstanding upconverting emissions and large X-ray absorption coefficient/K-edge value of Lu and doped Yb ions, the obtained nanoprobes were successfully used as luminescent nanoprobes and X-ray contrast agents for in vivo synergistic upconversion luminescence and X-ray bioimaging. The in vivo biodistribution of these nanoprobes were observed, and the results based on long-term tracking reveal that the as-prepared nanoprobes first aggregated in the lung of the mouse, transferred to the liver, and finally moved to the spleen.


Subject(s)
Erbium/chemistry , Fluorides/chemistry , Lutetium/chemistry , Polyethylene Glycols/chemistry , Ytterbium/chemistry , Animals , Erbium/pharmacokinetics , Fluorides/pharmacokinetics , Luminescence , Lutetium/pharmacokinetics , Mice , Optical Imaging , Polyethylene Glycols/pharmacokinetics , Radiography , X-Rays , Ytterbium/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL