Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 6.351
Filter
1.
Adv Surg ; 58(1): 49-64, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39089786

ABSTRACT

Colorectal cancer (CRC) with peritoneal metastases is a complex disease and its management presents significant clinical challenges. In well-selected patients at experienced centers, CRS/hyperthermic intraperitoneal chemotherapy (HIPEC) can be performed with acceptable morbidity and is associated with prolonged survival. Based on the results of recent randomized controlled trials, HIPEC using oxaliplatin after CRS with shortened perfusion periods (30 minutes) is no longer recommended. There is a movement toward utilizing mitomycin C as a first-line intraperitoneal agent with extended perfusion times (90-120 minutes); however, there is currently little prospective evidence to support its widespread use.


Subject(s)
Colonic Neoplasms , Hyperthermic Intraperitoneal Chemotherapy , Mitomycin , Peritoneal Neoplasms , Humans , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/drug therapy , Colonic Neoplasms/pathology , Colonic Neoplasms/therapy , Mitomycin/administration & dosage , Oxaliplatin/administration & dosage , Antineoplastic Agents/administration & dosage , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Cytoreduction Surgical Procedures , Treatment Outcome
2.
Cancer Med ; 13(15): e70037, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39109683

ABSTRACT

BACKGROUND: Gastric cancer (GC), particularly for advanced stage of GC, commonly undergoes peritoneal metastasis (PM), which is the leading cause of GC-related death. However, there currently has no reliable biomarker to predict the onset of GCPM. It is well known that the imbalance of gut microbiota contributes to the development and metastasis of gastrointestinal tumors. Unfortunately, little is known about how the alternation in gut microbiota is associated with the onset of GCPM. METHODS: Our current study analyzed structural characteristics and functional prediction of gut microbiota in GC patients with PM (PM group) and without PM (non-PM group). Fresh fecal samples were collected from a discovery cohort (PM = 38, non-PM = 54) and a validation cohort (PM = 15, non-PM = 21) of GC patients and their 16S ribosomal RNA (16s rRNA) gene amplicons were sequenced, followed by bioinformatics. RESULTS: The results indicated an increase in the biodiversity of gut microbiota in the non-PM group of the discovery cohort, compared with the PM group. Moreover, LEfSe analysis found 31 significantly different microorganisms, of which the Roseburia ranked the fifth in the random forest (RF) model. The characteristics of intestinal microbiota in GCPM patients were changed, and the abundance of Roseburia in gut microbiota from the GCPM patients was reduced and receiver operating characteristic (ROC) analysis revealed that the reduced abundance of gut Roseburia effectively predicted the onset of GCPM. CONCLUSION: This signature was also observed in the validation cohort. Therefore, Roseburia is a protective microbial marker and the reduced abundance of Roseburia in gut microbiota may help early diagnosis of GCPM.


Subject(s)
Feces , Gastrointestinal Microbiome , Peritoneal Neoplasms , RNA, Ribosomal, 16S , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/microbiology , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/microbiology , Male , Female , Middle Aged , RNA, Ribosomal, 16S/genetics , Feces/microbiology , Biomarkers, Tumor/genetics , Aged , Clostridiales/isolation & purification , Clostridiales/genetics
3.
Sci Rep ; 14(1): 19103, 2024 08 17.
Article in English | MEDLINE | ID: mdl-39154083

ABSTRACT

Peritoneal recurrence (PR) in gastric cancer after curative resection has poor prognosis. Therefore, we aimed to construct a nomogram to predict PR, and establish PR score for risk stratification to guide adjuvant chemotherapy. A total of 315 patients with gastric cancer after radical surgery were included, and randomly stratified into training group (n = 221) and validation group (n = 94). Univariate and multivariate analyses were used to determine predictive factors of PR. The nomogram was constructed to predict the risk of PR. We utilized the time-dependent area under the receiver operating characteristic (ROC) curves (AUCs), calibration curves, and decision curve analysis (DCA) to evaluate the performance of the nomogram. Multivariate analysis showed that tumor site, N stage, preoperative CEA, and postoperative CA199 were independent predictors of PR. A nomogram was constructed to predict PR based on these factors. The AUC value was 0.755 in the training group and 0.715 in the validation group. The calibration curves showed good agreement between prediction and observation in the training and validation groups. The decision curve analysis displayed a good net benefit of the nomogram. The novel PR score was developed and patients were stratified into the low-, medium-, and high -risk groups. For the high-risk group, postoperative adjuvant chemotherapy significantly improved patients' overall survival (OS) and disease-free survival (DFS). The establishment of nomogram facilitates the prediction of PR after radical gastrectomy, and a novel PR score may help guide adjuvant chemotherapy for gastric cancer.


Subject(s)
Neoplasm Recurrence, Local , Nomograms , Peritoneal Neoplasms , Stomach Neoplasms , Humans , Stomach Neoplasms/surgery , Stomach Neoplasms/pathology , Stomach Neoplasms/mortality , Male , Female , Middle Aged , Peritoneal Neoplasms/surgery , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/mortality , Aged , Risk Assessment/methods , ROC Curve , Prognosis , Gastrectomy , Chemotherapy, Adjuvant , Adult , Risk Factors
4.
Sci Rep ; 14(1): 16208, 2024 07 13.
Article in English | MEDLINE | ID: mdl-39003337

ABSTRACT

The study aims to investigate the predictive capability of machine learning algorithms for omental metastasis in locally advanced gastric cancer (LAGC) and to compare the performance metrics of various machine learning predictive models. A retrospective collection of 478 pathologically confirmed LAGC patients was undertaken, encompassing both clinical features and arterial phase computed tomography images. Radiomic features were extracted using 3D Slicer software. Clinical and radiomic features were further filtered through lasso regression. Selected clinical and radiomic features were used to construct omental metastasis predictive models using support vector machine (SVM), decision tree (DT), random forest (RF), K-nearest neighbors (KNN), and logistic regression (LR). The models' performance metrics included accuracy, area under the curve (AUC) of the receiver operating characteristic curve, sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV). In the training cohort, the RF predictive model surpassed LR, SVM, DT, and KNN in terms of accuracy, AUC, sensitivity, specificity, PPV, and NPV. Compared to the other four predictive models, the RF model significantly improved PPV. In the test cohort, all five machine learning predictive models exhibited lower PPVs. The DT model demonstrated the most significant variation in performance metrics relative to the other models, with a sensitivity of 0.231 and specificity of 0.990. The LR-based predictive model had the lowest PPV at 0.210, compared to the other four models. In the external validation cohort, the performance metrics of the predictive models were generally consistent with those in the test cohort. The LR-based model for predicting omental metastasis exhibited a lower PPV. Among the machine learning algorithms, the RF predictive model demonstrated higher accuracy and improved PPV relative to LR, SVM, KNN, and DT models.


Subject(s)
Machine Learning , Omentum , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/diagnostic imaging , Male , Female , Omentum/pathology , Omentum/diagnostic imaging , Middle Aged , Retrospective Studies , Aged , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/diagnostic imaging , Tomography, X-Ray Computed/methods , Support Vector Machine , ROC Curve , Algorithms , Adult , Decision Trees , Radiomics
5.
Int J Mol Sci ; 25(13)2024 Jul 08.
Article in English | MEDLINE | ID: mdl-39000603

ABSTRACT

Although several studies have been completed to investigate the effect of cytoreductive surgery (CRS) with or without hyperthermic intraperitoneal chemotherapy (HIPEC) in endometrial cancer with peritoneal metastasis (ECPM), a direct comparison was not performed previously. A meta-analysis was performed to investigate the suspected additional survival benefits of CRS plus HIPEC over CRS only. Twenty-one and ten studies with a total number of 1116 and 152 cases investigating CRS only and CRS plus HIPEC were identified, respectively. When all articles were analyzed, the 1-year survival rate was 17.60% higher for CRS plus HIPEC (82.28% vs. 64.68%; p = 0.0102). The same tendency was observed for the 2-year (56.07% vs. 36.95%; difference: 19.12%; p = 0.0014), but not for the 5-year (21.88% vs. 16.45%; difference: 5.43%; p = 0.3918) survival rates. The same clinical significance, but statistically less strong observations, could be made if only the studies published after 2010 were investigated (1-year survival rate: 12.08% and p = 0.0648; 2-year survival rate: 10.90% and p = 0.0988). CRS remains one of the core elements of ECPM treatment, but the addition of HIPEC to CRS can increase the positive clinical outcome, especially in the first 2 years.


Subject(s)
Cytoreduction Surgical Procedures , Endometrial Neoplasms , Hyperthermic Intraperitoneal Chemotherapy , Peritoneal Neoplasms , Humans , Female , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/mortality , Endometrial Neoplasms/pathology , Endometrial Neoplasms/therapy , Endometrial Neoplasms/mortality , Cytoreduction Surgical Procedures/methods , Hyperthermic Intraperitoneal Chemotherapy/methods , Combined Modality Therapy , Survival Rate
6.
BMJ Case Rep ; 17(7)2024 Jul 22.
Article in English | MEDLINE | ID: mdl-39038876

ABSTRACT

Low-grade appendiceal mucinous neoplasm (LAMN) may culminate as a mucin-secreting disease known as pseudomyxoma peritonei (PMP). Once the diagnosis of LAMN and PMP is made, cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS and HIPEC) are indicated.Herein, we present a female patient in her 50s who was diagnosed with an ovarian mass for which she underwent laparoscopic oophorectomy. As the pathology of the ovary showed a tumour of gastrointestinal origin, she then underwent CRS and HIPEC with a final pathology of LAMN. Six weeks later, a mucinous lesion confined to the abdominal wall was detected on a postoperative CT. Suspected for port-site metastasis at the laparoscopic trocar site, we treated this lesion using the same principles of treatment as the intra-abdominal disease. The abdominal wall mass was surgically resected, and the cavity created was irrigated with mitomycin C. On 30 months of follow-up, the patient had no evidence of disease.


Subject(s)
Appendiceal Neoplasms , Cytoreduction Surgical Procedures , Hyperthermic Intraperitoneal Chemotherapy , Laparoscopy , Ovarian Neoplasms , Ovariectomy , Humans , Female , Ovarian Neoplasms/therapy , Ovarian Neoplasms/pathology , Middle Aged , Ovariectomy/adverse effects , Laparoscopy/adverse effects , Cytoreduction Surgical Procedures/adverse effects , Hyperthermic Intraperitoneal Chemotherapy/adverse effects , Appendiceal Neoplasms/therapy , Appendiceal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Adenocarcinoma, Mucinous/therapy
8.
Front Immunol ; 15: 1415457, 2024.
Article in English | MEDLINE | ID: mdl-39044825

ABSTRACT

Background: The occurrence of peritoneal metastasis (PM) in patients with colorectal cancer (CRC) has a dismal prognosis. There is often limited response to systemic- and immunotherapy, even in microsatellite unstable (MSI) CRC. To overcome therapy resistance, it is critical to understand local immune environment in the peritoneal cavity, and to develop models to study anti-tumor immune responses. Here, we defined the peritoneal immune system (PerIS) in PM-CRC patients and evaluate the pre-clinical potential of a humanized immune system (HIS) mouse model for PM-CRC. Methods: We studied the human PerIS in PM-CRC patients (n=20; MSS 19/20; 95%) and in healthy controls (n=3). HIS mice (NODscid gamma background; n=18) were generated, followed by intraperitoneal injection of either saline (HIS control; n=3) or human MSS/MSI CRC cell lines HUTU80, MDST8 and HCT116 (HIS-PM, n=15). Immune cells in peritoneal fluid and peritoneal tumors were analyzed using cytometry by time of flight (CyTOF). Results: The human and HIS mouse homeostatic PerIS was equally populated by NK cells and CD4+- and CD8+ T cells, however differences were observed in macrophage and B cell abundance. In HIS mice, successful peritoneal engraftment of both MSI and MSS tumors was observed (15/15; 100%). Both in human PM-CRC and in the HIS mouse PM-CRC model, we observed that MSS PM-CRC triggered a CD4+ Treg response in the PerIS, while MSI PM-CRC drives CD8+ TEMs responses. Conclusion: In conclusion, T cell responses in PM-CRC in HIS mice mirror those in human PM-CRC, making this model suitable to study antitumor T cell responses in PM-CRC.


Subject(s)
Colorectal Neoplasms , Disease Models, Animal , Peritoneal Neoplasms , Animals , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/immunology , Humans , Mice , Male , Female , Cell Line, Tumor , Mice, Inbred NOD , Mice, SCID , Middle Aged , Aged , Tumor Microenvironment/immunology , Killer Cells, Natural/immunology
9.
Technol Cancer Res Treat ; 23: 15330338241264169, 2024.
Article in English | MEDLINE | ID: mdl-39051686

ABSTRACT

Objective: The prognosis of malignant tumors with peritoneal metastases and cancerous ascites has generally been poor, with limited treatment options. The PRaG regimen, which comprised of hypofractionated radiotherapy, programmed cell death-1 (PD-1) inhibitor, and granulocyte-macrophage colony-stimulating factor (GM-CSF), showed a survival advantage in patients with advanced solid tumors who failed at least the first line of standard systemic treatment. Intraperitoneal infusion of PD-1 inhibitors may be a novel therapeutic strategy for managing malignant ascites. Integrating the PRaG regimen with intraperitoneal perfusion of a PD-1 inhibitor might control malignant ascites and provide further survival benefits in these patients. This proposed study aims to investigate the safety and efficacy of intraperitoneal infusion of serplulimab in combination with the PRaG regimen in patients with simultaneous advanced solid tumors and cancerous ascites who fail at least the first-line treatment. Methods: This proposed study is a prospective, single-arm, open-label, multicenter clinical trial. All eligible patients will receive 2 cycles of intensive treatment, a combination of PRaG regimen with an intraperitoneal infusion of PD-1 inhibitor. The patients who are beneficially treated with intensive treatment will receive consolidation treatment every 2 weeks until ascites disappear, disease progression occurs, intolerable toxicity occurs, or for up to 1 year. Phase I of this study will be conducted using a modified 3 + 3 design. The dose of intraperitoneal infusion of PD-1 inhibitor for phase II will be determined according to dose-limiting toxicity evaluation in the phase I study. Conclusion: This prospective, open-label, multicenter study will potentially lead to intraperitoneal perfusion of a PD-1 inhibitor being a new strategy for malignant ascites patients and provide a meaningful efficacy and safety of the combination of PRaG regimen with an intraperitoneal infusion of PD-1 inhibitor for these patients.


Subject(s)
Ascites , Immune Checkpoint Inhibitors , Infusions, Parenteral , Neoplasms , Humans , Ascites/etiology , Ascites/drug therapy , Ascites/pathology , Female , Male , Middle Aged , Neoplasms/drug therapy , Neoplasms/complications , Neoplasms/pathology , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Adult , Aged , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Treatment Outcome , Prospective Studies
10.
Curr Oncol ; 31(7): 3657-3668, 2024 Jun 25.
Article in English | MEDLINE | ID: mdl-39057141

ABSTRACT

BACKGROUND: Cytoreductive surgery (CRS) combined with hyperthermic intraperitoneal chemotherapy (HIPEC) is a major treatment of colorectal peritoneal carcinomatosis (CPC). The aim was to determine the disease-free survival (DFS) and overall survival (OS) of patients undergoing CRS-HIPEC for CPC and factors associated with long-term survival (LTS). METHODS: consecutive CPC patients who underwent CRS-HIPEC at a HIPEC center between 2007 and 2021 were included. Actual survival was calculated, and Cox proportional hazards models were used to identify factors associated with OS, DFS and LTS. RESULTS: there were 125 patients with CPC who underwent primary CRS-HIPEC, with mean age of 54.5 years. Median follow-up was 31 months. Average intraoperative PCI was 11, and complete cytoreduction (CC-0) was achieved in 96.8%. Median OS was 41.6 months (6-196). The 2-year and 5-year OS were 68% and 24.8%, respectively, and the 2-year DFS was 28.8%. Factors associated with worse OS included pre-HIPEC systemic therapy, synchronous extraperitoneal metastasis, and PCI ≥ 20 (p < 0.05). Progression prior to CRS-HIPEC was associated with worse DFS (p < 0.05). Lower PCI, fewer complications, lower recurrence and longer DFS were associated with LTS (p < 0.05). CONCLUSION: CRS and HIPEC improve OS in CPC patients but they have high disease recurrence. Outcomes depend on preoperative therapy response, extraperitoneal metastasis, and peritoneal disease burden.


Subject(s)
Colorectal Neoplasms , Cytoreduction Surgical Procedures , Hyperthermic Intraperitoneal Chemotherapy , Peritoneal Neoplasms , Humans , Cytoreduction Surgical Procedures/methods , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/secondary , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Female , Hyperthermic Intraperitoneal Chemotherapy/methods , Male , Middle Aged , Aged , Adult , Treatment Outcome , Combined Modality Therapy , Retrospective Studies
11.
Sci Rep ; 14(1): 17522, 2024 07 30.
Article in English | MEDLINE | ID: mdl-39080370

ABSTRACT

Peritoneal metastasis (PM), the regional progression of intra-abdominal malignancies, is a common sequelae of colorectal cancer (CRC). Immunotherapy is slated to be effective in generating long-lasting anti-tumour response as it utilizes the specificity and memory of the immune system. In the tumour microenvironment, tumour associated macrophages (TAMs) are posited to create an anti-inflammatory pro-tumorigenic environment. In this paper, we aimed to identify immunomodulatory factors associated with colorectal PM (CPM). A publicly available colorectal single cell database (GSE183916) was analysed to identify possible immunological markers that are associated with the activation of macrophages in cancers. Immunohistochemical analysis for V-set and immunoglobin containing domain 4 (VSIG4) expression was performed on tumour microarrays (TMAs) of tumours of colorectal origin (n = 211). Expression of VSIG4 in cell-free ascites obtained from CPM patients (n = 39) was determined using enzyme-linked immunosorbent assay (ELISA). CD163-positive TAMs cluster expression was extracted from a publicly available single cell database and evaluated for the top 100 genes. From these macrophage-expressed genes, VSIG4, a membrane protein produced by the M2 macrophages, mediates the up-regulation of anti-inflammatory and down-regulation of pro-inflammatory macrophages, contributing to an overall anti-inflammatory state. CRC TMA IHC staining showed that low expression of VSIG4 in stromal tissues of primary CRC are associated with poor prognosis (p = 0.0226). CPM ascites also contained varying concentrations of VSIG4, which points to a possible role of VSIG4 in the ascites. The contribution of VSIG4 to CPM development can be further evaluated for its potential as an immunotherapeutic agent.


Subject(s)
Colorectal Neoplasms , Peritoneal Neoplasms , Humans , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/immunology , Female , Male , Tumor Microenvironment/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Middle Aged , Paracrine Communication , Aged , Gene Expression Regulation, Neoplastic , Immunomodulation
12.
ACS Nano ; 18(29): 18963-18979, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39004822

ABSTRACT

Intraperitoneal co-delivery of chemotherapeutic drugs (CDs) and immune checkpoint inhibitors (ICIs) brings hope to improve treatment outcomes in patients with peritoneal metastasis from ovarian cancer (OC). However, current intraperitoneal drug delivery systems face issues such as rapid drug clearance from lymphatic drainage, heterogeneous drug distribution, and uncontrolled release of therapeutic agents into the peritoneal cavity. Herein, we developed an injectable nanohydrogel by combining carboxymethyl chitosan (CMCS) with bioadhesive nanoparticles (BNPs) based on polylactic acid-hyperbranched polyglycerol. This system enables the codelivery of CD and ICI into the intraperitoneal space to extend drug retention. The nanohydrogel is formed by cross-linking of aldehyde groups on BNPs with amine groups on CMCS via reversible Schiff base bonds, with CD and ICI loaded separately into BNPs and CMCS network. BNP/CMCS nanohydrogel maintained the activity of the biomolecules and released drugs in a sustained manner over a 7 day period. The adhesive property, through the formation of Schiff bases with peritoneal tissues, confers BNPs with an extended residence time in the peritoneal cavity after being released from the nanohydrogel. In a mouse model, BNP/CMCS nanohydrogel loaded with paclitaxel (PTX) and anti-PD-1 antibodies (αPD-1) significantly suppressed peritoneal metastasis of OC compared to all other tested groups. In addition, no systemic toxicity of nanohydrogel-loaded PTX and αPD-1 was observed during the treatment, which supports potential translational applications of this delivery system.


Subject(s)
Chitosan , Drug Delivery Systems , Hydrogels , Immune Checkpoint Inhibitors , Nanocomposites , Ovarian Neoplasms , Peritoneal Neoplasms , Animals , Hydrogels/chemistry , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/pathology , Mice , Chitosan/chemistry , Chitosan/analogs & derivatives , Female , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/chemistry , Immune Checkpoint Inhibitors/administration & dosage , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Nanocomposites/chemistry , Humans , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Mice, Inbred BALB C , Glycerol/chemistry , Glycerol/analogs & derivatives , Cell Line, Tumor , Polymers/chemistry , Polyesters
13.
World J Surg Oncol ; 22(1): 205, 2024 Jul 31.
Article in English | MEDLINE | ID: mdl-39085860

ABSTRACT

BACKGROUND: Cytoreductive surgery and chemotherapy reportedly improve the prognosis of patients with metachronous peritoneal metastases. However, the types of peritoneal metastases indicated for cytoreductive surgery remains unclear. Therefore, we aimed to clarify the category of cases for which cytoreductive surgery would be effective and report the prognosis associated with cytoreductive surgery for metachronous peritoneal metastases. METHODS: This study included 52 consecutive patients who underwent cytoreductive surgery for metachronous peritoneal metastases caused by colorectal cancer between January 2005 and December 2018 and fulfilled the selection criteria. The median follow-up period was 54.9 months. Relapse-free survival was calculated as the time from cytoreductive surgery of metachronous peritoneal metastases to recurrence. Overall survival was defined as the time from cytoreductive surgery of metachronous peritoneal metastases to death or the end of the follow-up period. RESULTS: The 5-year relapse-free survival rate was 30.0% and the 5-year overall survival rate was 72.3%. None of the patients underwent hyperthermic intraperitoneal chemotherapy. The analysis indicated no potential risk factors for 5-year relapse-free survival. However, for 5-year overall survival, the multivariate analysis revealed that time to diagnosis of metachronous peritoneal metastases of < 2 years after primary surgery (hazard ratio = 4.1, 95% confidence interval = 2.0-8.6, p = 0.0002) and number of metachronous peritoneal metastases ≥ 3 (hazard ratio = 9.8, 95% confidence interval = 2.3-42.3, p = 0.002) as independent factors associated with a poor prognosis. CONCLUSIONS: Long intervals of more than 2 years after primary surgery and 2 or less metachronous peritoneal metastases were good selection criteria for cytoreductive surgery for metachronous peritoneal metastases from colorectal cancer.


Subject(s)
Colorectal Neoplasms , Cytoreduction Surgical Procedures , Peritoneal Neoplasms , Humans , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/surgery , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/drug therapy , Cytoreduction Surgical Procedures/mortality , Cytoreduction Surgical Procedures/methods , Male , Female , Colorectal Neoplasms/pathology , Colorectal Neoplasms/surgery , Colorectal Neoplasms/mortality , Colorectal Neoplasms/drug therapy , Middle Aged , Aged , Survival Rate , Prognosis , Follow-Up Studies , Adult , Retrospective Studies , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/surgery , Neoplasms, Second Primary/surgery , Neoplasms, Second Primary/pathology , Neoplasms, Second Primary/mortality , Neoplasms, Second Primary/drug therapy , Hyperthermic Intraperitoneal Chemotherapy/methods , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
14.
J Transl Med ; 22(1): 646, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982444

ABSTRACT

BACKGROUND: Patients with peritoneal metastasis from colorectal cancer (PM-CRC) have inferior prognosis and respond particularly poorly to chemotherapy. This study aims to identify the molecular explanation for the observed clinical behavior and suggest novel treatment strategies in PM-CRC. METHODS: Tumor samples (230) from a Norwegian national cohort undergoing surgery and hyperthermic intraperitoneal chemotherapy (HIPEC) with mitomycin C (MMC) for PM-CRC were subjected to targeted DNA sequencing, and associations with clinical data were analyzed. mRNA sequencing was conducted on a subset of 30 samples to compare gene expression in tumors harboring BRAF or KRAS mutations and wild-type tumors. RESULTS: BRAF mutations were detected in 27% of the patients, and the BRAF-mutated subgroup had inferior overall survival compared to wild-type cases (median 16 vs 36 months, respectively, p < 0.001). BRAF mutations were associated with RNF43/RSPO aberrations and low expression of negative Wnt regulators (ligand-dependent Wnt activation). Furthermore, BRAF mutations were associated with gene expression changes in transport solute carrier proteins (specifically SLC7A6) and drug metabolism enzymes (CES1 and CYP3A4) that could influence the efficacy of MMC and irinotecan, respectively. BRAF-mutated tumors additionally exhibited increased expression of members of the novel butyrophilin subfamily of immune checkpoint molecules (BTN1A1 and BTNL9). CONCLUSIONS: BRAF mutations were frequently detected and were associated with particularly poor survival in this cohort, possibly related to ligand-dependent Wnt activation and altered drug transport and metabolism that could confer resistance to MMC and irinotecan. Drugs that target ligand-dependent Wnt activation or the BTN immune checkpoints could represent two novel therapy approaches.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm , Mutation , Peritoneal Neoplasms , Proto-Oncogene Proteins B-raf , Humans , Proto-Oncogene Proteins B-raf/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/drug therapy , Mutation/genetics , Female , Male , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Middle Aged , Aged , Gene Expression Regulation, Neoplastic , Molecular Targeted Therapy , Adult
15.
Exp Cell Res ; 441(1): 114155, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-39002689

ABSTRACT

At least one-third of patients with epithelial ovarian cancer (OC) present ascites at diagnosis and almost all have ascites at recurrence especially because of the propensity of the OC cells to spread in the abdominal cavity leading to peritoneal metastasis. The influence of ascites on the development of pre-metastatic niches, and on the biological mechanisms leading to cancer cell colonization of the mesothelium, remains poorly understood. Here, we show that ascites weakens the mesothelium by affecting the morphology of mesothelial cells and by destabilizing their distribution in the cell cycle. Ascites also causes destabilization of the integrity of mesothelium by modifying the organization of cell junctions, but it does not affect the synthesis of N-cadherin and ZO-1 by mesothelial cells. Moreover, ascites induces disorganization of focal contacts and causes actin cytoskeletal reorganization potentially dependent on the activity of Rac1. Ascites allows the densification and reorganization of ECM proteins of the mesothelium, especially fibrinogen/fibrin, and indicates that it is a source of the fibrinogen and fibrin surrounding OC spheroids. The fibrin in ascites leads to the adhesion of OC spheroids to the mesothelium, and ascites promotes their disaggregation followed by the clearance of mesothelial cells. Both αV and α5ß1 integrins are involved. In conclusion ascites and its fibrinogen/fibrin composition affects the integrity of the mesothelium and promotes the integrin-dependent implantation of OC spheroids in the mesothelium.


Subject(s)
Ascites , Fibrin , Fibrinogen , Integrin alpha5beta1 , Ovarian Neoplasms , Spheroids, Cellular , Tumor Microenvironment , Humans , Female , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ascites/pathology , Ascites/metabolism , Integrin alpha5beta1/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Fibrinogen/metabolism , Fibrin/metabolism , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/metabolism , Peritoneal Neoplasms/pathology , Carcinoma, Ovarian Epithelial/metabolism , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Receptors, Vitronectin/metabolism , rac1 GTP-Binding Protein/metabolism , Cell Adhesion , Peritoneum/pathology , Peritoneum/metabolism , Epithelium/metabolism , Epithelium/pathology , Cadherins/metabolism , Tumor Cells, Cultured
16.
J Gastric Cancer ; 24(3): 246-256, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38960884

ABSTRACT

PURPOSE: Peritoneal carcinomatosis (PC) presents a major challenge in the treatment of late-stage, solid tumors, with traditional therapies limited by poor drug penetration. We evaluated a novel hyperthermic pressurized intraperitoneal aerosol chemotherapy (HPIPAC) system using a human abdominal cavity model for its efficacy against AGS gastric cancer cells. MATERIALS AND METHODS: A model simulating the human abdominal cavity and AGS gastric cancer cell line cultured dishes were used to assess the efficacy of the HPIPAC system. Cell viability was measured to evaluate the impact of HPIPAC under 6 different conditions: heat alone, PIPAC with paclitaxel (PTX), PTX alone, normal saline (NS) alone, heat with NS, and HPIPAC with PTX. RESULTS: Results showed a significant reduction in cell viability with HPIPAC combined with PTX, indicating enhanced cytotoxic effects. Immediately after treatment, the average cell viability was 66.6%, which decreased to 49.2% after 48 hours and to a further 19.6% after 120 hours of incubation, demonstrating the sustained efficacy of the treatment. In contrast, control groups exhibited a recovery in cell viability; heat alone showed cell viability increasing from 90.8% to 94.4%, PIPAC with PTX from 82.7% to 89.7%, PTX only from 73.3% to 74.8%, NS only from 90.9% to 98.3%, and heat with NS from 74.4% to 84.7%. CONCLUSIONS: The HPIPAC system with PTX exhibits a promising approach in the treatment of PC in gastric cancer, significantly reducing cell viability. Despite certain limitations, this study highlights the system's potential to enhance treatment outcomes. Future efforts should focus on refining HPIPAC and validating its effectiveness in clinical settings.


Subject(s)
Aerosols , Cell Survival , Hyperthermic Intraperitoneal Chemotherapy , Paclitaxel , Peritoneal Neoplasms , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/pathology , Peritoneal Neoplasms/therapy , Paclitaxel/pharmacology , Paclitaxel/administration & dosage , Hyperthermic Intraperitoneal Chemotherapy/methods , Cell Survival/drug effects , Cell Line, Tumor , Hyperthermia, Induced/methods , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/pharmacology
17.
J Gastric Cancer ; 24(3): 291-299, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38960888

ABSTRACT

PURPOSE: Gastric cancer treated with curative resection exhibits several recurrence patterns. The peritoneum is the most common site of recurrence. Some reports have indicated different prognostic influences according to the recurrence sites in other cancers, such as esophageal and colorectal cancers. This study investigated whether the recurrence sites influenced the prognosis of patients with recurrent gastric cancer. MATERIALS AND METHODS: The data of 115 patients who experienced tumor recurrence after curative gastrectomy were retrospectively reviewed. The sites of recurrence were divided into 4 groups: lymph node (LN), peritoneum, other single organs, and multiple lesions. Clinicopathological features were compared between the sites of recurrence. Prognosis after resection and recurrence were also compared. RESULTS: The peritoneum was the primary site of recurrence in 38 patients (33%). The tumor differentiation and pathological stages were significantly different. Survival after surgery did not show a statistically significant difference (hazard ratio [HR] of LN: 1, peritoneum: 1.083, other single organs: 1.025, and multiple lesions: 1.058; P=1.00). Survival after recurrence was significantly different (HR of LN, 1; peritoneum, 2.164; other single organs, 1.092; multiple lesions, 1.554; P=0.01), and patients with peritoneal and multiple lesion recurrences had worse prognosis. Furthermore, peritoneal recurrence seemed to occur later than that at other sites; the median times to recurrence in LN, peritoneal, other single-organ, and multiple lesions were 265, 722, 372, and 325 days, respectively. CONCLUSIONS: The sites of gastric cancer recurrence may have different prognostic effects. Peritoneal recurrence may be less sensitive to chemotherapy and occur during the late phase of recurrence.


Subject(s)
Gastrectomy , Neoplasm Recurrence, Local , Stomach Neoplasms , Humans , Stomach Neoplasms/surgery , Stomach Neoplasms/pathology , Stomach Neoplasms/mortality , Male , Female , Retrospective Studies , Neoplasm Recurrence, Local/pathology , Middle Aged , Prognosis , Aged , Adult , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/surgery , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/pathology , Aged, 80 and over , Lymphatic Metastasis/pathology
18.
Zhonghua Wei Chang Wai Ke Za Zhi ; 27(7): 740-748, 2024 Jul 25.
Article in Chinese | MEDLINE | ID: mdl-39004991

ABSTRACT

Peritoneal metastasis in gastric cancer is associated with rapid disease progression. Hyperthermic intraoperative peritoneal chemotherapy (HIPEC) done immediately after cytoreductive surgery (CRS) has become an important treatment for peritoneal metastasis in gastric cancer patients. However, different treatment options for HIPEC exist with potential influence on survival rates and prognosis in patients, exist. These treatment options include open or closed abdomen technique, perfusion solution, number of catheters, temperature, duration, and drug regimens. This paper aims to provide more evidence on standardization of HIPEC treatment options and technologies by systematically reviewing different drug regimens and technical approaches. The study included 2 randomized controlled trials, 3 phase I/II clinical trials, 2 prospective cohort studies, and 34 retrospective cohort studies, involving 1511 patients. The most common HIPEC option is to dissolve 50-75 mg/m2 of Cisplatin and 30-40 mg/m2 of Mitomycin C in 3-4 L saline solution at 42-43℃. After gastrointestinal anastomosis, 2-3 catheters are used in the HIPEC system with a perfusion flow rate of 500 ml/min. The duration is 60-90 minutes. Anastomotic leakage was low in studies where HIPEC was performed after gastrointestinal anastomosis. The utilization of open HIPEC and a two-drug regimen resulted in improved overall survival rates. The future development of HIPEC aims to enhance tumor-specific therapy by optimizing various aspects, such as identifying the safest and most effective chemotherapy regimens, refining patient selection criteria, and improving perioperative care.


Subject(s)
Cytoreduction Surgical Procedures , Hyperthermic Intraperitoneal Chemotherapy , Peritoneal Neoplasms , Stomach Neoplasms , Humans , Cisplatin/administration & dosage , Cisplatin/therapeutic use , Combined Modality Therapy , Cytoreduction Surgical Procedures/methods , Hyperthermia, Induced/methods , Hyperthermic Intraperitoneal Chemotherapy/methods , Mitomycin/administration & dosage , Mitomycin/therapeutic use , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Peritoneal Neoplasms/drug therapy , Stomach Neoplasms/therapy
19.
J Immunother Cancer ; 12(7)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38955417

ABSTRACT

BACKGROUND: Tertiary lymphoid structures (TLSs) are thought to stimulate antitumor immunity and positively impact prognosis and response to immune checkpoint blockade. In gastric cancers (GCs), however, TLSs are predominantly found in GC with poor prognosis and limited treatment response. We, therefore, hypothesize that immune cell composition and function of TLS depends on tumor location and the tumor immune environment. METHODS: Spatial transcriptomics and immunohistochemistry were used to characterize the phenotype of CD45+ immune cells inside and outside of TLS using archival resection specimens from GC primary tumors and peritoneal metastases. RESULTS: We identified significant intrapatient and interpatient diversity of the cellular composition and maturation status of TLS in GC. Tumor location (primary vs metastatic site) accounted for the majority of differences in TLS maturity, as TLS in peritoneal metastases were predominantly immature. This was associated with higher levels of tumor-infiltrating macrophages and Tregs and less plasma cells compared with tumors with mature TLS. Furthermore, mature TLSs were characterized by overexpression of antitumor immune pathways such as B cell-related pathways, MHC class II antigen presentation while immature TLS were associated with protumor pathways, including T cell exhaustion and enhancement of DNA repair pathways in the corresponding cancer. CONCLUSION: The observation that GC-derived peritoneal metastases often contain immature TLS which are associated with immune suppressive regulatory tumor-infiltrating leucocytes, is in keeping with the lack of response to immune checkpoint blockade and the poor prognostic features of peritoneal metastatic GC, which needs to be taken into account when optimizing immunomodulatory strategies for metastatic GC.


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , Tertiary Lymphoid Structures , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/immunology , Tertiary Lymphoid Structures/immunology , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/immunology , Male , Female , Tumor Microenvironment
20.
Sci Rep ; 14(1): 17648, 2024 07 31.
Article in English | MEDLINE | ID: mdl-39085339

ABSTRACT

Claudin 18.2 has emerged as a viable therapeutic target in gastric cancer (GC), but little is known about the heterogeneity of its expression in GC. This study investigated the heterogeneity of claudin 18.2 expression in 166 patients with metastatic GC whose surgical or paired primary-metastatic specimens were available. The prevalence of claudin 18.2 positivity (moderate-to-strong expression in ≥ 75% by the 43-14A clone) was 47.0%. Claudin 18.2-positive tumors exhibited more frequent peritoneal metastasis and a lower incidence of hepatic and distant lymph node involvement. Survival outcomes were comparable between patients with claudin 18.2-positive and -negative tumors. Intratumoral heterogeneity was noted in 38.5% of surgical specimens. Paired primary-metastatic site analysis revealed that 25.2% of patients had discordant results for claudin 18.2 positivity. Across different metastatic organ categories, peritoneal lesions showed the highest positivity rate (44.3%) and positive concordance rate (31.4%), whereas liver lesions had the lowest positivity rate (17.9%) and concordance rate (12.8%). In conclusion, claudin 18.2 expression exhibits intratumoral and intrapatient spatial heterogeneity in metastatic GC. Claudin 18.2 positivity is associated with more frequent peritoneal metastasis, and peritoneal lesions are more likely to have positively concordant claudin 18.2 results with the primary site than other metastatic sites.


Subject(s)
Claudins , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Male , Female , Middle Aged , Aged , Claudins/metabolism , Claudins/genetics , Biomarkers, Tumor/metabolism , Neoplasm Metastasis , Adult , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/metabolism , Aged, 80 and over , Lymphatic Metastasis , Prognosis , Liver Neoplasms/secondary , Liver Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL