Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 165.744
Filter
Add more filters








Publication year range
1.
Cancer Med ; 13(15): e70043, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39087856

ABSTRACT

BACKGROUND: Recent research indicates a positive correlation between DEP structural domain-containing 1B (DEPDC1B) and the cell cycle in various tumors. However, the role of DEPDC1B in the infiltration of the tumor immune microenvironment (TIME) remains unexplored. METHODS: We analyzed the differential expression and prognostic significance of DEPDC1B in colon adenocarcinoma (COAD) using the R package "limma" and the Gene Expression Profiling Interactive Analysis (GEPIA) website. Gene set enrichment analysis (GSEA) was employed to investigate the functions and interactions of DEPDC1B expression in COAD. Cell Counting Kit-8 (CCK-8) assays and colony formation assays were utilized to assess the proliferative function of DEPDC1B. Correlations between DEPDC1B expression and tumor-infiltrating immune cells, immune checkpoints, tumor mutational burden (TMB), and microsatellite instability (MSI) status were examined using Spearman correlation analysis and CIBERSORT. RESULTS: DEPDC1B was highly expressed in COAD. Elevated DEPDC1B expression was associated with lower epithelial-to-mesenchymal transition (EMT) and TNM stages, leading to a favorable prognosis. DEPDC1B mRNA was prominently expressed in COAD cell lines. CCK-8 and colony formation assays demonstrated that DEPDC1B inhibited the proliferation of COAD cells. Analysis using the CIBERSORT database and Spearman correlation revealed that DEPDC1B correlated with four types of tumor-infiltrating immune cells. Furthermore, high DEPDC1B expression was linked to the expression of PD-L1, CTLA4, SIGLEC15, PD-L2, TMB, and MSI-H. High DEPDC1B expression also indicated responsiveness to anti-PD-L1 immunotherapy. CONCLUSIONS: DEPDC1B inhibits the proliferation of COAD cells and positively regulates the cell cycle, showing a positive correlation with CCNB1 and PBK expression. DEPDC1B expression in COAD is associated with tumor-infiltrating immune cells, immune checkpoints, TMB, and MSI-H in the tumor immune microenvironment. This suggests that DEPDC1B may serve as a novel prognostic marker and a potential target for immunotherapy in COAD.


Subject(s)
Adenocarcinoma , Colonic Neoplasms , GTPase-Activating Proteins , Gene Expression Regulation, Neoplastic , Tumor Microenvironment , Humans , Colonic Neoplasms/genetics , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Colonic Neoplasms/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Prognosis , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Epithelial-Mesenchymal Transition/genetics , Epithelial-Mesenchymal Transition/immunology , Genes, Tumor Suppressor , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Microsatellite Instability , Male , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cyclin B1/genetics , Cyclin B1/metabolism , Female
2.
Cancer Med ; 13(15): e70075, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39087899

ABSTRACT

OBJECTIVES: To determine the imaging details and diagnostic information of the treatment response to neoadjuvant chemoradiotherapy (nCRT) of rectal adenocarcinoma at 9.4T magnetic resonance imaging (MRI) by ex vivo. METHODS: Fifteen cases with locally advanced rectal cancer (LARC) followed by radical surgery after nCRT between September 2022 and February 2023 were recruited. Resected specimens were fixed in a perfluoropolyether-filled test tube and scanned with a 3.0T and 9.4T MRI system ex vivo. The residual tumor depth and MRI-based tumor regression grade (TRG) were subjectively assessed and then compared with the pathological findings. RESULTS: The ex vivo 9.4T T2WI without fat suppression clearly differentiated tumor tissue, fibrosis and normal rectal wall, which clearly corresponded to the pathologic tissues of the rectal specimens. The TRG could be accurately assessed on ex vivo 9.4T images in 13/15 specimens (86.7%), while in 11/15 specimens (73.3%) on ex vivo 3.0T images. CONCLUSION: Ex vivo 9.4T MR imaging clearly displayed the components of rectal wall and proved excellent diagnostic performance for evaluating the treatment response to nCRT, which allow radiologists to understand and then assess more accurately the TRG of LARC after nCRT.


Subject(s)
Adenocarcinoma , Magnetic Resonance Imaging , Neoadjuvant Therapy , Rectal Neoplasms , Humans , Rectal Neoplasms/therapy , Rectal Neoplasms/diagnostic imaging , Rectal Neoplasms/pathology , Neoadjuvant Therapy/methods , Magnetic Resonance Imaging/methods , Male , Female , Adenocarcinoma/therapy , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Middle Aged , Aged , Adult , Treatment Outcome , Rectum/diagnostic imaging , Rectum/pathology , Rectum/surgery , Chemoradiotherapy/methods
3.
Asian J Endosc Surg ; 17(4): e13366, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39090801

ABSTRACT

Subtotal colectomy is often performed on patients with synchronous colorectal cancer. However, compared with colorectal anastomosis, ileorectal anastomosis with subtotal colectomy is more likely to result in bowel dysfunction. The Deloyers procedure is useful in preserving bowel function in a patient with synchronous colorectal cancer. An 87-year-old man presented with bloody stool. Colonoscopy showed masses in the cecum, transverse colon, rectosigmoid, and rectum above the peritoneal reflection. Computed tomography scan revealed no evidence of regional lymph node swelling and distant metastasis. Therefore, robot-assisted low anterior resection, laparoscopic extended left hemicolectomy, laparoscopic cecal resection, and diverting ileostomy were performed. The patient was discharged from the hospital without complications. There was no recurrence, and the patient did not have complaints such as urgency, fecal incontinence, and excretory dysfunction. Hence, minimally invasive coloproctectomy using the Deloyers procedure can be safe and useful in preserving postoperative bowel function in elderly patients.


Subject(s)
Colectomy , Humans , Male , Aged, 80 and over , Colectomy/methods , Robotic Surgical Procedures/methods , Colorectal Neoplasms/surgery , Colorectal Neoplasms/pathology , Laparoscopy/methods , Neoplasms, Multiple Primary/surgery , Neoplasms, Multiple Primary/pathology , Proctectomy/methods , Adenocarcinoma/surgery , Adenocarcinoma/pathology
4.
J Investig Med High Impact Case Rep ; 12: 23247096241262515, 2024.
Article in English | MEDLINE | ID: mdl-39087613

ABSTRACT

Studies have highlighted a potential link between malignancies and immunoglobulin A nephropathy (IgAN). In such studies, the treatment of malignancy improved the symptoms of IgAN. Here, we report a patient case involving a history of hypertension, tobacco use disorder, and chronic kidney disease (CKD) presenting with hematuria with acute renal failure secondary to IgAN per renal biopsy. Prompted by this association, a malignancy workup was performed including computed tomography (CT) body imaging and biopsies of mediastinal and cervical lymph nodes which revealed a metastatic adenocarcinoma. Current knowledge includes a general mechanism behind the development of IgAN that points toward glomerular deposition of tumor-specific immunoglobulin A (IgA) immunoglobulins. However, the association of IgAN and malignancy has no definitive management guidelines. This clinical case serves as an important contribution in the hopes of future development of guidelines regarding the surveillance and management of IgAN in the setting of malignancy.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Glomerulonephritis, IGA , Lung Neoplasms , Tomography, X-Ray Computed , Humans , Glomerulonephritis, IGA/complications , Glomerulonephritis, IGA/pathology , Glomerulonephritis, IGA/diagnosis , Lung Neoplasms/pathology , Lung Neoplasms/complications , Lung Neoplasms/diagnosis , Carcinoma, Non-Small-Cell Lung/complications , Carcinoma, Non-Small-Cell Lung/secondary , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/pathology , Male , Acute Kidney Injury/etiology , Middle Aged , Hematuria/etiology , Adenocarcinoma/secondary , Adenocarcinoma/complications , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Biopsy , Kidney/pathology
5.
BMC Cancer ; 24(1): 934, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090577

ABSTRACT

BACKGROUND: Due to the close proximity of the prostate and rectum, rectal toxicity remains a major problem in patient treated by radiotherapy for prostate adenocarcinoma. One method of increasing the distance between the prostate and the rectum is to use a spacer implanted into the rectoprostatic space. This report describes the long-term outcomes obtained with a new ballon spacer. METHODS: Patients treated with curative radiotherapy for low- or intermediate-risk prostate adenocarcinoma, who underwent insertion of the ProSpace® (BioProtect Ltd, Tzur Yigal, Israel) rectal-prostate balloon spacer, were included. The main objective was to evaluate the dosimetric benefit of the spacer for OARs. The secondary objectives were to evaluate the feasibility and tolerability of ProSpace® balloon placement and to evaluate its long-term therapeutic efficacy and tolerance. RESULTS: Between October 2013 and March 2015, 16 patients were enrolled in the Pasteur Clinic, Toulouse, France. The median follow-up was 85.5 months. From top to bottom, the space created was a mean of 16.3 mm (range: 11-20.5 mm) at the base of the prostate, 12.1 mm (range: 4-16 mm) at the middle and 8.9 mm at the apex (range: 5-15 mm). On average, rectal volumes receiving a dose of 70 Gy, 60 Gy and 50 Gy were significantly lower after balloon implantation: -4.81 cc (1.5 vs. 6.3; p < 0.0005), -8.08 cc (6.4 vs. 14.5; p = 0.002) and -9.06 cc (16.7 vs. 25.7; p = 0.003), respectively. There were significant differences in coverage after balloon implantation: Median V95% (p < 0.0005), median Dmin (p = 0.01) and median V98% (p < 0.001) were higher after balloon implantation. At 5 years, cumulative gastrointestinal toxicity was grade 1 in 6% (1/16 patients). No toxicity of grade 2 or higher was found. At 5 years, no urinary toxicity grade 3 or 4 toxicity was found. The QoL was not deteriorated. CONCLUSIONS: The use of the ProSpace® balloon seems to be well accepted by patients, allowing a double dosimetric gain: a decrease in doses received by the rectum and an improvement in the coverage of the high-risk PTV. The long-term gastrointestinal toxicity remains low and QoL is preserved in all treated patients.


Subject(s)
Prostatic Neoplasms , Rectum , Humans , Male , Prostatic Neoplasms/radiotherapy , Aged , Rectum/radiation effects , Middle Aged , Radiotherapy Dosage , Adenocarcinoma/radiotherapy , Prostate/radiation effects , Prostate/pathology , Aged, 80 and over , Treatment Outcome , Radiation Injuries/etiology , Radiation Injuries/prevention & control , Follow-Up Studies
6.
Cancer Imaging ; 24(1): 101, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39090668

ABSTRACT

OBJECTIVES: The roles of magnetic resonance imaging (MRI) -based radiomics approach and deep learning approach in cervical adenocarcinoma (AC) have not been explored. Herein, we aim to develop prognosis-predictive models based on MRI-radiomics and clinical features for AC patients. METHODS: Clinical and pathological information from one hundred and ninety-seven patients with cervical AC was collected and analyzed. For each patient, 107 radiomics features were extracted from T2-weighted MRI images. Feature selection was performed using Spearman correlation and random forest (RF) algorithms, and predictive models were built using support vector machine (SVM) technique. Deep learning models were also trained with T2-weighted MRI images and clinicopathological features through Convolutional Neural Network (CNN). Kaplan-Meier curve was analyzed using significant features. In addition, information from another group of 56 AC patients was used for the independent validation. RESULTS: A total of 107 radiomics features and 6 clinicopathological features (age, FIGO stage, differentiation, invasion depth, lymphovascular space invasion (LVSI), and lymph node metastasis (LNM) were included in the analysis. When predicting the 3-year, 4-year, and 5-year DFS, the model trained solely on radiomics features achieved AUC values of 0.659 (95%CI: 0.620-0.716), 0.791 (95%CI: 0.603-0.922), and 0.853 (95%CI: 0.745-0.912), respectively. However, the combined model, incorporating both radiomics and clinicopathological features, outperformed the radiomics model with AUC values of 0.934 (95%CI: 0.885-0.981), 0.937 (95%CI: 0.867-0.995), and 0.916 (95%CI: 0.857-0.970), respectively. For deep learning models, the MRI-based models achieved an AUC of 0.857, 0.777 and 0.828 for 3-year DFS, 4-year DFS and 5-year DFS prediction, respectively. And the combined deep learning models got a improved performance, the AUCs were 0.903. 0.862 and 0.969. In the independent test set, the combined model achieved an AUC of 0.873, 0.858 and 0.914 for 3-year DFS, 4-year DFS and 5-year DFS prediction, respectively. CONCLUSIONS: We demonstrated the prognostic value of integrating MRI-based radiomics and clinicopathological features in cervical adenocarcinoma. Both radiomics and deep learning models showed improved predictive performance when combined with clinical data, emphasizing the importance of a multimodal approach in patient management.


Subject(s)
Adenocarcinoma , Deep Learning , Magnetic Resonance Imaging , Radiomics , Uterine Cervical Neoplasms , Adult , Aged , Female , Humans , Middle Aged , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/pathology , Adenocarcinoma/surgery , Lymphatic Metastasis/diagnostic imaging , Magnetic Resonance Imaging/methods , Neoplasm Staging , Prognosis , Retrospective Studies , Uterine Cervical Neoplasms/diagnostic imaging , Uterine Cervical Neoplasms/pathology
7.
PLoS One ; 19(8): e0308051, 2024.
Article in English | MEDLINE | ID: mdl-39093890

ABSTRACT

Preclinical models that replicate patient tumours as closely as possible are crucial for translational cancer research. While in vitro cancer models have many advantages in assessing tumour response therapy, in vivo systems are essential to enable evaluation of the role of the tumour cell extrinsic factors, such as the tumour microenvironment and host immune system. The requirement for a functional immune system is particularly important given the current focus on immunotherapies. Therefore, we set out to generate an immunocompetent, transplantable model of colorectal cancer suitable for in vivo assessment of immune-based therapeutic approaches. Intestinal tumours from a genetically engineered mouse model, driven by expression of a Pik3ca mutation and loss of Apc, were transplanted into wild type C57BL/6 host mice and subsequently passaged to form a novel syngeneic transplant model of colorectal cancer. Our work confirms the potential to develop a panel of mouse syngeneic grafts, akin to human PDX panels, from different genetically engineered, or carcinogen-induced, mouse models. Such panels would allow the in vivo testing of new pharmaceutical and immunotherapeutic treatment approaches across a range of tumours with a variety of genetic driver mutations.


Subject(s)
Adenocarcinoma , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Mice, Inbred C57BL , Animals , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Mice , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/pathology , Adenomatous Polyposis Coli Protein/genetics , Phosphatidylinositol 3-Kinases/metabolism , Transplantation, Isogeneic , Mutation , Humans , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology
8.
Head Neck Pathol ; 18(1): 67, 2024 Aug 05.
Article in English | MEDLINE | ID: mdl-39101976

ABSTRACT

BACKGROUND: High-risk human papillomavirus (HR-HPV) infection has been increasingly recognized as a risk factor for sinonasal tract carcinomas. However the prevalence and prognostic significance of HPV-associated sinonasal carcinomas is not well known due to limited studies and inconsistency in HPV testing modalities in literatures. Morphologically, HPV-associated sinonasal carcinomas encompass a diverse group of tumors. HPV-associated sinonasal adenocarcinoma has not been reported. The purpose of this study was to determine the prevalence, morphologic spectrum and prognostic implication of HPV-associated sinonasal carcinomas. METHODS: This cohort included 153 sinonasal carcinomas. Tissue microarrays were constructed. P16 immunohistochemistry and HR-HPV E6/7 in-situ Hybridization (ISH) were performed. Carcinomas were deemed HPV-associated based on a positive ISH testing. Clinicopathologic data was collected. RESULTS: 28/153 (18%) sinonasal carcinomas were HPV-associated. HPV-associated carcinomas consisted of 26 (93%) squamous cell carcinomas and variants, 1 (3.5%) HPV-related multiphenotypic sinonasal carcinoma and 1 (3.5%) adenocarcinoma. The HPV-associated adenocarcinoma closely resembled HPV-associated endocervical adenocarcinoma morphologically. HPV-associated carcinomas occurred in 8 (29%) women and 20 (71%) men with a median age of 66 years old. HPV-associated carcinomas were predominantly located at nasal cavity. A trend toward improved overall survival and progression free survival in HPV-associated carcinomas patients was observed, yet without statistical significance. CONCLUSION: Our study identifies a novel HPV-associated sinonasal adenocarcinoma subtype, highlights the broad morphologic spectrum of HPV-associated sinonasal carcinomas, and supports routine p16 testing during pathology practice regardless of tumor subtype followed by a confirmatory HR-HPV testing. This practice is critical for studying the clinical behavior of HPV-associated sinonasal carcinomas.


Subject(s)
Papillomavirus Infections , Paranasal Sinus Neoplasms , Humans , Male , Female , Papillomavirus Infections/complications , Aged , Middle Aged , Paranasal Sinus Neoplasms/virology , Paranasal Sinus Neoplasms/pathology , Adult , Aged, 80 and over , Adenocarcinoma/virology , Adenocarcinoma/pathology
9.
Cell Mol Biol (Noisy-le-grand) ; 70(7): 100-105, 2024 Jul 28.
Article in English | MEDLINE | ID: mdl-39097890

ABSTRACT

Goosecoid (GSC), translated from a homeobox gene, is a protein that participates in metastasis of various cancers. Pancreatic adenocarcinoma (PAAD) is one of the deadliest malignancies associated with a poor diagnosis and prognosis. To develop new treatment target or biomarker for PAAD, this study intended to assess the effects and the molecular mechanism of GSC on PAAD metastasis. The expressive discrepancy of GSC in PAAD and normal tissues/cells was compared by both the quantitative PCR and western blot. The effects of GSC silencing and GSC over-expression on PAAD cells and TGF-ß signaling were proved by wound-healing assay, cell counting kit-8, Transwell assay and western blot. From the results, GSC mRNA and protein levels were enriched in PAAD cancer tissues and cells. GSC silencing prohibited metastasis of PAAD cells including the ability to invade, migrate and epithelial-mesenchymal transition (EMT), whereas GSC upregulation stimulated these cells behaviors above. GSC silencing reversed the effects on cellular processes induced by activation of the TGF-ß pathway. Furthermore, silencing of GSC postponed tumor growth in xenograft model. In summary, GSC was abundantly expressed in PAAD, which activated the TGF-ß pathway to enhance cell metastasis and tumor development.


Subject(s)
Adenocarcinoma , Epithelial-Mesenchymal Transition , Neoplasm Metastasis , Pancreatic Neoplasms , Signal Transduction , Transforming Growth Factor beta , Animals , Humans , Mice , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Gene Silencing , Intercellular Signaling Peptides and Proteins , Mice, Inbred BALB C , Mice, Nude , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/genetics , Transforming Growth Factor beta/metabolism
11.
Front Endocrinol (Lausanne) ; 15: 1217250, 2024.
Article in English | MEDLINE | ID: mdl-39104815

ABSTRACT

Background: Gallbladder mixed neuroendocrine-non-neuroendocrine neoplasm generally consists of a gallbladder neuroendocrine tumor and a non-neuroendocrine component. The World Health Organization (WHO) in 2019 established a guideline requiring each component, both neuroendocrine and non-neuroendocrine, to account for a minimum of 30% of the tumor mass. Methods: Patients after surgery resection and diagnosed at microscopy evaluation with pure gallbladder neuroendocrine carcinoma (GBNEC), gallbladder mixed adeno-neuroendocrine carcinoma (GBMANEC, GBNEC≥30%), and gallbladder carcinoma mixed with a small fraction of GBNEC (GBNEC <30%) between 2010 and 2022 at West China Hospital of Sichuan University were collated for the analyses. Demographic features, surgical variables, and tumor characteristics were evaluated for association with patients' overall and recurrence-free survival (OS and RFS). Results: The study included 26 GBNEC, 11 GBMANEC, 4 gallbladder squamous-cell carcinoma (GBSCC), and 7 gallbladder adenocarcinoma (GBADC) mixed with a small fraction of GBNEC. All patients had stage III or higher tumors (AJCC8th edition). The majority of included patients (79.17%) underwent curative surgical resection (R0), with only ten patients having tumoral resection margins. In the analysis comparing patients with GBNEC percentage (GBNEC≥30% vs. GBNEC<30%), the basic demographics and tumor characteristics of most patients were comparable. The prognosis of these patients was also comparable, with a median OS of 23.65 months versus 20.40 months (P=0.13) and a median RFS of 17.1 months versus 12.3 months (P=0.24). However, patients with GBADC or GBSCC mixed with GBNEC <30% had a statistically significant decreased OS and RFS (both P<0.0001)) compared with GBNEC and GBMANEC. Patients with GBNEC who exhibited advanced tumor stages and lymphovascular invasion had a higher risk of experiencing worse overall survival (OS) and recurrence-free survival (RFS). However, a 30% GBNEC component was not identified as an independent risk factor. Conclusion: Patients with GBNEC were frequently diagnosed at advanced stages and their prognosis is poor. The 30% percentage of the GBNEC component is not related to the patient's survival.


Subject(s)
Carcinoma, Neuroendocrine , Gallbladder Neoplasms , Humans , Gallbladder Neoplasms/pathology , Gallbladder Neoplasms/mortality , Gallbladder Neoplasms/surgery , Female , Male , Middle Aged , Prognosis , Carcinoma, Neuroendocrine/pathology , Carcinoma, Neuroendocrine/mortality , Carcinoma, Neuroendocrine/surgery , Carcinoma, Neuroendocrine/diagnosis , Aged , Adult , Retrospective Studies , Adenocarcinoma/pathology , Adenocarcinoma/mortality , Adenocarcinoma/surgery , Survival Rate
12.
Int Braz J Urol ; 50(5): 659-660, 2024.
Article in English | MEDLINE | ID: mdl-39106120

ABSTRACT

OBJECTIVE: We present a novel technique to perform single-port (SP) robot-assisted partial cystectomy with excision of the urachal remnant and bilateral pelvic lymph node dissection for urachal adenocarcinoma (1-7). MATERIALS AND METHODS: A 41-year-old male presented to the clinic for multiple episodes of hematuria and mucousuria. Office cystoscopy revealed a small solitary tumor at the dome of the bladder, with a diagnostic bladder biopsy revealing a tubule-villous bladder adenoma. Cross-sectional imaging of the chest/abdomen/pelvis revealed a 4.5 cm cystic mass arising from the urachus without evidence of local invasion and metastatic spread. He underwent SP robotic-assisted partial cystectomy with excision of the urachal remnant and bilateral pelvic lymph node dissection. Surgical steps include: 1) peritoneal incision to release the urachus and drop bladder 2) identification of urachal tumor 3) intraoperative live cystoscopic identification of bladder mass and scoring of tumor margins using Toggle Pro feature 4) tumor excision with partial cystectomy 5) cystorrhaphy 6) bilateral pelvic lymph node dissection 7) peritoneal interposition flap to mitigate lymphocele formation. RESULTS: Surgery was successful, with no intraoperative complications, an operative time of 100 minutes, and estimated blood loss of 20 mL. The patient was discharged on post-op day one, and the Foley catheter removed one week after surgery. Final pathology revealed a 7.5 cm infiltrating urachal muscle-invasive adenocarcinoma of the bladder (pT2b). Negative surgical margins were achieved. CONCLUSIONS: Single-port robot-assisted partial cystectomy for urachal adenocarcinoma is safe and can achieve equivalent oncologic outcomes to the standard of care with minimally invasive and open techniques.


Subject(s)
Adenocarcinoma , Cystectomy , Lymph Node Excision , Robotic Surgical Procedures , Urinary Bladder Neoplasms , Humans , Male , Adenocarcinoma/surgery , Adenocarcinoma/pathology , Robotic Surgical Procedures/methods , Cystectomy/methods , Urinary Bladder Neoplasms/surgery , Urinary Bladder Neoplasms/pathology , Adult , Lymph Node Excision/methods , Treatment Outcome , Operative Time , Reproducibility of Results
13.
Medicine (Baltimore) ; 103(31): e39189, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39093786

ABSTRACT

INTRODUCTION AND IMPORTANCE: Lung adenocarcinoma may resemble the clinical presentation of an infectious or inflammatory lung disease. The coexistence of lung cancer, and polyserous effusions is uncommon, which may cause a diagnostic challenge. However, any polyserous effusions at a young age must always be suspicious for malignancy. CASE PRESENTATION: We report a case of 38-year-old male patient with polyserous effusions and pneumonia who was treated accordingly and showed clinical improvement with a significant reduction of pericardial and pleural effusions. Subsequent testing and a biopsy resulted in the histopathological diagnosis of an adenocarcinoma of the lung. CLINICAL DISCUSSION: Nonrecurrent polyserous effusions in lung adenocarcinoma are uncommon, and negative cytology results may not exclude malignancy due to the moderate sensitivity of pleural and pericardial fluid cytology. Clinicians should remain vigilant for false-negative results, especially in younger patients. Malignancy should not be ruled out because pleural and pericardial fluid cytology have a sensitivity of 60% and 92%, respectively. CONCLUSION: Our case highlights the diagnostic challenges posed by atypical presentations of lung adenocarcinoma and emphasizes the importance of considering malignancy in the differential diagnosis of polyserous effusions, even when initial cytology results are negative. Clarifying the rationale for this study enhances its relevance and impact.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Pneumonia , Humans , Male , Adult , Diagnosis, Differential , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Lung Neoplasms/complications , Adenocarcinoma of Lung/diagnosis , Adenocarcinoma of Lung/pathology , Pneumonia/diagnosis , Pericardial Effusion/etiology , Pericardial Effusion/diagnosis , Pericardial Effusion/pathology , Pleural Effusion/diagnosis , Pleural Effusion/etiology , Adenocarcinoma/diagnosis , Adenocarcinoma/pathology , Pleural Effusion, Malignant/pathology , Pleural Effusion, Malignant/diagnosis , Pleural Effusion, Malignant/etiology , Cytology
14.
BMC Cancer ; 24(1): 941, 2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39095759

ABSTRACT

BACKGROUND: Advanced pancreatic adenocarcinoma lacks effective treatment options, and systemic gemcitabine-based chemotherapy offers only marginal survival benefits at the cost of significant toxicities and adverse events. New therapeutic options with better drug availability are warranted. This study aims to evaluate the safety and efficacy of digital subtraction angiography (DSA)-guided pancreatic arterial infusion (PAI) versus intravenous chemotherapy (IVC) using the gemcitabine and oxaliplatin (GEMOX) regimen in unresectable locally advanced or metastatic pancreatic cancer (PC) patients. MATERIALS AND METHODS: This study prospectively enrolled 51 eligible treatment-naive patients with unresectable PC to receive GEMOX treatment via PAI or IVC (1:1 ratio randomization) from December 2015 to December 2019. Cycles were repeated monthly, and each process consisted of two treatments administered bi-weekly. Overall survival (OS), progression-free survival (PFS), objective response rate (ORR), disease control rate (DCR), 1-year survival, 6-month survival, tumor-site subgroup survival, and incidences of adverse events were compared. RESULTS: The median OS of the PAI and IVC groups were 9.93 months and 10.07 months, respectively (p = 0.3049). The median PFS of the PAI and IVC groups were 5.07 months and 4.23 months (p = 0.1088). No significant differences were found in the ORR (11.54% vs. 4%, p = 0.6312), DCR (53.85% vs. 44%, p = 0.482), and 1-year OS rate (44% vs. 20.92%, p = 0.27) in PAI and IVC groups. The 6-month OS rate was significantly higher in the PAI group (100%) than in the IVC group (83.67%) (p = 0.0173). The median OS of patients in PAI group with pancreatic head and neck tumors were significantly higher than those of body and tail tumors (12.867 months vs. 9 months, p = 0.0214). The incidences of hematologic disorders, liver function disorders, and digestive disorders in the IVC group were higher than in the PAI group (p < 0.05). CONCLUSION: GEMOX PAI therapy presented a higher 6-month OS rate and fewer adverse events than IVC in advanced pancreatic adenocarcinoma patients. Those with pancreatic head and neck tumors may yield a superior treatment outcome from PAI treatment. TRIAL REGISTRATION NUMBER: NCT02635971. DATE OF REGISTRATION: 21/12/2015.


Subject(s)
Adenocarcinoma , Angiography, Digital Subtraction , Antineoplastic Combined Chemotherapy Protocols , Deoxycytidine , Pancreatic Neoplasms , Humans , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/mortality , Male , Female , Middle Aged , Deoxycytidine/analogs & derivatives , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Adenocarcinoma/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Infusions, Intra-Arterial , Adult , Prospective Studies , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Gemcitabine , Infusions, Intravenous , Pancreas/pathology , Pancreas/diagnostic imaging , Organoplatinum Compounds
15.
Clin Nucl Med ; 49(9): e482-e483, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39086049

ABSTRACT

ABSTRACT: Relapsing polychondritis (RP) is an uncommon autoimmune disease that causes inflammation of the cartilage and proteoglycan-rich structures, including the ear, nose, and airway. Paraneoplastic RP is a subset of RP that occurs in some individuals following the detection and treatment of certain types of cancers. FDG PET/CT helps with early diagnosis of RP, identifying inflammatory areas even in the absence of symptoms, and guiding the selection of appropriate biopsy sites. Here, we present a case of adenocarcinoma of the lung presenting with paraneoplastic symptoms of RP as initial presentation, and symptoms were resolved after 3 cycles of chemotherapy.


Subject(s)
Adenocarcinoma of Lung , Adenocarcinoma , Fluorodeoxyglucose F18 , Lung Neoplasms , Polychondritis, Relapsing , Positron Emission Tomography Computed Tomography , Humans , Polychondritis, Relapsing/diagnostic imaging , Polychondritis, Relapsing/complications , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/complications , Adenocarcinoma of Lung/diagnostic imaging , Adenocarcinoma of Lung/complications , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/complications , Paraneoplastic Syndromes/diagnostic imaging , Male , Middle Aged , Tomography, X-Ray Computed
16.
JAMA Netw Open ; 7(8): e2425581, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39093560

ABSTRACT

Importance: The prognosis of patients with adenocarcinoma of the esophagus and esophagogastric junction (AEG) is poor. From current evidence, it remains unclear to what extent preoperative chemoradiotherapy (CRT) or preoperative and/or perioperative chemotherapy achieve better outcomes than surgery alone. Objective: To assess the association of preoperative CRT and preoperative and/or perioperative chemotherapy in patients with AEG with overall survival and other outcomes. Data Sources: Literature search in PubMed, Cochrane Library, Cumulative Index to Nursing and Allied Health Literature, ClinicalTrials.gov, and International Clinical Trials Registry Platform was performed from inception to April 21, 2023. Study Selection: Two blinded reviewers screened for randomized clinical trials comparing preoperative CRT plus surgery with preoperative and/or perioperative chemotherapy plus surgery, 1 intervention with surgery alone, or all 3 treatments. Only data from participants with AEG were included from trials that encompassed mixed histology or gastric cancer. Among 2768 initially identified studies, 17 (0.6%) met the selection criteria. Data Extraction and Synthesis: The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) reporting guidelines were followed for extracting data and assessing data quality by 2 independent extractors. A bayesian network meta-analysis was conducted using the 2-stage approach. Main Outcomes and Measures: Overall and disease-free survival, postoperative morbidity, and mortality. Results: The analyses included 2549 patients (2206 [86.5%] male; mean [SD] age, 61.0 [9.4] years) from 17 trials (conducted from 1989-2016). Both preoperative CRT plus surgery (hazard ratio [HR], 0.75 [95% credible interval (CrI), 0.62-0.90]; 3-year difference, 105 deaths per 1000 patients) and preoperative and/or perioperative chemotherapy plus surgery (HR, 0.78 [95% CrI, 0.64-0.91]; 3-year difference, 90 deaths per 1000 patients) showed longer overall survival than surgery alone. Comparing the 2 modalities yielded similar overall survival (HR, 1.04 [95% CrI], 0.83-1.28]; 3-year difference, 15 deaths per 1000 patients fewer for CRT). Similarly, disease-free survival was longer for both modalities compared with surgery alone. Postoperative morbidity was more frequent after CRT plus surgery (odds ratio [OR], 2.94 [95% CrI, 1.01-8.59]) than surgery alone. Postoperative mortality was not significantly more frequent after CRT plus surgery than surgery alone (OR, 2.50 [95% CrI, 0.66-10.56]) or after chemotherapy plus surgery than CRT plus surgery (OR, 0.44 [95% CrI, 0.08-2.00]). Conclusions and Relevance: In this meta-analysis of patients with AEG, both preoperative CRT and preoperative and/or perioperative chemotherapy were associated with longer survival without relevant differences between the 2 modalities. Thus, either of the 2 treatments may be recommended to patients.


Subject(s)
Adenocarcinoma , Chemoradiotherapy , Esophageal Neoplasms , Esophagogastric Junction , Network Meta-Analysis , Stomach Neoplasms , Humans , Adenocarcinoma/therapy , Adenocarcinoma/mortality , Esophagogastric Junction/pathology , Esophageal Neoplasms/therapy , Esophageal Neoplasms/mortality , Chemoradiotherapy/methods , Stomach Neoplasms/therapy , Stomach Neoplasms/mortality , Stomach Neoplasms/drug therapy , Male , Preoperative Care/methods , Middle Aged , Female , Aged , Disease-Free Survival
17.
J Cancer Res Clin Oncol ; 150(8): 381, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097562

ABSTRACT

BACKGROUND: High-grade non-intestinal-type sinonasal adenocarcinoma (non-ITAC) is a rare and aggressive form of adenocarcinoma with poor prognosis. The current standard treatment approach involves surgery combined with radiation therapy. However, there is a need for exploring additional treatment modalities to improve patient outcomes. CASE PRESENTATION: We present a case of a 65-year-old male patient who presented with pain in the right maxillary sinus and was diagnosed with high-grade non-ITAC following surgery. Postoperative pathology revealed tumor invasion into bone tissue and vascular invasion, necessitating further treatment. The patient underwent radiation therapy, followed by immunotherapy with carilizumab combined with chemotherapy. During the maintenance immunotherapy period, tumor progression was observed, and genetic testing identified EGFR and TP53 mutations. Consequently, the patient was treated with gefitinib, a targeted therapy drug. Notably, the patient's lung metastases showed a gradual reduction in size, indicating a favorable treatment response. The patient is currently undergoing oral treatment with gefitinib. CONCLUSIONS: This case report highlights the potential benefit of combining immunotherapy and targeted therapy in the treatment of high-grade non-ITAC. Despite the rarity of this cancer type, this approach may offer an alternative treatment strategy for patients with this aggressive disease. We hope that this case can contribute to a deeper understanding of high-grade non-ITAC and promote the application of immunotherapy and targeted therapy in improving survival rates for patients with this condition.


Subject(s)
Adenocarcinoma , Humans , Male , Aged , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Adenocarcinoma/drug therapy , Maxillary Sinus Neoplasms/pathology , Maxillary Sinus Neoplasms/therapy , Maxillary Sinus Neoplasms/drug therapy , Molecular Targeted Therapy , Immunotherapy/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/administration & dosage , Gefitinib/therapeutic use , Maxillary Sinus/pathology , Paranasal Sinus Neoplasms/pathology , Paranasal Sinus Neoplasms/therapy , Paranasal Sinus Neoplasms/drug therapy , Neoplasm Grading
18.
Clin Epigenetics ; 16(1): 102, 2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39097736

ABSTRACT

BACKGROUND: Oesophageal cancer remains a challenging disease with high mortality rates and few therapeutic options. In view of these difficulties, epigenetic drugs have emerged as potential alternatives for patient care. The goal of this study was to evaluate the effect and biological consequences of Panobinostat treatment, an HDAC (histone deacetylase) inhibitor already approved for treatment of patients with multiple myeloma, in oesophageal cell lines of normal and malignant origin, with the latter being representative of the two main histological subtypes: adenocarcinoma and squamous cell carcinoma. RESULTS: Panobinostat treatment inhibited growth and hindered proliferation, colony formation and invasion of oesophageal cancer cells. Considering HDAC tissue expression, HDAC1 was significantly upregulated in normal oesophageal epithelium in comparison with tumour tissue, whereas HDAC3 was overexpressed in oesophageal cancer compared to non-malignant mucosa. No differences between normal and tumour tissue were observed for HDAC2 and HDAC8 expression. CONCLUSIONS: Panobinostat exposure effectively impaired malignant features of oesophageal cancer cells. Because HDAC3 was shown to be overexpressed in oesophageal tumour samples, this epigenetic drug may represent an alternative therapeutic option for oesophageal cancer patients.


Subject(s)
Adenocarcinoma , Carcinoma, Squamous Cell , Cell Proliferation , Esophageal Neoplasms , Histone Deacetylase Inhibitors , Histone Deacetylases , Panobinostat , Humans , Panobinostat/pharmacology , Panobinostat/therapeutic use , Panobinostat/administration & dosage , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Cell Line, Tumor , Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Cell Proliferation/drug effects , Carcinoma, Squamous Cell/drug therapy , Carcinoma, Squamous Cell/genetics , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Repressor Proteins/genetics , Hydroxamic Acids/pharmacology , Hydroxamic Acids/therapeutic use , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Indoles/pharmacology , Indoles/therapeutic use , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology
19.
BMC Cancer ; 24(1): 979, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118022

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a major contributor to cancer-related mortality. Glycolysis plays a pivotal role in tumor microenvironment (TME) reprogramming. In this research, the functions of glycolysis-associated genes (GRGs) were evaluated to predict the outcome and reveal the characteristics of the immune microenvironment in individuals with stomach cancer. METHODS: The Cancer Genome Atlas (TCGA)-stomach adenocarcinoma (STAD) cohort provided gene expression and clinical data for gastric cancer (GC) patients, which were further authenticated using datasets sourced from the Gene Expression Omnibus (GEO). By referencing the Molecular Signatures Database (MSigDB), a total of 326 GRGs were pinpointed. The various subtypes of GC were outlined through consensus clustering, derived from the expression patterns of these GRGs. Utilizing multivariate Cox regression analysis, a multigene risk score model was formulated. Both the CIBERSORT and ESTIMATE algorithms played a pivotal role in assessing the immune microenvironment. To delve into the biological functions of the key genes, wound healing, transwell invasion, and MTT assays were conducted. RESULTS: Based on the expression patterns of GRGs, patients were categorized into two distinct groups: the metabolic subtype, designated as cluster A, and the immune subtype, labeled as cluster B. Patients belonging to cluster B exhibited a poorer prognosis. A prognostic risk score model, formulated upon the expression levels of six key GRGs - ME1, PLOD2, NUP50, CXCR4, SLC35A3, and SRD35A3 - emerged as a viable tool for predicting patient outcomes. The downregulation of CXCR4 notably diminished the glycolytic capacity of gastric cancer (GC) cells, alongside their migratory, invasive, and proliferative capabilities. Intriguingly, despite the adverse prognostic implications associated with both the immune subtype (cluster B) and the high-risk cohort, these groups exhibited a favorable immune microenvironment coupled with elevated expression of immune checkpoint genes. Our investigations revealed a positive correlation between high CXCR4 expression and low ME1 expression with the infiltration of CD8+ T cells, as well as an enhanced responsiveness to treatment with an anti-PD-1 immune checkpoint inhibitor. CONCLUSIONS: In this study, we discovered that the expression profiles of GRGs hold the potential to forecast the prognosis of gastric cancer (GC) patients, thereby possibly aiding in clinical treatment decision-making.


Subject(s)
Glycolysis , Stomach Neoplasms , Tumor Microenvironment , Stomach Neoplasms/genetics , Stomach Neoplasms/immunology , Stomach Neoplasms/pathology , Stomach Neoplasms/mortality , Humans , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Prognosis , Glycolysis/genetics , Gene Expression Regulation, Neoplastic , Male , Biomarkers, Tumor/genetics , Female , Gene Expression Profiling , Middle Aged , Adenocarcinoma/genetics , Adenocarcinoma/immunology , Adenocarcinoma/pathology , Cell Line, Tumor
20.
BMC Cancer ; 24(1): 978, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39118103

ABSTRACT

BACKGROUND: The unfolded protein response (UPR) is associated with immune cells that regulate the biological behavior of tumors. This article aims to combine UPR-associated genes with immune cells to find a prognostic marker and to verify its connection to the UPR. METHODS: Univariate cox analysis was used to screen prognostically relevant UPRs and further screened for key UPRs among them by machine learning. ssGSEA was used to calculate immune cell abundance. Univariate cox analysis was used to screen for prognostically relevant immune cells. Multivariate cox analysis was used to calculate UPR_score and Tumor Immune Microenvironment score (TIME_score). WGCNA was used to screen UPR-Immune-related (UI-related) genes. Consensus clustering analysis was used to classify patients into molecular subtype. Based on the UI-related genes, we classified colon adenocarcinoma (COAD) samples by cluster analysis. Single-cell analysis was used to analyze the role of UI-related genes. We detected the function of TIMP1 by cell counting and transwell. Immunoblotting was used to detect whether TIMP1 was regulated by key UPR genes. RESULTS: Combined UPR-related genes and immune cells can determine the prognosis of COAD patients. Cluster analysis showed that UI-related genes were associated with clinical features of COAD. Single-cell analysis revealed that UI-related genes may act through stromal cells. We defined three key UI-related genes by machine learning algorithms. Finally, we found that TIMP1, regulated by key genes of UPR, promoted colon cancer proliferation and metastasis. CONCLUSIONS: We found that TIMP1 was a prognostic marker and experimentally confirmed that TIMP1 was regulated by key genes of UPR.


Subject(s)
Biomarkers, Tumor , Colonic Neoplasms , Tissue Inhibitor of Metalloproteinase-1 , Tumor Microenvironment , Unfolded Protein Response , Humans , Unfolded Protein Response/genetics , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/mortality , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Prognosis , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Tissue Inhibitor of Metalloproteinase-1/genetics , Tissue Inhibitor of Metalloproteinase-1/metabolism , Gene Expression Regulation, Neoplastic , Cluster Analysis , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Machine Learning , Single-Cell Analysis/methods , Female , Cell Line, Tumor , Male
SELECTION OF CITATIONS
SEARCH DETAIL