Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.412
Filtrer
1.
BMC Surg ; 24(1): 285, 2024 Oct 04.
Article de Anglais | MEDLINE | ID: mdl-39367354

RÉSUMÉ

Pancreatic cancer is a highly aggressive cancer with unfavorable prognosis despite the therapeutic interventions. Intraperitoneal chemotherapy has recently shown potential outcomes in the presence of peritoneal metastases. However, a consensus is still lacking on different methods for intraperitoneal chemotherapy in pancreatic cancer. A variety of drugs and doses via three types of intraperitoneal chemotherapy have been studied. The prognosis and treatment strategies for pancreatic ductal adenocarcinoma (PDAC) will be significantly influenced by peritoneal dissemination and resectability of the macroscopic disease. Normothermic intraperitoneal chemotherapy (NIPEC) has been used for the treatment of peritoneal metastases of pancreatic carcinomas. Intraperitoneal chemotherapy is often combined with systemic therapies or surgical procedures which may lead to favorable combination therapies such as cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS/HIPEC). Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a relatively new approach that provides a homogenous and deep penetration of the chemotherapy into the peritoneum by producing aerosols. The present study aims to review the literature for recent evidence on intraperitoneal chemotherapy in pancreatic cancer.


Sujet(s)
Tumeurs du pancréas , Tumeurs du péritoine , Humains , Tumeurs du pancréas/traitement médicamenteux , Tumeurs du pancréas/thérapie , Tumeurs du pancréas/anatomopathologie , Tumeurs du péritoine/traitement médicamenteux , Tumeurs du péritoine/thérapie , Tumeurs du péritoine/secondaire , Antinéoplasiques/administration et posologie , Chimiothérapie hyperthermique intrapéritonéale , Carcinome du canal pancréatique/traitement médicamenteux , Carcinome du canal pancréatique/thérapie , Résultat thérapeutique , Perfusions parentérales
2.
BMJ Open ; 14(10): e083659, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39353699

RÉSUMÉ

BACKGROUND: Gastric cancer (GC) is the fourth leading cause of cancer mortality worldwide. Peritoneal metastasis (PM) is a significant cause of death in patients with GC, and presents a major challenge in clinical diagnosis and treatment. Predicting the occurrence of PM in high-risk patients, and diagnosing and treating PM in advance to improve patient survival, remains an unsolved problem in clinical practice. Given the low positive rate of cytology and difficulty in diagnosing occult PM, new molecular markers and detection technologies for early diagnosis require urgent validation. The primary objective of this study is to observe and evaluate the predictive effect of intraoperative peritoneal lavage fluid (PLF) circulating tumour cells (CTC) and circulating tumour DNA (ctDNA) levels in patients with pT4NxM0/pT1-3N+M0 GC on metachronous PM after R0 resection. METHODS AND ANALYSIS: This prospective single-centre clinical study is conducted at Renji Hospital, Shanghai Jiao Tong University School of Medicine. In this study, 200 cases of patients with pT4NxM0/pT1-3N+M0 gastric adenocarcinoma older than 18 years will be screened. Participants will undergo intraoperative PLF CTC and ctDNA testing and will be followed up for 2 years, with imaging assessments performed every 3-6 months until PM occurrs. The primary outcome is the incidence of PM 1 year after surgery, which will be estimated using Clopper-Pearson method, with 95% CIs calculated and compared between groups. Secondary outcome include the incidence of PM 2 years after surgery, overall survival and disease progression. Data will be analysed using the Kaplan-Meier method and the log-rank test. ETHICS AND COMMUNICATION: Informed consent has been obtained from all subjects. This protocol has been approved by the Ethics Committee of Renji Hospital, Shanghai Jiao Tong University School of Medicine (LY2023-142-B). The findings will be disseminated through peer-reviewed manuscripts, reports and presentations. TRIAL REGISTRATION NUMBER: ChiCTR2300074910.


Sujet(s)
ADN tumoral circulant , Cellules tumorales circulantes , Lavage péritonéal , Tumeurs du péritoine , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/génétique , Tumeurs de l'estomac/chirurgie , Études prospectives , ADN tumoral circulant/sang , ADN tumoral circulant/génétique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/génétique , Cellules tumorales circulantes/métabolisme , Cellules tumorales circulantes/anatomopathologie , Mâle , Femelle , Liquide d'ascite/métabolisme , Chine , Adulte d'âge moyen , Marqueurs biologiques tumoraux/sang , Adénocarcinome/chirurgie , Adénocarcinome/génétique , Adénocarcinome/anatomopathologie , Adénocarcinome/secondaire , Valeur prédictive des tests , Gastrectomie/méthodes , Stadification tumorale
4.
Biomed Pharmacother ; 179: 117294, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39226726

RÉSUMÉ

Peritoneal metastasis (PM) pathophysiology is complex and not fully understood. PM, originating from gastrointestinal (GI) cancer, is a condition that significantly worsens patient prognosis due to its complex nature and limited treatment options. The non-coding RNAs (ncRNAs) have been shown to play pivotal roles in cancer biology, influencing tumorigenesis, progression, metastasis, and therapeutic resistance. Increasing evidence has demonstrated the regulatory functions of different classes of ncRNAs, including microRNAs (miRNAs) and long non-coding RNAs (lncRNAs) in PM. Identifying biomarkers for early detection of PM is a crucial step towards improving patient outcomes, and how ncRNA profiles correlate with survival rates, response to therapy, and recurrence risks have raised much attention in recent years. Additionally, exploring innovative therapeutic approaches utilizing ncRNAs, such as targeted therapy and gene silencing, may offer new horizons in treating this dire condition. Recent advances in systemic treatments and the development of novel loco-regional therapies have opened doors to multimodal treatment approaches. Radical surgeries combined with hyperthermic intraperitoneal chemotherapy (HIPEC) have shown promising results, leading to extended patient survival. Current research is focused on the molecular characterization of PM, which is crucial for early detection and developing future therapeutic strategies. By summarizing the latest findings, this study underscores the transformative potential of ncRNAs in enhancing the diagnosis, prognosis, and treatment of PM in GI cancer, paving the way for more personalized and effective clinical strategies.


Sujet(s)
Marqueurs biologiques tumoraux , Tumeurs du péritoine , Humains , Marqueurs biologiques tumoraux/génétique , Tumeurs du péritoine/génétique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/thérapie , Animaux , ARN non traduit/génétique , ARN long non codant/génétique , Régulation de l'expression des gènes tumoraux , Tumeurs gastro-intestinales/génétique , Tumeurs gastro-intestinales/anatomopathologie , Tumeurs gastro-intestinales/thérapie , microARN/génétique
5.
Health Technol Assess ; 28(51): 1-139, 2024 09.
Article de Anglais | MEDLINE | ID: mdl-39254852

RÉSUMÉ

Background: We compared the relative benefits, harms and cost-effectiveness of hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery ± systemic chemotherapy versus cytoreductive surgery ± systemic chemotherapy or systemic chemotherapy alone in people with peritoneal metastases from colorectal, gastric or ovarian cancers by a systematic review, meta-analysis and model-based cost-utility analysis. Methods: We searched MEDLINE, EMBASE, Cochrane Library and the Science Citation Index, ClinicalTrials.gov and WHO ICTRP trial registers until 14 April 2022. We included only randomised controlled trials addressing the research objectives. We used the Cochrane risk of bias tool version 2 to assess the risk of bias in randomised controlled trials. We used the random-effects model for data synthesis when applicable. For the cost-effectiveness analysis, we performed a model-based cost-utility analysis using methods recommended by The National Institute for Health and Care Excellence. Results: The systematic review included a total of eight randomised controlled trials (seven randomised controlled trials, 955 participants included in the quantitative analysis). All comparisons other than those for stage III or greater epithelial ovarian cancer contained only one trial, indicating the paucity of randomised controlled trials that provided data. For colorectal cancer, hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy probably results in little to no difference in all-cause mortality (60.6% vs. 60.6%; hazard ratio 1.00, 95% confidence interval 0.63 to 1.58) and may increase the serious adverse event proportions compared to cytoreductive surgery ± systemic chemotherapy (25.6% vs. 15.2%; risk ratio 1.69, 95% confidence interval 1.03 to 2.77). Hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy probably decreases all-cause mortality compared to fluorouracil-based systemic chemotherapy alone (40.8% vs. 60.8%; hazard ratio 0.55, 95% confidence interval 0.32 to 0.95). For gastric cancer, there is high uncertainty about the effects of hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy versus cytoreductive surgery + systemic chemotherapy or systemic chemotherapy alone on all-cause mortality. For stage III or greater epithelial ovarian cancer undergoing interval cytoreductive surgery, hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy probably decreases all-cause mortality compared to cytoreductive surgery + systemic chemotherapy (46.3% vs. 57.4%; hazard ratio 0.73, 95% confidence interval 0.57 to 0.93). Hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy may not be cost-effective versus cytoreductive surgery + systemic chemotherapy for colorectal cancer but may be cost-effective for the remaining comparisons. Limitations: We were unable to obtain individual participant data as planned. The limited number of randomised controlled trials for each comparison and the paucity of data on health-related quality of life mean that the recommendations may change as new evidence (from trials with a low risk of bias) emerges. Conclusions: In people with peritoneal metastases from colorectal cancer with limited peritoneal metastases and who are likely to withstand major surgery, hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy should not be used in routine clinical practice (strong recommendation). There is considerable uncertainty as to whether hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy or cytoreductive surgery + systemic chemotherapy should be offered to patients with gastric cancer and peritoneal metastases (no recommendation). Hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy should be offered routinely to women with stage III or greater epithelial ovarian cancer and metastases confined to the abdomen requiring and likely to withstand interval cytoreductive surgery after chemotherapy (strong recommendation). Future work: More randomised controlled trials are necessary. Study registration: This study is registered as PROSPERO CRD42019130504. Funding: This award was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme (NIHR award ref: 17/135/02) and is published in full in Health Technology Assessment; Vol. 28, No. 51. See the NIHR Funding and Awards website for further award information.


Cancers of the bowel, ovary or stomach can spread to the lining of the abdomen ('peritoneal metastases'). Chemotherapy (the use of drugs that aim to kill cancer cells) given by injection or tablets ('systemic chemotherapy') is one of the main treatment options. There is uncertainty about whether adding cytoreductive surgery (cytoreductive surgery; an operation to remove the cancer) and 'hyperthermic intraoperative peritoneal chemotherapy' (warm chemotherapy delivered into the lining of the abdomen during cytoreductive surgery) are beneficial. We reviewed all the information from medical literature published until 14 April 2022, to answer the above uncertainty. We found the following from eight trials, including about 1000 participants. In people with peritoneal metastases from bowel cancer, hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy probably does not provide any benefits and increases harm compared to cytoreductive surgery + systemic chemotherapy, while cytoreductive surgery + systemic chemotherapy appears to increase survival compared to systemic chemotherapy alone. There is uncertainty about the best treatment for people with peritoneal metastases from stomach cancer. In women with peritoneal metastases from ovarian cancer who require systemic chemotherapy before cytoreductive surgery to shrink the cancer to allow surgery ('advanced ovarian cancer'), hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy probably increases survival compared to cytoreductive surgery + systemic chemotherapy. In people who can withstand a major operation and in whom cancer can be removed, cytoreductive surgery + systemic chemotherapy should be offered to people with peritoneal metastases from bowel cancer, while hyperthermic intraoperative peritoneal chemotherapy + cytoreductive surgery + systemic chemotherapy should be offered to women with peritoneal metastases from 'advanced ovarian cancer'. Uncertainty in treatment continues for gastric cancer. This award was funded by the National Institute for Health and Care Research (NIHR) Health Technology Assessment programme (NIHR award ref: 17/135/02) and is published in full in Health Technology Assessment; Vol. 28, No. 51. See the NIHR Funding and Awards website for further award information.


Sujet(s)
Analyse coût-bénéfice , Interventions chirurgicales de cytoréduction , Chimiothérapie hyperthermique intrapéritonéale , Tumeurs du péritoine , Humains , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/thérapie , Tumeurs du péritoine/traitement médicamenteux , Interventions chirurgicales de cytoréduction/économie , Évaluation de la technologie biomédicale , Essais contrôlés randomisés comme sujet , Femelle , Années de vie ajustées sur la qualité , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/chirurgie , Tumeurs de l'ovaire/thérapie , Hyperthermie provoquée/économie , Évaluation du Coût-Efficacité
6.
Biomolecules ; 14(9)2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39334866

RÉSUMÉ

Ovarian cancer remains a leading cause of death among gynecological cancers, largely due to its propensity for peritoneal metastasis and the development of drug resistance. This review concentrates on the molecular underpinnings of these two critical challenges. We delve into the role of exosomes, the nano-sized vesicles integral to cellular communication, in orchestrating the complex interactions within the tumor microenvironment that facilitate metastatic spread and thwart therapeutic efforts. Specifically, we explore how exosomes drive peritoneal metastasis by promoting epithelial-mesenchymal transition in peritoneal mesothelial cells, altering the extracellular matrix, and supporting angiogenesis, which collectively enable the dissemination of cancer cells across the peritoneal cavity. Furthermore, we dissect the mechanisms by which exosomes contribute to the emergence of drug resistance, including the sequestration and expulsion of chemotherapeutic agents, the horizontal transfer of drug resistance genes, and the modulation of critical DNA repair and apoptotic pathways. By shedding light on these exosome-mediated processes, we underscore the potential of exosomal pathways as novel therapeutic targets, offering hope for more effective interventions against ovarian cancer's relentless progression.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Exosomes , Tumeurs de l'ovaire , Tumeurs du péritoine , Microenvironnement tumoral , Exosomes/métabolisme , Humains , Femelle , Tumeurs de l'ovaire/anatomopathologie , Tumeurs de l'ovaire/métabolisme , Tumeurs de l'ovaire/traitement médicamenteux , Tumeurs de l'ovaire/génétique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/traitement médicamenteux , Tumeurs du péritoine/métabolisme , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Animaux
7.
ESMO Open ; 9(9): 103681, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39288528

RÉSUMÉ

INTRODUCTION: Pressurized intraperitoneal aerosol chemotherapy-oxaliplatin (PIPAC-OX) induces direct DNA damage and immunogenic cell death in patients with gastric cancer peritoneal metastases (GCPM). Combining PIPAC-OX with immune checkpoint inhibition remains untested. We conducted a phase I first-in-human trial evaluating the safety and efficacy of PIPAC-OX combined with systemic nivolumab (NCT03172416). METHODS: Patients with GCPM who experienced disease progression on at least first-line systemic therapy were recruited across three centers in Singapore and Belgium. Patients received PIPAC-OX at 90 mg/m2 every 6 weeks and i.v. nivolumab 240 mg every 2 weeks. Translational studies were carried out on GCPM samples acquired during PIPAC-OX procedures. RESULTS: In total, 18 patients with GCPM were prospectively recruited. The PIPAC-OX and nivolumab combination was well tolerated with manageable treatment-related adverse events, although one patient suffered from grade 4 vomiting. At second and third PIPAC-OX, respectively, the median decrease in peritoneal cancer index (PCI) was -5 (interquartile range: -12 to +1) and -7 (interquartile range: -6 to -20) and peritoneal regression grade 1 or 2 was observed in 66.7% (6/9) and 100% (3/3). Translational analyses of 43 GCPM samples revealed enrichment of immune/stromal infiltration and inflammatory signatures in peritoneal tumors after PIPAC-OX and nivolumab. M2 macrophages were reduced in treated peritoneal tumor samples while memory CD4+, CD8+ central memory and naive CD8+ T-cells were increased. CONCLUSIONS: The first-in-human trial combining PIPAC-OX and nivolumab demonstrated safety and tolerability, coupled with enhanced T-cell infiltration within peritoneal tumors. This trial sets the stage for future combinations of systemic immunotherapy with locoregional intraperitoneal treatments.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Nivolumab , Oxaliplatine , Tumeurs du péritoine , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/traitement médicamenteux , Tumeurs de l'estomac/anatomopathologie , Nivolumab/pharmacologie , Nivolumab/administration et posologie , Nivolumab/usage thérapeutique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/traitement médicamenteux , Femelle , Mâle , Oxaliplatine/administration et posologie , Oxaliplatine/usage thérapeutique , Oxaliplatine/pharmacologie , Adulte d'âge moyen , Sujet âgé , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte , Résultat thérapeutique
8.
World J Surg Oncol ; 22(1): 244, 2024 Sep 11.
Article de Anglais | MEDLINE | ID: mdl-39256787

RÉSUMÉ

BACKGROUND: Clinical T4 (cT4) stage gastric cancer presents with frequent postoperative recurrence and poor prognosis. This study is to evaluate the oncological efficacy of laparoscopic radical total gastrectomy combined with postoperative prophylactic hyperthermic intraperitoneal chemotherapy (HIPEC) in patients with cT4N + M0 gastric cancer who received neoadjuvant chemotherapy. METHODS: We reviewed the clinicopathological data of 174 patients with clinical T4 gastric cancer who underwent neoadjuvant chemotherapy followed by laparoscopic radical total gastrectomy between June 2017 and December 2021. Among them, 142 were included in the non-HIPEC group, and 32 in the HIPEC group. Patients in both groups were paired based on propensity score in a 2:1 ratio to assess disparities in tumor recurrence and long-term survival. RESULTS: After matching, there were no significant differences in the clinicopathological data between the two groups. The peritoneum (16.1%) and distant organs (10.9%) were the most frequent locations for recurrence. Prior to matching, the recurrence rates were similar at all sites for both groups. Compared with those in the non-HIPEC cohort, the recurrence rates at all sites, the lung, and the peritoneum were notably lower in the HIPEC cohort. Prior to matching, the 3-year overall survival and disease-free survival rates were similar between the two groups; following matching, the HIPEC group exhibited notably greater survival rates than did the non-HIPEC group. The disparities in survival rates between the groups became even more pronounced after conducting a stratified analysis among patients with stage III disease. CONCLUSIONS: Neoadjuvant chemotherapy combined with prophylactic HIPEC after laparoscopic radical gastrectomy can effectively reduce the rate of peritoneal metastasis in patients with cT4N + M0 advanced gastric cancer and significantly improve the prognosis of such patients, which is of great clinical value.


Sujet(s)
Gastrectomie , Chimiothérapie hyperthermique intrapéritonéale , Laparoscopie , Traitement néoadjuvant , Récidive tumorale locale , Score de propension , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/thérapie , Tumeurs de l'estomac/chirurgie , Tumeurs de l'estomac/mortalité , Gastrectomie/méthodes , Mâle , Femelle , Études rétrospectives , Adulte d'âge moyen , Laparoscopie/méthodes , Traitement néoadjuvant/méthodes , Taux de survie , Pronostic , Chimiothérapie hyperthermique intrapéritonéale/méthodes , Études de suivi , Récidive tumorale locale/prévention et contrôle , Récidive tumorale locale/anatomopathologie , Récidive tumorale locale/thérapie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Stadification tumorale , Association thérapeutique , Sujet âgé , Traitement médicamenteux adjuvant/méthodes , Tumeurs du péritoine/thérapie , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/mortalité , Adulte
9.
PLoS One ; 19(9): e0307965, 2024.
Article de Anglais | MEDLINE | ID: mdl-39255313

RÉSUMÉ

Pressurized intraperitoneal aerosol chemotherapy (PIPAC) is a valuable therapeutic alternative for patients with peritoneal metastases. PIPAC uses a hyaluronic acid-based gel to reduce surgically induced adhesions. The aim of this study was to evaluate the effects of the hyaluronic acid-based gel on tumor dissemination. First, we explored whether the survival of CT26 luciferase-expressing murine colonic tumor cells was correlated with the dose of HyaRegen® Gel, and we determined the half-maximal inhibitory concentration (the IC50) of the gel. Next, we performed an in vitro study of cell survival rates after gel application on day 0 (D0) and day 1 (D1). Finally, we intraperitoneally administered the gel to mice with immunocompetent BALB/c colonic peritoneal metastases (on D0, D5, D10, D14, and D18). Tumor growth was regularly monitored using a bioluminescence assay (on D11, D17, and D21). After all mice had been sacrificed on D21, the body weights and the volumes of intraperitoneal ascites were measured; the Peritoneal Carcinosis Index (PCI) and Ki-antigen 67 scores were calculated. The IC50 value was 70 µL of gel in a total volume of 100 µL. The cell survival rates on D4 were identical in the control group and the two groups that had been treated with gel on D0 and D1. The bioluminescence levels over time were similar in the gel and control groups. The PCI scores were 35.5 ± 2.89 for the control group and 36 ± 2.45 for the gel group (p = 0.8005). The mean Ki-67 index percentages were 37.28 ±1 1.75 for the control group and 34.03 ± 8.62 for the gel group (p = 0.1971). This in vitro and in vivo study using a mouse model of immunocompetent metastatic peritoneal cancer did not reveal any pro- or anti-tumoral effect of HyaRegen® Gel. These findings indicate that the gel can be used to treat PIPACs with minimal apprehension.


Sujet(s)
Prolifération cellulaire , Tumeurs colorectales , Gels , Acide hyaluronique , Souris de lignée BALB C , Tumeurs du péritoine , Animaux , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/traitement médicamenteux , Souris , Prolifération cellulaire/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Acide hyaluronique/pharmacologie , Femelle , Survie cellulaire/effets des médicaments et des substances chimiques , Humains
10.
World J Surg Oncol ; 22(1): 212, 2024 Sep 02.
Article de Anglais | MEDLINE | ID: mdl-39218891

RÉSUMÉ

OBJECTIVES: Pancreatic cancer with peritoneal metastasis presents a challenging prognosis, with limited effective treatment options available. This study aims to evaluate the efficacy and safety of combining cytoreductive surgery (CRS) with hyperthermic intraperitoneal chemotherapy (HIPEC) as a treatment strategy for this patient group. METHODS: A retrospective analysis was conducted on patients with peritoneal metastasis of pancreatic cancer who underwent CRS + HIPEC treatment at Beijing Shijitan Hospital from March 2017 to December 2023. The study focused on assessing clinical features, the incidence of sever adverse events (SAEs), and overall survival (OS). RESULTS: A total of 10 patients were enrolled in this study. The median OS was 24.2 months, suggesting an improvement over traditional therapies. While SAEs were noted, including two cases of severe complications necessitating additional surgical interventions, no perioperative fatalities were recorded. The overall survival time for patients with CC0/1 was not significantly different from that of patients with CC2/3, and no prognostic predictors were identified. CONCLUSIONS: The combination of CRS and HIPEC appears to be a viable and promising treatment modality for patients with peritoneal metastasis of pancreatic cancer, offering an improved survival rate with manageable safety concerns. Further research is needed to refine patient selection criteria and to explore the long-term benefits of this approach.


Sujet(s)
Interventions chirurgicales de cytoréduction , Chimiothérapie hyperthermique intrapéritonéale , Tumeurs du pancréas , Tumeurs du péritoine , Humains , Tumeurs du péritoine/thérapie , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/mortalité , Interventions chirurgicales de cytoréduction/méthodes , Interventions chirurgicales de cytoréduction/mortalité , Mâle , Femelle , Tumeurs du pancréas/anatomopathologie , Tumeurs du pancréas/thérapie , Chimiothérapie hyperthermique intrapéritonéale/méthodes , Études rétrospectives , Adulte d'âge moyen , Taux de survie , Association thérapeutique , Pronostic , Sujet âgé , Études de suivi , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adulte
11.
Sci Rep ; 14(1): 22324, 2024 09 27.
Article de Anglais | MEDLINE | ID: mdl-39333597

RÉSUMÉ

Multimodal therapy for peritoneal metastasis (PM) including cytoreductive surgery (CRS) and hyperthermic intraperitoneal chemotherapy (HIPEC) provides long-term survival in highly selected colorectal cancer patients. Mechanisms behind HIPEC are unknown and may include induction of adaptive immunity. We therefore analyzed human PM samples and explored the impact of HIPEC in experimental models. Human samples from colorectal primary tumors (n = 19) and PM lesions (n = 37) were examined for the presence of CD8 + T-cells and their association with disease free (DFS) and overall survival (OS). CD8 + T cell response after HIPEC was assessed using an in-vivo PM mouse model, tumor cell lines and patient-derived tumor organoids. Patients with high intraepithelial CD8 + T cell counts showed longer DFS and OS. In the mouse model, HIPEC controlled growth of PM and increased numbers of functional granzyme positive CD8 + T cells within tumors. Cell lines and human organoids that were treated with heated chemotherapies showed immunogenic changes, reflected by significantly higher levels of MHC-class I molecules and expression of Cancer Testis Antigens Cyclin A1 and SSX-4. Using in-vitro co-culture assays, we noticed that cancer cells treated with heated chemotherapy primed dendritic cells, which subsequently enhanced effector functions of CD8 + T cells. The presence of CD8 + T-cells within PM lesions is associated with prolonged survival of patients with PM. Data from PM mouse model and in-vitro assay show that heated chemotherapies induce immunogenic changes on cancer cells leading to induction of CD8 + T-cells mediated immunity, which seems to control growth of PM lesions in mice after HIPEC.


Sujet(s)
Lymphocytes T CD8+ , Tumeurs colorectales , Chimiothérapie hyperthermique intrapéritonéale , Tumeurs du péritoine , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/thérapie , Tumeurs du péritoine/immunologie , Humains , Animaux , Chimiothérapie hyperthermique intrapéritonéale/méthodes , Souris , Lymphocytes T CD8+/immunologie , Tumeurs colorectales/anatomopathologie , Tumeurs colorectales/thérapie , Tumeurs colorectales/immunologie , Lignée cellulaire tumorale , Femelle , Mâle
12.
JCO Precis Oncol ; 8: e2400149, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39259912

RÉSUMÉ

PURPOSE: High-grade appendiceal adenocarcinomas (HGAA) with peritoneal metastases (PMs) are associated with poor survival. Hyperthermic intraperitoneal chemotherapy (HIPEC) is a novel treatment approach for unresectable HGAA-PM. However, its influence on immunogenomic profiles has not yet been fully explored. MATERIALS AND METHODS: We obtained 79 samples of metastatic peritoneal tumor deposits from patients diagnosed with HGAA and performed whole-exome sequencing, RNA sequencing, and immunoprofiling before and after HIPEC. Tumor biopsies were subjected to immunogenomic profiling to detect mutational signatures and immune populations associated with oncologic outcomes. RESULTS: Fifteen patients with HGAA-PMs were included in the study. The median progression-free survival (PFS) was 6.7 months (2.7-25.3) and the median overall survival was 11.4 months (4.7-42). Mucin-associated genes (MUC16, MUC3A, and MUC5AC) and titin (TTN) had the highest mutation frequencies. Mutational signatures such as single-base substitution 29 and doublet-base substitution 11 were present in >50% of single-base and double-base mutations. Higher PD-L1 coexpression on CD8+ T cells demonstrated a higher PFS both intratumorally (P = .019) and at the margin (P = .025). CONCLUSION: HIPEC-associated mutational signatures were identified in HGAA-PMs. Elevated PD-L1+ cytotoxic T-cell populations after HIPEC had better PFS, offering valuable insights for prognostication in the context of HIPEC treatment.


Sujet(s)
Tumeurs de l'appendice , Chimiothérapie hyperthermique intrapéritonéale , Mutation , Tumeurs du péritoine , Microenvironnement tumoral , Humains , Mâle , Femelle , Tumeurs de l'appendice/génétique , Tumeurs de l'appendice/anatomopathologie , Tumeurs de l'appendice/thérapie , Adulte d'âge moyen , Sujet âgé , Tumeurs du péritoine/génétique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/thérapie , Adulte , Adénocarcinome/génétique , Adénocarcinome/thérapie , Adénocarcinome/traitement médicamenteux , Adénocarcinome/anatomopathologie , Grading des tumeurs
13.
BMJ Case Rep ; 17(9)2024 Sep 26.
Article de Anglais | MEDLINE | ID: mdl-39327038

RÉSUMÉ

Cotyledonoid dissecting leiomyoma (CDL) is a rare benign uterine leiomyoma that macroscopically shows multinodular placenta-like growth. Its border with the myometrial layer is unclear, making it clinically difficult to differentiate from uterine sarcoma. CDL is often misdiagnosed. We report a case of CDL in which a subserosal myoma was suspected preoperatively and an abdominal myomectomy was performed. However, due to intraoperative findings and intraoperative rapid histopathological diagnosis, the procedure was changed to total hysterectomy. Peritoneal dissemination had also occurred and was resected simultaneously. It has been reported that CDL is generally a disease with good prognosis and that fertility preservation may be considered depending on the case. On the other hand, some cases of large tumours have caused peritoneal dissemination. We did a literature review of CDL and compared a group with peritoneal dissemination who underwent disseminated resection simultaneously and a group without peritoneal dissemination. We found that the tumour diameter was significantly greater in the peritoneal dissemination group.


Sujet(s)
Hystérectomie , Léiomyome , Tumeurs du péritoine , Tumeurs de l'utérus , Humains , Femelle , Léiomyome/chirurgie , Léiomyome/anatomopathologie , Léiomyome/diagnostic , Tumeurs de l'utérus/anatomopathologie , Tumeurs de l'utérus/chirurgie , Tumeurs de l'utérus/diagnostic , Tumeurs du péritoine/anatomopathologie , Tumeurs du péritoine/chirurgie , Tumeurs du péritoine/diagnostic , Tumeurs du péritoine/secondaire , Diagnostic différentiel , Myomectomie de l'utérus , Adulte
14.
BMJ Case Rep ; 17(8)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39214581

RÉSUMÉ

Diagnosing small bowel adenocarcinomas presents challenges due to non-specific symptoms, rarity and gastroscopy and colonoscopy's limited small intestine access, highlighting targeted diagnostic procedures' necessity. We present a late-diagnosed metastatic small bowel adenocarcinoma case in a man in his 80s who had asymptomatic mild iron-deficiency anaemia 1 year before diagnosis, with no active bleeding found on endoscopies. He experienced a single rectal bleeding episode 9 months prediagnosis, with subsequent severe iron-deficiency anaemia and no clear gastrointestinal source identified on gastroscopy. For 2 months, he had intermittent postprandial diarrhoea without abdominal pain, infectious or inflammatory causes. He experienced significant weight loss over 3 months prediagnosis. Subsequent gastroscopy indicated duodenal-gastric food retropulsion, suggesting a downstream blockage. Magnetic resonance enterography showed proximal jejunum thickening. Push enteroscopy confirmed jejunum adenocarcinoma. CT scans detected liver and peritoneal metastases. After one chemotherapy cycle, his condition worsened, leading to his passing 2 months post diagnosis.


Sujet(s)
Adénocarcinome , Tumeurs du jéjunum , Humains , Mâle , Adénocarcinome/secondaire , Adénocarcinome/diagnostic , Adénocarcinome/imagerie diagnostique , Adénocarcinome/anatomopathologie , Tumeurs du jéjunum/secondaire , Tumeurs du jéjunum/imagerie diagnostique , Tumeurs du jéjunum/diagnostic , Sujet âgé de 80 ans ou plus , Anémie par carence en fer/étiologie , Tumeurs du foie/secondaire , Tumeurs du foie/imagerie diagnostique , Tumeurs du foie/diagnostic , Hémorragie gastro-intestinale/étiologie , Tomodensitométrie , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/imagerie diagnostique , Tumeurs du péritoine/diagnostic , Intestin grêle/anatomopathologie , Intestin grêle/imagerie diagnostique , Diagnostic différentiel , Imagerie par résonance magnétique
15.
Gan To Kagaku Ryoho ; 51(7): 767-769, 2024 Jul.
Article de Japonais | MEDLINE | ID: mdl-39191697

RÉSUMÉ

A 64-year-old man complained of lower abdominal pain and vomiting. The CT scan showed adhesional ileus; therefore, small bowel resection procedure was performed. Histological findings showed signet-ring cell carcinoma and poorly differentiated adenocarcinoma. We performed ileocecal resection as an additional surgery. The operative findings revealed peritoneal nodules. The histological findings suggested goblet cell carcinoid with peritoneal dissemination. mFOLFOX+bevacizumab was administered, and no progression was observed for 30 months after the surgery. Appendiceal goblet cell carcinoid is rare and its prognosis is poor. Here, we report a case of appendiceal GCC that achieved a relatively long-term survival despite peritoneal dissemination.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs de l'appendice , Tumeur carcinoïde , Tumeurs du péritoine , Humains , Mâle , Adulte d'âge moyen , Tumeurs de l'appendice/anatomopathologie , Tumeurs de l'appendice/traitement médicamenteux , Tumeurs de l'appendice/chirurgie , Tumeur carcinoïde/traitement médicamenteux , Tumeur carcinoïde/chirurgie , Tumeur carcinoïde/anatomopathologie , Tumeur carcinoïde/secondaire , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/traitement médicamenteux , Tumeurs du péritoine/chirurgie , Facteurs temps , Leucovorine/administration et posologie , Leucovorine/usage thérapeutique , Bévacizumab/administration et posologie , Fluorouracil/administration et posologie , Composés organiques du platine/administration et posologie
16.
Glycobiology ; 34(10)2024 Aug 30.
Article de Anglais | MEDLINE | ID: mdl-39163480

RÉSUMÉ

Peritoneal metastasis frequently accompanies metastatic and/or recurrent gastric cancer, leading to a poor prognosis owing to a lack of effective treatment. Hence, there is a pressing need to enhance our understanding of the mechanisms and molecules driving peritoneal metastasis. In a previous study, galectin-4 inhibition impeded peritoneal metastasis in a murine model. This study examined the glycan profiles of cell surface proteins and glycosphingolipids (GSLs) in cells with varying tumorigenic potentials to understand the intricate mechanisms underlying galectin-4-mediated regulation, particularly glycosylation. Detailed mass spectrometry analysis showed that galectin-4 knockout cells exhibit increased expression of lacto-series GSLs with ß1,3-linked galactose while showing no significant alterations in neolacto-series GSLs. We conducted real-time polymerase chain reaction (PCR) analysis to identify candidate glycosyltransferases that synthesize increased levels of GSLs. Subsequently, we introduced the candidate B3GALT5 gene and selected the clones with high expression levels. B3GALT5 gene-expressing clones showed GSL glycan profiles like those of knockout cells and significantly reduced tumorigenic ability in mouse models. These clones exhibited diminished proliferative capacity and showed reduced expression of galectin-4 and activated AKT. Moreover, co-localization of galectin-4 with flotillin-2 (a raft marker) decreased in B3GALT5-expressing cells, implicating GSLs in galectin-4 localization to lipid rafts. D-threo-1-phenyl-2-decanoylamino-3-morpholino-1-propanol (a GSL synthase inhibitor) also affected galectin-4 localization in rafts, suggesting the involvement of GSL microdomains. We discovered that B3GALT5 plays a crucial role in regulating peritoneal metastasis of malignant gastric cancer cells by suppressing cell proliferation and modulating lipid rafts and galectin-4 via mechanisms that are yet to be elucidated.


Sujet(s)
Galactosyltransferases , Galectine 4 , Tumeurs de l'estomac , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Animaux , Humains , Souris , Galactosyltransferases/métabolisme , Galactosyltransferases/génétique , Galectine 4/métabolisme , Galectine 4/génétique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/métabolisme , Tumeurs du péritoine/génétique , Prolifération cellulaire , Différenciation cellulaire , Lignée cellulaire tumorale
17.
Adv Surg ; 58(1): 49-64, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39089786

RÉSUMÉ

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.


Sujet(s)
Tumeurs du côlon , Chimiothérapie hyperthermique intrapéritonéale , Mitomycine , Tumeurs du péritoine , Humains , Tumeurs du péritoine/thérapie , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/traitement médicamenteux , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/thérapie , Mitomycine/administration et posologie , Oxaliplatine/administration et posologie , Antinéoplasiques/administration et posologie , Composés organiques du platine/administration et posologie , Composés organiques du platine/usage thérapeutique , Interventions chirurgicales de cytoréduction , Résultat thérapeutique
18.
Eur J Surg Oncol ; 50(10): 108569, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39134081

RÉSUMÉ

BACKGROUND: Since 2016, staging laparoscopy has been implemented in the diagnostic workup of patients with gastric cancer. Staging laparoscopy aims to detect incurable disease (peritoneal metastases and irresectable tumors) and to prevent futile laparotomies. METHODS: In this population-based nationwide study, we sought patient- and tumor characteristics associated with undergoing a staging laparoscopy. Additionally, we analyzed the prevalence of synchronous peritoneal metastases, the outcome of the staging laparoscopy and its clinical impact on treatment decisions. All patients diagnosed with non-cardia gastric cancer from the Netherlands Cancer Registry between 2016 and 2021 were included. RESULTS: Alongside tumor characteristics, patient characteristics such as younger age, absence of comorbidities and lower WHO performance status were associated with performing a staging laparoscopy. In the study period, an increase in the proportion of patients who underwent a staging laparoscopy was observed, from 19.6% in 2016 to 32.3% in 2021 (p-value<0.001). In the same period, the prevalence of synchronous peritoneal metastases increased from 25% to 31%. In 37.6% of the patients who had the outcome of their staging laparoscopy reported, had incurable disease diagnosed during staging laparoscopy. Significantly less of these patients were treated with triplet regimens as compared to patients with a negative staging laparoscopy (18.5 vs. 76.3%; p-value<0.001). CONCLUSION: The implementation of staging laparoscopy in gastric cancer patients paralleled the increase in diagnosis of incurable disease and a decrease in the application of triplet systemic therapies in these patients.


Sujet(s)
Laparoscopie , Stadification tumorale , Tumeurs du péritoine , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/diagnostic , Femelle , Mâle , Pays-Bas/épidémiologie , Sujet âgé , Adulte d'âge moyen , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/diagnostic , Tumeurs du péritoine/épidémiologie , Enregistrements , Adulte , Sujet âgé de 80 ans ou plus , Facteurs âges
19.
Oncogene ; 43(37): 2768-2780, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39138263

RÉSUMÉ

Peritoneal metastasis is one of the most common risk factors contributing to the poor prognosis of gastric cancer. We previously reported that extracellular vesicles from gastric cancer cells could facilitate peritoneal metastasis. However, their impact on gastric cancer-induced peritoneal metastasis under hypoxic conditions remains unclear. This study aims to elucidate how hypoxia-resistant gastric cancer cell-derived extracellular vesicles affect the peritoneal metastasis of normoxic gastric cancer cells. Proteomic analysis revealed elevated levels of Caveolin1 and Laminin ß2 in hypoxia-resistant gastric cancer cells and their corresponding extracellular vesicles. Importantly, Caveolin1 was found to play a central role in mediating Laminin ß2 sorting into extracellular vesicles derived from hypoxia-resistant gastric cancer cells, and subsequently, extracellular vesicle-associated Laminin ß2 promoted peritoneal metastasis in normoxic gastric cancer cells by activating the AKT pathway. Further investigation confirmed that Caveolin1 activation by Rho-related Coiled-coil kinase 1-mediated phosphorylation of Y14 residue is a key factor facilitating Laminin ß2 sorting into extracellular vesicles. Moreover, Y14 phosphorylated- Caveolin1 enhanced Laminin ß2 sorting by activating Rab11. Finally, our study demonstrated that a combined assessment of plasma extracellular vesicle-associated Caveolin1 and extracellular vesicle-associated Laminin ß2 could provide an accurate predictive tool for peritoneal metastasis occurrence in gastric cancer.


Sujet(s)
Cavéoline-1 , Vésicules extracellulaires , Tumeurs du péritoine , Tumeurs de l'estomac , Protéines G rab , rho-Associated Kinases , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/métabolisme , Tumeurs de l'estomac/génétique , Humains , Cavéoline-1/métabolisme , Cavéoline-1/génétique , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/métabolisme , Animaux , rho-Associated Kinases/métabolisme , Vésicules extracellulaires/métabolisme , Souris , Protéines G rab/métabolisme , Protéines G rab/génétique , Lignée cellulaire tumorale , Transduction du signal , Mâle , Femelle
20.
Sci Rep ; 14(1): 19103, 2024 08 17.
Article de Anglais | MEDLINE | ID: mdl-39154083

RÉSUMÉ

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.


Sujet(s)
Récidive tumorale locale , Nomogrammes , Tumeurs du péritoine , Tumeurs de l'estomac , Humains , Tumeurs de l'estomac/chirurgie , Tumeurs de l'estomac/anatomopathologie , Tumeurs de l'estomac/mortalité , Mâle , Femelle , Adulte d'âge moyen , Tumeurs du péritoine/chirurgie , Tumeurs du péritoine/secondaire , Tumeurs du péritoine/mortalité , Sujet âgé , Appréciation des risques/méthodes , Courbe ROC , Pronostic , Gastrectomie , Traitement médicamenteux adjuvant , Adulte , Facteurs de risque
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE