1
|
Lee J, Han Y, Kim S, Jo H, Wang W, Cho U, Kim SI, Kim B, Song YS. Mitochondrial fission enhances IL-6-induced metastatic potential in ovarian cancer via ERK1/2 activation. Cancer Sci 2024; 115:1536-1550. [PMID: 38433313 PMCID: PMC11093201 DOI: 10.1111/cas.16064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 11/22/2023] [Accepted: 12/18/2023] [Indexed: 03/05/2024] Open
Abstract
Ovarian cancer is a lethal gynecologic cancer mostly diagnosed in an advanced stage with an accumulation of ascites. Interleukin-6 (IL-6), a pro-inflammatory cytokine is highly elevated in malignant ascites and plays a pleiotropic role in cancer progression. Mitochondria are dynamic organelles that undergo fission and fusion in response to external stimuli and dysregulation in their dynamics has been implicated in cancer progression and metastasis. Here, we investigate the effect of IL-6 on mitochondrial dynamics in ovarian cancer cells (OVCs) and its impact on metastatic potential. Treatment with IL-6 on ovarian cancer cell lines (SKOV3 and PA-1) led to an elevation in the metastatic potential of OVCs. Interestingly, a positive association was observed between dynamin-related protein 1 (Drp1), a regulator of mitochondrial fission, and IL-6R in metastatic ovarian cancer tissues. Additionally, IL-6 treatment on OVCs was linked to the activation of Drp1, with a notable increase in the ratio of the inhibitory form p-Drp1(S637) to the active form p-Drp1(S616), indicating enhanced mitochondrial fission. Moreover, IL-6 treatment triggered the activation of ERK1/2, and inhibiting ERK1/2 mitigated IL-6-induced mitochondrial fission. Suppressing mitochondrial fission through siRNA transfection and a pharmacological inhibitor reduced the IL-6-induced migration and invasion of OVCs. This was further supported by 3D invasion assays using patient-derived spheroids. Altogether, our study suggests the role of mitochondrial fission in the metastatic potential of OVCs induced by IL-6. The inhibition of mitochondrial fission could be a potential therapeutic approach to suppress the metastasis of ovarian cancer.
Collapse
Affiliation(s)
- Juwon Lee
- WCU Biomodulation, Department of Agricultural BiotechnologySeoul National UniversitySeoulKorea
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
| | - Youngjin Han
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
| | - Soochi Kim
- Department of Neurology and Neurological SciencesStanford University School of MedicineStanfordCaliforniaUSA
- Paul F. Glenn Laboratories for the Biology of AgingStanford University School of MedicineStanfordCaliforniaUSA
| | - HyunA Jo
- WCU Biomodulation, Department of Agricultural BiotechnologySeoul National UniversitySeoulKorea
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
| | - Wenyu Wang
- Department of Medical Oncology, The First Affiliated Hospital, College of MedicineZhejiang UniversityHangzhouChina
| | - Untack Cho
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
| | - Se Ik Kim
- Department of Obstetrics and Gynecology, College of MedicineSeoul National UniversitySeoulKorea
| | - Boyun Kim
- Department of SmartBio, College of Life and Health ScienceKyungsung UniversityBusanKorea
| | - Yong Sang Song
- WCU Biomodulation, Department of Agricultural BiotechnologySeoul National UniversitySeoulKorea
- Cancer Research Institute, College of MedicineSeoul National UniversitySeoulKorea
- Department of Obstetrics and Gynecology, College of MedicineSeoul National UniversitySeoulKorea
| |
Collapse
|
2
|
Mulugeta A, Lumsden AL, Madakkatel I, Stacey D, Lee SH, Mäenpää J, Oehler MK, Hyppönen E. Phenome-wide association study of ovarian cancer identifies common comorbidities and reveals shared genetics with complex diseases and biomarkers. Cancer Med 2024; 13:e7051. [PMID: 38457211 PMCID: PMC10923028 DOI: 10.1002/cam4.7051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 01/29/2024] [Accepted: 02/09/2024] [Indexed: 03/09/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is commonly diagnosed among older women who have comorbidities. This hypothesis-free phenome-wide association study (PheWAS) aimed to identify comorbidities associated with OC, as well as traits that share a genetic architecture with OC. METHODS We used data from 181,203 white British female UK Biobank participants and analysed OC and OC subtype-specific genetic risk scores (OC-GRS) for an association with 889 diseases and 43 other traits. We conducted PheWAS and colocalization analyses for individual variants to identify evidence for shared genetic architecture. RESULTS The OC-GRS was associated with 10 diseases, and the clear cell OC-GRS was associated with five diseases at the FDR threshold (p = 5.6 × 10-4 ). Mendelian randomizaiton analysis (MR) provided robust evidence for the association of OC with higher risk of "secondary malignant neoplasm of digestive systems" (OR 1.64, 95% CI 1.33, 2.02), "ascites" (1.48, 95% CI 1.17, 1.86), "chronic airway obstruction" (1.17, 95% CI 1.07, 1.29), and "abnormal findings on examination of the lung" (1.51, 95% CI 1.22, 1.87). Analyses of lung spirometry measures provided further support for compromised respiratory function. PheWAS on individual OC variants identified five genetic variants associated with other diseases, and seven variants associated with biomarkers (all, p ≤ 4.5 × 10-8 ). Colocalization analysis identified rs4449583 (from TERT locus) as the shared causal variant for OC and seborrheic keratosis. CONCLUSIONS OC is associated with digestive and respiratory comorbidities. Several variants affecting OC risk were associated with other diseases and biomarkers, with this study identifying a novel genetic locus shared between OC and skin conditions.
Collapse
Affiliation(s)
- Anwar Mulugeta
- Australian Centre for Precision Health, Unit of Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- South Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- Department of Pharmacology and Clinical Pharmacy, College of Health ScienceAddis Ababa UniversityAddis AbabaEthiopia
| | - Amanda L. Lumsden
- Australian Centre for Precision Health, Unit of Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- South Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - Iqbal Madakkatel
- Australian Centre for Precision Health, Unit of Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- South Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - David Stacey
- Australian Centre for Precision Health, Unit of Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- South Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| | - S. Hong Lee
- Australian Centre for Precision Health, Unit of Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- South Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
- UniSA Allied Health & Human PerformanceUniversity of South AustraliaAdelaideSouth AustraliaAustralia
| | - Johanna Mäenpää
- Faculty of Medicine and Medical TechnologyTampere UniversityTampereFinland
- Cancer Centre, Tampere University and University HospitalTampereFinland
| | - Martin K. Oehler
- Department of Gynaecological OncologyRoyal Adelaide HospitalAdelaideSouth AustraliaAustralia
- Adelaide Medical School, Robinson Research Institute, University of AdelaideAdelaideSouth AustraliaAustralia
| | - Elina Hyppönen
- Australian Centre for Precision Health, Unit of Clinical and Health SciencesUniversity of South AustraliaAdelaideSouth AustraliaAustralia
- South Australian Health and Medical Research InstituteAdelaideSouth AustraliaAustralia
| |
Collapse
|
3
|
Berger JM, Alany A, Berchtold L, Puhr R, Friedrich A, Scheiner B, Prager GW, Preusser M, Berghoff AS, Bergen ES. Prognosticators of survival in patients with metastatic pancreatic cancer and ascites. ESMO Open 2023; 8:102048. [PMID: 37977000 PMCID: PMC10774951 DOI: 10.1016/j.esmoop.2023.102048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/27/2023] [Accepted: 09/28/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND Identification of factors associated with survival after ascites diagnosis in metastatic pancreatic cancer (mPC) patients may guide treatment decisions and help to maintain quality of life in this highly symptomatic patient collective. PATIENTS AND METHODS All patients treated for mPC at the Medical University of Vienna between 2010 and 2019 developing ascites throughout their course of disease were identified by retrospective chart review. General risk factors, metastatic sites, systemic inflammation and liver function parameters, as well as type of treatment after ascites diagnosis were investigated for associations with survival. RESULTS One hundred and seventeen mPC patients with ascites were included in this study. Median time from mPC to ascites diagnosis was 8.9 months (range 0-99 months) and median overall survival (OS) after ascites diagnosis was 27.4 days (range 21.3-42.6 days). Identified prognostic factors at ascites diagnosis independently associated with an impaired OS were presence of liver metastases [hazard ratio (HR): 2.07, 95% confidence interval (CI) 1.13-3.79, P = 0.018), peritoneal carcinomatosis (HR: 1.74, 95% CI 1.11-2.71, P = 0.015), and portal vein obstruction (HR: 2.52, 95% CI 1.29-4.90, P = 0.007). Compared with best supportive care, continuation of systemic therapy after ascites diagnosis was independently associated with survival (HR: 0.35, 95% CI 0.20-0.61, P < 0.001) with a median OS of 62 days (95% CI 51-129 days, P < 0.001) versus 16 days (95% CI 11-24 days), respectively. CONCLUSIONS Liver and peritoneal metastases as well as portal vein obstruction were found to be prognostic factors after ascites diagnosis in mPC patients. Continuation of systemic therapy after ascites diagnosis was associated with a longer OS, which needs to be evaluated in larger clinical trials including quality-of-life assessment.
Collapse
Affiliation(s)
- J M Berger
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - A Alany
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - L Berchtold
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - R Puhr
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - A Friedrich
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - B Scheiner
- Division of Gastroenterology and Hepatology, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - G W Prager
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna
| | - M Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - A S Berghoff
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna; Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Vienna
| | - E S Bergen
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Vienna.
| |
Collapse
|
4
|
Qian L, Sun R, Xue Z, Guo T. Mass Spectrometry-based Proteomics of Epithelial Ovarian Cancers: a Clinical Perspective. Mol Cell Proteomics 2023:100578. [PMID: 37209814 PMCID: PMC10388592 DOI: 10.1016/j.mcpro.2023.100578] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 05/08/2023] [Accepted: 05/16/2023] [Indexed: 05/22/2023] Open
Abstract
Increasing proteomic studies focused on epithelial ovarian cancer (EOC) have attempted to identify early disease biomarkers, establish molecular stratification, and discover novel druggable targets. Here we review these recent studies from a clinical perspective. Multiple blood proteins have been used clinically as diagnostic markers. The ROMA test integrates CA125 and HE4, while the OVA1 and OVA2 tests analyze multiple proteins identified by proteomics. Targeted proteomics has been widely used to identify and validate potential diagnostic biomarkers in EOCs, but none has yet been approved for clinical adoption. Discovery proteomic characterization of bulk EOC tissue specimens has uncovered a large number of dysregulated proteins, proposed new stratification schemes, and revealed novel targets of therapeutic potential. A major hurdle facing clinical translation of these stratification schemes based on bulk proteomic profiling is intra-tumor heterogeneity, namely that single tumor specimens may harbor molecular features of multiple subtypes. We reviewed over 2500 interventional clinical trials of ovarian cancers since 1990, and cataloged 22 types of interventions adopted in these trials. Among 1418 clinical trials which have been completed or are not recruiting new patients, about 50% investigated chemotherapies. Thirty-seven clinical trials are at phase 3 or 4, of which 12 focus on PARP, 10 on VEGFR, 9 on conventional anti-cancer agents, and the remaining on sex hormones, MEK1/2, PD-L1, ERBB, and FRα. Although none of the foregoing therapeutic targets were discovered by proteomics, newer targets discovered by proteomics, including HSP90 and cancer/testis antigens, are being tested also in clinical trials. To accelerate the translation of proteomic findings to clinical practice, future studies need to be designed and executed to the stringent standards of practice-changing clinical trials. We anticipate that the rapidly evolving technology of spatial and single-cell proteomics will deconvolute the intra-tumor heterogeneity of EOCs, further facilitating their precise stratification and superior treatment outcomes.
Collapse
Affiliation(s)
- Liujia Qian
- iMarker lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang, 310030, China.
| | - Rui Sun
- iMarker lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang, 310030, China
| | - Zhangzhi Xue
- iMarker lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang, 310030, China
| | - Tiannan Guo
- iMarker lab, Westlake Laboratory of Life Sciences and Biomedicine, Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang Province, China; Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang Province, China; Research Center for Industries of the Future, Westlake University, 600 Dunyu Road, Hangzhou, Zhejiang, 310030, China.
| |
Collapse
|
5
|
Berger J, Alany A, Puhr R, Berchtold L, Friedrich A, Scheiner B, Prager G, Berghoff A, Preusser M, Bergen E. Clinical risk factors for ascites in metastatic pancreatic cancer. ESMO Open 2023; 8:101200. [PMID: 36989885 PMCID: PMC10163163 DOI: 10.1016/j.esmoop.2023.101200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/22/2023] [Accepted: 02/23/2023] [Indexed: 03/29/2023] Open
Abstract
BACKGROUND Malignant ascites is common in metastatic pancreatic cancer (mPC) and its management still remains a clinical challenge. Early identification of patients at risk for ascites development may support and guide treatment decisions. MATERIALS AND METHODS Data of patients treated for mPC at the Medical University of Vienna between 2010 and 2019 were collected by retrospective chart review. Ascites was defined as clinically relevant accumulation of intraperitoneal fluid diagnosed by ultrasound or computer tomography scan of the abdomen. We investigated the association between general risk factors, metastatic sites, liver function, systemic inflammation as well as portal vein obstruction (PVO) and ascites development. RESULTS Among 581 patients with mPC included in this study, 122 (21.0%) developed ascites after a median of 8.7 months after diagnosis of metastatic disease. The occurrence of ascites led to an 8.9-fold increased risk of death [confidence interval (CI) 7.2-11, P < 0.001] with a median overall survival of 1 month thereafter. Clinical risk factors for ascites were male sex [hazard ratio (HR) 1.71, CI 1.00-2.90, P = 0.048], peritoneal carcinomatosis (HR 6.79, CI 4.09-11.3, P < 0.001), liver metastases (HR 2.16, CI 1.19-3.91, P = 0.011), an albumin-bilirubin (ALBI) score grade 3 (HR 6.79, CI 2.11-21.8, P = 0.001), PVO (HR 2.28, CI 1.15-4.52, P = 0.019), and an elevated C-reactive protein (CRP) (HR 4.19, CI 1.58-11.1, P = 0.004). CONCLUSIONS Survival after diagnosis of ascites is very limited in mPC patients. Male sex, liver and peritoneal metastases, impaired liver function, PVO, as well as systemic inflammation were identified as independent risk factors for ascites development in this uniquely large real-life patient cohort.
Collapse
|
6
|
Han MY, Borazanci EH. Malignant ascites in pancreatic cancer: Pathophysiology, diagnosis, molecular characterization, and therapeutic strategies. Front Oncol 2023; 13:1138759. [PMID: 37007072 PMCID: PMC10060830 DOI: 10.3389/fonc.2023.1138759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 03/06/2023] [Indexed: 03/18/2023] Open
Abstract
Malignant ascites is the accumulation of fluid in the peritoneum as a result of advanced cancer and often signifies the terminal phase of the disease. Management of malignant ascites remains a clinical challenge as symptom palliation is the current standard of cure. Previously, studies examining malignant ascites largely focused on ovarian and gastric cancer. In recent years, there has been a significant increase in research on malignant ascites in pancreatic cancer. Malignant ascites is usually diagnosed based on positive cytology, but cytology is not always diagnostic, indicating the need for novel diagnostic tools and biomarkers. This review aims to summarize the current understanding of malignant ascites in pancreatic cancer and the recent advances in the molecular characterization of malignant ascites fluid from patients with pancreatic cancer including analysis of soluble molecules and extracellular vesicles. Current standard of care treatment options such as paracenteses and diuretics are outlined along with new emerging treatment strategies such as immunotherapy and small-molecule based therapies. New potential investigative directions resulting from these studies are also highlighted.
Collapse
Affiliation(s)
- Margaret Y. Han
- Department of Biosciences, Rice University, Houston, TX, United States
| | - Erkut H. Borazanci
- Department of Oncology, HonorHealth Research Institute, Scottsdale, AZ, United States
- *Correspondence: Erkut H. Borazanci,
| |
Collapse
|
7
|
Jha DK, Rohilla M, Das CK, Irrinki S, Singh H, Arora A, Saha SC, Gupta P, Mandavdhare HS, Dutta U, Sharma A, Sharma V. Randomized crossover trial of 'Roll-over' technique of abdominal paracentesis versus standard technique in suspected malignant ascites. Expert Rev Gastroenterol Hepatol 2023; 17:295-300. [PMID: 36795510 DOI: 10.1080/17474124.2023.2181785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 02/14/2023] [Indexed: 02/17/2023]
Abstract
BACKGROUND The sensitivity of single abdominal paracentesis for diagnosis of peritoneal carcinomatosis (PC) varies from 40-70%. We hypothesized that rolling-over the patient before paracentesis might improve the cytological yield. RESEARCH DESIGN AND METHODS This was a single center pilot study with a randomized cross-over design. We compared the cytological yield of fluid obtained by roll-over technique (ROG) with standard paracentesis (SPG) in suspected PC. In the ROG group, patients were rolled side-to-side thrice, and the paracentesis was done within 1 minute. Each patient served as their own control, and the outcome assessor (cytopathologist) was blinded. The primary objective was to compare the tumor cell positivity between SPG and ROG groups. RESULTS Of 71 patients, 62 were analyzed. Of 53 patients with malignancy-related ascites, 39 had PC. Most of the tumor cells were adenocarcinoma (30, 94%) with one patient each having suspicious cytology and one having lymphoma. The sensitivity for diagnosis of PC was (31/39) 79.49% in SPG group and (32/39) 82.05% in ROG group (p = 1.00). The cellularity was similar between both the groups (good cellularity in 58% of SPG and 60% of ROG, p = 1.00). CONCLUSIONS Rollover paracentesis did not improve the cytological yield of abdominal paracentesis. TRIAL REGISTRATION CTRI/2020/06/025887 and NCT04232384.
Collapse
Affiliation(s)
- Daya Krishna Jha
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Manish Rohilla
- Department of Cytopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Chandan K Das
- Department of Medical Oncology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Santhosh Irrinki
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harjeet Singh
- Department of Surgical Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aashima Arora
- Department of Obstetrics and Gynecology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Subhas C Saha
- Department of Obstetrics and Gynecology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Pankaj Gupta
- Department of Radiodiagnosis, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Harshal S Mandavdhare
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Usha Dutta
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Aman Sharma
- Internal Medicine, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vishal Sharma
- Department of Gastroenterology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
8
|
Seah DS, Wilcock A, Chang S, Sousa MS, Sinnarajah A, Teoh CO, Allan S, Chye R, Doogue M, Hunt J, Agar M, Currow DC. Paracentesis for cancer-related ascites in palliative care: An international, prospective cohort study. Palliat Med 2022; 36:1408-1417. [PMID: 36113139 DOI: 10.1177/02692163221122326] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Paracentesis is commonly undertaken in patients with cancer-related ascites. AIM To systematically investigate the symptomatic benefits and harms experienced by patients with cancer undergoing paracentesis using real-world data in the palliative care setting. DESIGN Prospective, multisite, observational, consecutive cohort study. Benefits and harms of paracentesis were assessed between 01/07/2018 and 31/02/2021 as part of routine clinical assessments by treating clinicians at four timepoints: (T0) before paracentesis; (T1) once drainage ceased; (T2) 24 h after T1 and (T3) 28 days after T1 or next paracentesis, if sooner. SETTING/PARTICIPANTS Data were collected from 11 participating sites across five countries (Australia, England, Hong Kong, Malaysia and New Zealand) on 111 patients undergoing paracentesis via a temporary (73%) or indwelling (21%) catheter: 51% male, median age 69 years, Australia-modified Karnofsky Performance Score 50. RESULTS At T1 (n = 100), symptoms had improved for most patients (81%), specifically abdominal distension (61%), abdominal pain (49%) and nausea (27%), with two-thirds experiencing improvement in ⩾2 symptoms. In the remaining patients, symptoms were unchanged (7%) or worse (12%). At least one harm occurred in 32% of patients, the most common being an ascitic leak (n = 14). By T3, 89% of patients had experienced some benefit and 36% some harm, including four patients who experienced serious harm, one of which was a fatal bowel perforation. CONCLUSION Most patients obtained rapid benefits from paracentesis. Harms were less frequent and generally mild, but occasionally serious and fatal. Our findings help inform clinician-patient discussions about the potential outcomes of paracentesis in this frail population.
Collapse
Affiliation(s)
- Davinia Se Seah
- IMPACCT-Improving Palliative, Aged and Chronic Care through Clinical Research and Translation, Faculty of Health, University of Technology Sydney, Sydney, NSW, Australia.,Sacred Heart Health Service, Sydney, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Andrew Wilcock
- Hayward House Specialist Palliative Care Unit, School of Clinical Oncology, University of Nottingham, Nottingham, England
| | - Sungwon Chang
- IMPACCT-Improving Palliative, Aged and Chronic Care through Clinical Research and Translation, Faculty of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Mariana S Sousa
- IMPACCT-Improving Palliative, Aged and Chronic Care through Clinical Research and Translation, Faculty of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Aynharan Sinnarajah
- Division of Palliative Medicine, Department of Medicine, School of Medicine, Queen's University, Kingston, ON, Canada
| | | | | | - Richard Chye
- IMPACCT-Improving Palliative, Aged and Chronic Care through Clinical Research and Translation, Faculty of Health, University of Technology Sydney, Sydney, NSW, Australia.,Sacred Heart Health Service, Sydney, NSW, Australia.,St Vincent's Clinical School, University of New South Wales, Sydney, NSW, Australia
| | - Matthew Doogue
- University of Otago - Christchurch & Canterbury District Health Board, Christchurch, New Zealand
| | - Jane Hunt
- IMPACCT-Improving Palliative, Aged and Chronic Care through Clinical Research and Translation, Faculty of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - Meera Agar
- IMPACCT-Improving Palliative, Aged and Chronic Care through Clinical Research and Translation, Faculty of Health, University of Technology Sydney, Sydney, NSW, Australia
| | - David C Currow
- Faculty of Science, Medicine and Health, University of Wollongong, Wollongong, Australia
| |
Collapse
|
9
|
Hendrikson J, Liu Y, Ng WH, Lee JY, Lim AH, Loh JW, Ng CC, Ong WS, Tan JWS, Tan QX, Ng G, Shannon NB, Lim WK, Lim TK, Chua C, Wong JSM, Tan GHC, So JBY, Yeoh KG, Teh BT, Chia CS, Soo KC, Kon OL, Tan IB, Chan JY, Teo MCC, Ong CAJ. Ligand-mediated PAI-1 inhibition in a mouse model of peritoneal carcinomatosis. Cell Rep Med 2022; 3:100526. [PMID: 35243423 PMCID: PMC8861959 DOI: 10.1016/j.xcrm.2022.100526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 12/28/2022]
Abstract
Peritoneal carcinomatosis (PC) present a ubiquitous clinical conundrum in all intra-abdominal malignancies. Via functional and transcriptomic experiments of ascites-treated PC cells, we identify STAT3 as a key signaling pathway. Integrative analysis of publicly available databases and correlation with clinical cohorts (n = 7,359) reveal putative clinically significant activating ligands of STAT3 signaling. We further validate a 3-biomarker prognostic panel in ascites independent of clinical covariates in a prospective study (n = 149). Via single-cell sequencing experiments, we uncover that PAI-1, a key component of the prognostic biomarker panel, is largely secreted by fibroblasts and mesothelial cells. Molecular stratification of ascites using PAI-1 levels and STAT3 activation in ascites-treated cells highlight a therapeutic opportunity based on a phenomenon of paracrine addiction. These results are recapitulated in patient-derived ascites-dependent xenografts. Here, we demonstrate therapeutic proof of concept of direct ligand inhibition of a prognostic target within an enclosed biological space.
Collapse
Affiliation(s)
- Josephine Hendrikson
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Ying Liu
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Wai Har Ng
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Abner Herbert Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jui Wan Loh
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Cedric C.Y. Ng
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Whee Sze Ong
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Joey Wee-Shan Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Qiu Xuan Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Gillian Ng
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Nicholas B. Shannon
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Weng Khong Lim
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169609, Singapore
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Tony K.H. Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
- Pathology Academic Clinical Program, SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
| | - Clarinda Chua
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jolene Si Min Wong
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Grace Hwei Ching Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Jimmy Bok Yan So
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Division of Surgical Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Khay Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore 119074, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
| | - Claramae Shulyn Chia
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Khee Chee Soo
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Oi Lian Kon
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Iain Beehuat Tan
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- Laboratory of Applied Cancer Genomics, Genome Institute of Singapore, A∗STAR Research Entities, Singapore 138672, Singapore
| | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Melissa Ching Ching Teo
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Chin-Ann J. Ong
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
| | - on behalf of the Singapore Peritoneal Oncology Study (SPOS) Group and Singapore Gastric Cancer Consortium (SGCC)
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169609, Singapore
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
- Pathology Academic Clinical Program, SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Division of Surgical Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
- Laboratory of Applied Cancer Genomics, Genome Institute of Singapore, A∗STAR Research Entities, Singapore 138672, Singapore
| |
Collapse
|
10
|
Sun Y, Hu Y, Wan C, Lovell JF, Jin H, Yang K. Local biomaterial-assisted antitumour immunotherapy for effusions in the pleural and peritoneal cavities caused by malignancies. Biomater Sci 2021; 9:6381-6390. [PMID: 34582527 DOI: 10.1039/d1bm00971k] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Malignant pleural effusion (MPE) and malignant ascites (MA), which are common but serious conditions caused by malignancies, are related to poor quality of life and high mortality. Current treatments, including therapeutic thoracentesis and indwelling pleural catheters or paracentesis and catheter drainage, are largely palliative. An effective treatment is urgently needed. MPE and MA are excellent candidates for intratumoural injections that have direct contact with tumour cells and kill tumour cells more effectively and efficiently with fewer side effects, and the fluid environment of MPE and MA can provide a homogeneous area for drug distribution. The immunosuppressive environments within the pleural and peritoneal cavities suggest the feasibility of local immunotherapy. In this review, we introduce the current management of MPE and MA, discuss the latest advances and challenges in utilizing local biomaterial-assisted antitumour therapies for the treatment of MPE and MA, and discuss further opportunities in this field.
Collapse
Affiliation(s)
- Yajie Sun
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Yan Hu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Chao Wan
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Jonathan F Lovell
- Department of Chemical and Biological Engineering, University at Buffalo, State University of New York. Buffalo, New York, 14260, USA
| | - Honglin Jin
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Kunyu Yang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
11
|
Zhang S, Xie B, Wang L, Yang H, Zhang H, Chen Y, Wang F, Liu C, He H. Macrophage-mediated vascular permeability via VLA4/VCAM1 pathway dictates ascites development in ovarian cancer. J Clin Invest 2021; 131:140315. [PMID: 33295887 DOI: 10.1172/jci140315] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 11/25/2020] [Indexed: 12/14/2022] Open
Abstract
The development of ascites correlates with advanced stage disease and poor prognosis in ovarian cancer. Vascular permeability is the key pathophysiological change involved in ascites development. Previously, we provided evidence that perivascular M2-like macrophages protect the vascular barrier through direct contact with endothelial cells (ECs). Here, we investigated the molecular mechanism and its clinical significance in the ovarian cancer setting. We found that upon direct coculture with the endothelium, M2 macrophages tuned down their VLA4 and reduced the levels of VCAM1 in ECs. On the other hand, ectopically overexpressing VLA4 in macrophages or VCAM1 in ECs induced hyperpermeability. Mechanistically, downregulation of VLA4 or VCAM1 led to reduced levels of RAC1 and ROS, which resulted in decreased phosphorylation of PYK2 (p-PYK2) and VE-cadherin (p-VE-cad), hence enhancing cell adhesion. Furthermore, targeting the VLA4/VCAM1 axis augmented vascular integrity and abrogated ascites formation in vivo. Finally, VLA4 expression on the macrophages isolated from ascites dictated permeability ex vivo. Importantly, VLA4 antibody acted synergistically with bevacizumab to further enhance the vascular barrier. Taking these data together, we reveal here that M2 macrophages regulate the vascular barrier though the VCAM1/RAC1/ROS/p-PYK2/p-VE-cad cascade, which provides specific therapeutic targets for the treatment of malignant ascites.
Collapse
Affiliation(s)
- Shibo Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Bingfan Xie
- Department of Gynaecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Lijie Wang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Hua Yang
- Department of Gynaecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Haopei Zhang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Yuming Chen
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Feng Wang
- Department of Gynaecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Changqing Liu
- Department of Gynaecology, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Huanhuan He
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
12
|
Chan KP, Badiei A, Tan CPS, Fitzgerald DB, Stanley C, Fysh ETH, Shrestha R, Muruganandan S, Read CA, Thomas R, Lee YCG. Use of indwelling pleural/peritoneal catheter in the management of malignant ascites: a retrospective study of 48 patients. Intern Med J 2021; 50:705-711. [PMID: 31566871 DOI: 10.1111/imj.14642] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 08/10/2019] [Accepted: 09/08/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND Patients suffering from malignant ascites usually require repeated large volume paracentesis (LVP) for symptomatic relief. This often requires hospital admission and has inherent risks. AIMS To report the first Australian experience of placing tunnelled indwelling peritoneal catheters (IPeC) for management of recurrent malignant ascites. METHODS A retrospective study was conducted of tunnelled IPeC use in patients with symptomatic malignant ascites in four hospitals in Western Australia (from 2010 to 2018). Procedure data, success rate and safety profile were collected from a database. RESULTS Forty-eight patients (median age 65 years; female 56%) underwent 51 peritoneal catheter insertion procedures that were performed mostly by pleural specialists. The majority of patients (96%) had prior LVP (median two drainages, interquartile range (IQR) 1-4) before IPeC insertion. The IPeC was inserted successfully under ultrasound guidance in all patients. The median length of hospital stay for IPeC insertion and initial ascites drainage was 2 days (IQR 2-3 days) and most patients (96%) did not require further paracentesis after IPeC placement. The majority (96%) of patients experienced relief from ascites symptoms after catheter insertion. Most IPeC-related adverse events were self-limiting, including pain (in 25% cases), transient hypotension after initial fluid drainage (10%), peritoneal fluid leakage (10%), bacterial peritonitis (8%), fluid loculation (2%) and catheter dislodgement (2%). Six (12%) patients had IPeC removed. All patients with bacterial peritonitis responded to antibiotics and one required catheter removal. CONCLUSIONS Use of tunnelled IPeC improves symptoms and can minimise further invasive drainage procedures in patients with symptomatic malignant ascites. Placement of IPeC was associated with a low rate of adverse events, most of which could be managed conservatively.
Collapse
Affiliation(s)
- Ka P Chan
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,Department of Medicine and Therapeutics, Chinese University of Hong Kong, Hong Kong
| | - Arash Badiei
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Carmen P S Tan
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Deirdre B Fitzgerald
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Christopher Stanley
- Department of Respiratory Medicine, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Edward T H Fysh
- Department of Respiratory Medicine, St John of God Midland Public Hospital, Perth, Western Australia, Australia
| | - Ranjan Shrestha
- Department of Respiratory Medicine, Fiona Stanley Hospital, Perth, Western Australia, Australia
| | - Sanjeevan Muruganandan
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Catherine A Read
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| | - Rajesh Thomas
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| | - Yun Chor Gary Lee
- Department of Respiratory Medicine, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia.,School of Medicine and Pharmacology, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
13
|
Asem M, Young A, Oyama C, ClaureDeLaZerda A, Liu Y, Ravosa MJ, Gupta V, Jewell A, Khabele D, Stack MS. Ascites-induced compression alters the peritoneal microenvironment and promotes metastatic success in ovarian cancer. Sci Rep 2020; 10:11913. [PMID: 32681052 PMCID: PMC7367827 DOI: 10.1038/s41598-020-68639-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 06/26/2020] [Indexed: 12/17/2022] Open
Abstract
The majority of women with recurrent ovarian cancer (OvCa) develop malignant ascites with volumes that can reach > 2 L. The resulting elevation in intraperitoneal pressure (IPP), from normal values of 5 mmHg to as high as 22 mmHg, causes striking changes in the loading environment in the peritoneal cavity. The effect of ascites-induced changes in IPP on OvCa progression is largely unknown. Herein we model the functional consequences of ascites-induced compression on ovarian tumor cells and components of the peritoneal microenvironment using a panel of in vitro, ex vivo and in vivo assays. Results show that OvCa cell adhesion to the peritoneum was increased under compression. Moreover, compressive loads stimulated remodeling of peritoneal mesothelial cell surface ultrastructure via induction of tunneling nanotubes (TNT). TNT-mediated interaction between peritoneal mesothelial cells and OvCa cells was enhanced under compression and was accompanied by transport of mitochondria from mesothelial cells to OvCa cells. Additionally, peritoneal collagen fibers adopted a more linear anisotropic alignment under compression, a collagen signature commonly correlated with enhanced invasion in solid tumors. Collectively, these findings elucidate a new role for ascites-induced compression in promoting metastatic OvCa progression.
Collapse
Affiliation(s)
- Marwa Asem
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Allison Young
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Carlysa Oyama
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Alejandro ClaureDeLaZerda
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Yueying Liu
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
| | - Matthew J Ravosa
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Vijayalaxmi Gupta
- Department of Obstetrics & Gynecology, Medical Center, University of Kansas, Kansas City, USA
| | - Andrea Jewell
- Department of Obstetrics & Gynecology, Medical Center, University of Kansas, Kansas City, USA
| | - Dineo Khabele
- Department of Obstetrics & Gynecology, Medical Center, University of Kansas, Kansas City, USA
| | - M Sharon Stack
- Department of Chemistry & Biochemistry, University of Notre Dame, Notre Dame, IN, USA.
- Harper Cancer Research Institute, University of Notre Dame, 1234 N. Notre Dame Ave., A200 Harper Hall, South Bend, IN, 46617, USA.
| |
Collapse
|
14
|
Ito T, Hanafusa N, Iwase S, Noiri E, Nangaku M, Nakagawa K, Miyagawa K. Ascitic IL-10 Concentration Predicts Prognosis of Patients Undergoing Cell-Free and Concentrated Ascites Reinfusion Therapy. Ther Apher Dial 2019; 24:90-95. [PMID: 31157953 DOI: 10.1111/1744-9987.12863] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 05/31/2019] [Indexed: 01/08/2023]
Abstract
Cell-free and concentrated ascites reinfusion therapy (CART) is now attracting rising attention as one of the strategies against cancer-related malignant ascites in Japan. Several studies report the safety, effectiveness, and complications of CART applied to patients with malignancies. However, its mechanism reflecting these effects still remains unclear. We evaluated concentration of inflammatory cytokines including interleukin (IL)-1β, IL-6, IL-8, IL-12, tumor necrosis factor (TNF)-α, and immunosuppressive cytokine IL-10 in ascites before CART procedures. We investigated their impacts on survival. IL-1β, IL-6, IL-8, TNF-α, and IL-10 were detected in ascites of the patients undergoing CART. Significant body temperature elevation, one potential complication of CART, was observed among the patients although it was not clinically important. There were no significant correlations between changes in body temperature and the concentration of IL-6, IL-8, and IL-10. The presence of IL-10 in ascites significantly related to longer survival after the first session of CART procedures. However, we observed no other clinically important correlation between cytokine concentrations and changes in WBC and CRP. Concentration of inflammatory cytokines in ascites did not relate to body temperature change, the chief complication of CART. Surprisingly, the presence of IL-10 in ascites related to longer survival after CART. Immunological environment of cancer-related ascites may reflect the outcome of CART and improve survival in those with malignancy.
Collapse
Affiliation(s)
- Tetsuya Ito
- Department of Palliative Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Norio Hanafusa
- Department of Blood Purification, Tokyo Women's Medical University, Tokyo, Japan
| | - Satoru Iwase
- Department of Palliative Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Eisei Noiri
- Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan
| | - Masaomi Nangaku
- Department of Hemodialysis and Apheresis, The University of Tokyo Hospital, Tokyo, Japan
| | - Keiichi Nakagawa
- Department of Palliative Medicine, The University of Tokyo Hospital, Tokyo, Japan
| | - Kiyoshi Miyagawa
- Laboratory of Molecular Radiology, Center for Disease Biology and Integrative Medicine, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
15
|
Armbrust R, Neeb C, Thuss-Patience P, Lüftner D, Pietzner K, Riess H, Oskay-Öczelik G, Richter R, Keller M, Sehouli J. Patients' perspectives towards malignant ascites: results of a prospective observational trial regarding expectations, characteristics and quality of life-a study of the North-Eastern-German Society of Gynecological Oncology. Arch Gynecol Obstet 2019; 299:1385-1389. [PMID: 30834969 DOI: 10.1007/s00404-019-05071-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 01/28/2019] [Indexed: 11/29/2022]
Abstract
PURPOSE Malignant ascites (MA) is a frequent and common symptom in (gyneco-) oncological patients. The present trial evaluated and assessed patients' characteristics, clinical features and the possible influence of MA on QoL measurements. METHODS A prospective observational trial was conducted from Oct 2013 until Nov 2016. Therefore an interdisciplinary questionnaire was developed. Overall 250 patients with histological confirmed MA were included with different cancer entities (gynecological, gastrointestinal). The correlation of MA caused symptoms and QoL measurements was assessed using Kendall's tau b. Multivariable logistic regression models were applied to analyze the risks of symptoms or severe limitation in daily activities. RESULTS 125 questionnaires could be analyzed. The majority of patients with MA had diagnosis of ovarian cancer (68.8%) and were under current cancer treatment (57.6%), mostly chemotherapy. Over 50% reported abdominal tension as major symptom, around 56% of the patients had MA when cancer was firstly diagnosed. Regression analysis showed that patients with MA above 2l were significantly more likely to be harmed in everyday activities. However, the age, gender, type of malignancy and the current treatment (chemotherapy vs. no chemotherapy) had no significant influence. CONCLUSION MA has a significantly impact on QoL measurements in cancer patients and might influence everyday activities including basic needs like eating, walking and body care. There is a high need for more information and education of patients with MA.
Collapse
Affiliation(s)
- Robert Armbrust
- Department of Gynecology With Center for Oncological Surgery, Charité European Competence Center for Ovarian Cancer, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany. .,North-Eastern German Society of Gynecological Oncology (NOGGO), Augustenburger Platz 1, 13353, Berlin, Germany.
| | - C Neeb
- Department of Gynecology With Center for Oncological Surgery, Charité European Competence Center for Ovarian Cancer, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - P Thuss-Patience
- Department of Hematology, Oncology and Tumor Immunology, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - D Lüftner
- Department of Hematology, Oncology and Tumor Immunology, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - K Pietzner
- Department of Gynecology With Center for Oncological Surgery, Charité European Competence Center for Ovarian Cancer, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,North-Eastern German Society of Gynecological Oncology (NOGGO), Augustenburger Platz 1, 13353, Berlin, Germany
| | - H Riess
- Department of Hematology, Oncology and Tumor Immunology, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - G Oskay-Öczelik
- Department of Gynecology With Center for Oncological Surgery, Charité European Competence Center for Ovarian Cancer, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,North-Eastern German Society of Gynecological Oncology (NOGGO), Augustenburger Platz 1, 13353, Berlin, Germany
| | - R Richter
- Department of Gynecology With Center for Oncological Surgery, Charité European Competence Center for Ovarian Cancer, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - M Keller
- North-Eastern German Society of Gynecological Oncology (NOGGO), Augustenburger Platz 1, 13353, Berlin, Germany
| | - J Sehouli
- Department of Gynecology With Center for Oncological Surgery, Charité European Competence Center for Ovarian Cancer, University Hospital Charité Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,North-Eastern German Society of Gynecological Oncology (NOGGO), Augustenburger Platz 1, 13353, Berlin, Germany
| |
Collapse
|
16
|
Nakano M, Ito M, Tanaka R, Ariyama H, Mitsugi K, Makiyama A, Uchino K, Esaki T, Tsuruta N, Hanamura F, Yamaguchi K, Okumura Y, Sagara K, Takayoshi K, Nio K, Tsuchihashi K, Tamura S, Shimokawa H, Arita S, Miyawaki K, Kusaba H, Akashi K, Baba E. Epithelial-mesenchymal transition is activated in CD44-positive malignant ascites tumor cells of gastrointestinal cancer. Cancer Sci 2018; 109:3461-3470. [PMID: 30142697 PMCID: PMC6215886 DOI: 10.1111/cas.13777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 08/20/2018] [Accepted: 08/22/2018] [Indexed: 12/18/2022] Open
Abstract
Disseminated cancer cells in malignant ascites possess unique properties that differ from primary tumors. However, the biological features of ascites tumor cells (ATC) have not been fully investigated. By analyzing ascites fluid from 65 gastrointestinal cancer patients, the distinguishing characteristics of ATC were identified. High frequency of CD44+ cells was observed in ATC using flow cytometry (n = 48). Multiplex quantitative PCR (n = 15) showed higher gene expression of epithelial‐mesenchymal transition (EMT)‐related genes and transforming growth factor beta (TGF‐beta)‐related genes in ATC than in the primary tissues. Immunohistochemistry (n = 10) showed that ATC also had much higher expression of phosphorylated SMAD2 than that in the corresponding primary tissues. TGF‐beta 1 was detected in all cases of malignant ascites by enzyme‐linked immunoassay (n = 38), suggesting the possible interaction of ATC and the ascites microenvironment. In vitro experiments revealed that these ATC properties were maintained by TGF‐beta 1 in cultured ATC(n = 3). Here, we showed that ATCrevealed high frequencies of CD44 and possessed distinct EMT features from primary tissues that were mainly maintained by TGF‐beta 1 in the ascites.
Collapse
Affiliation(s)
- Michitaka Nakano
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Mamoru Ito
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Risa Tanaka
- Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan
| | - Hiroshi Ariyama
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kenji Mitsugi
- Department of Medical Oncology, Hamanomachi Hospital, Fukuoka, Japan
| | - Akitaka Makiyama
- Department of Hematology/Oncology, Japan Community Healthcare Organization Kyushu Hospital, Kitakyushu, Japan
| | - Keita Uchino
- Department of Medical Oncology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Taito Esaki
- Department of Gastrointestinal and Medical Oncology, National Kyushu Cancer Center, Fukuoka, Japan
| | - Nobuhiro Tsuruta
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Fumiyasu Hanamura
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kyoko Yamaguchi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yuta Okumura
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kosuke Sagara
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kotoe Takayoshi
- Department of Gastrointestinal and Medical Oncology, National Kyushu Cancer Center, Fukuoka, Japan
| | - Kenta Nio
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kenji Tsuchihashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Shingo Tamura
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hozumi Shimokawa
- Department of Medical Oncology, Clinical Research Institute, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | - Shuji Arita
- Department of Comprehensive Clinical Oncology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Kohta Miyawaki
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Hitoshi Kusaba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Eishi Baba
- Department of Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.,Department of Comprehensive Clinical Oncology, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
17
|
Klymenko Y, Wates RB, Weiss-Bilka H, Lombard R, Liu Y, Campbell L, Kim O, Wagner D, Ravosa MJ, Stack MS. Modeling the effect of ascites-induced compression on ovarian cancer multicellular aggregates. Dis Model Mech 2018; 11:dmm034199. [PMID: 30254133 PMCID: PMC6176988 DOI: 10.1242/dmm.034199] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/29/2018] [Indexed: 12/12/2022] Open
Abstract
Epithelial ovarian cancer (EOC) is the most lethal gynecological malignancy. EOC dissemination is predominantly via direct extension of cells and multicellular aggregates (MCAs) into the peritoneal cavity, which adhere to and induce retraction of peritoneal mesothelium and proliferate in the submesothelial matrix to generate metastatic lesions. Metastasis is facilitated by the accumulation of malignant ascites (500 ml to >2 l), resulting in physical discomfort and abdominal distension, and leading to poor prognosis. Although intraperitoneal fluid pressure is normally subatmospheric, an average intraperitoneal pressure of 30 cmH2O (22.1 mmHg) has been reported in women with EOC. In this study, to enable experimental evaluation of the impact of high intraperitoneal pressure on EOC progression, two new in vitro model systems were developed. Initial experiments evaluated EOC MCAs in pressure vessels connected to an Instron to apply short-term compressive force. A Flexcell Compression Plus system was then used to enable longer-term compression of MCAs in custom-designed hydrogel carriers. Results show changes in the expression of genes related to epithelial-mesenchymal transition as well as altered dispersal of compressed MCAs on collagen gels. These new model systems have utility for future analyses of compression-induced mechanotransduction and the resulting impact on cellular responses related to intraperitoneal metastatic dissemination.This article has an associated First Person interview with the first authors of the paper.
Collapse
Affiliation(s)
- Yuliya Klymenko
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Rebecca B Wates
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Holly Weiss-Bilka
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Rachel Lombard
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Yueying Liu
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Leigh Campbell
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - Oleg Kim
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
- Department of Applied and Computational Mathematics and Statistics, University of Notre Dame, Notre Dame, IN 46556, USA
- Department of Mathematics, University of California, Riverside, CA 92521, USA
| | - Diane Wagner
- Department of Mechanical and Energy Engineering, Indiana University-Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Matthew J Ravosa
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
| | - M Sharon Stack
- Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN 46556, USA
| |
Collapse
|
18
|
Nakano M, Ito M, Tanaka R, Yamaguchi K, Ariyama H, Mitsugi K, Yoshihiro T, Ohmura H, Tsuruta N, Hanamura F, Sagara K, Okumura Y, Nio K, Tsuchihashi K, Arita S, Kusaba H, Akashi K, Baba E. PD-1+ TIM-3+ T cells in malignant ascites predict prognosis of gastrointestinal cancer. Cancer Sci 2018; 109:2986-2992. [PMID: 30187676 PMCID: PMC6125472 DOI: 10.1111/cas.13723] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 06/30/2018] [Indexed: 01/27/2023] Open
Abstract
The liquid biopsy of ascites fluid could be an excellent source of tumor and microenvironment for the study of prognostic biomarkers because of its accessibility. Tumor-infiltrating lymphocytes (TILs) can predict prognosis in multiple malignancies, including the response to immune checkpoint inhibitors, a breakthrough cancer therapy. However, TILs' profiles from malignant ascites have not been extensively studied. Using flow cytometric analysis, we quantified the proportion of exhausted T cells and memory/naive/effector T-cell subsets, among the CD4+ and CD8+ T-cell populations of paired TILs and peripheral blood T cell samples (n = 22). The correlation between CD4+ and CD8+ subset profiles suggested that the combined analysis of CD4+ and CD8+ cells in malignant ascites was clinically significant. We found that cells positive for the exhaustion markers programmed cell death-1 (PD-1), and T-cell immunoglobulin and mucin domain 3 (TIM-3), and cells coexpressing PD-1 and TIM-3 abundantly exist among malignant ascites TILs. Furthermore, patients with high frequency of PD-1+ TIM-3+ cells among the CD4+ and CD8+ T-cell population showed worse clinical outcome in multivariate analysis (n = 27). We propose that exhausted ascites TILs represent a clinically significant prognostic biomarker in advanced gastrointestinal cancer and represent an important target for immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Michitaka Nakano
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Mamoru Ito
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Risa Tanaka
- Department of Medical OncologyHamanomachi HospitalFukuokaJapan
| | - Kyoko Yamaguchi
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Hiroshi Ariyama
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Kenji Mitsugi
- Department of Medical OncologyHamanomachi HospitalFukuokaJapan
| | - Tomoyasu Yoshihiro
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Hirofumi Ohmura
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Nobuhiro Tsuruta
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Fumiyasu Hanamura
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Kosuke Sagara
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Yuta Okumura
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Kenta Nio
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Kenji Tsuchihashi
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Shuji Arita
- Faculty of Medical SciencesDepartment of Comprehensive Clinical OncologyKyushu UniversityFukuokaJapan
| | - Hitoshi Kusaba
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Koichi Akashi
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
| | - Eishi Baba
- Department of Medicine and Biosystemic ScienceKyushu University Graduate School of Medical SciencesFukuokaJapan
- Faculty of Medical SciencesDepartment of Comprehensive Clinical OncologyKyushu UniversityFukuokaJapan
| |
Collapse
|
19
|
Korpi S, Salminen VV, Piili RP, Paunu N, Luukkaala T, Lehto JT. Therapeutic Procedures for Malignant Ascites in a Palliative Care Outpatient Clinic. J Palliat Med 2018; 21:836-841. [DOI: 10.1089/jpm.2017.0616] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Säde Korpi
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Veera V. Salminen
- Department of Oncology, Palliative Care Unit, Tampere University Hospital, Tampere, Finland
| | - Reetta P. Piili
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Department of Oncology, Palliative Care Unit, Tampere University Hospital, Tampere, Finland
| | - Niina Paunu
- Department of Oncology, Palliative Care Unit, Tampere University Hospital, Tampere, Finland
| | - Tiina Luukkaala
- Research and Innovation Center, Tampere University Hospital, Tampere, Finland
- Faculty of Social Sciences, University of Tampere, Tampere, Finland
| | - Juho T. Lehto
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
- Department of Oncology, Palliative Care Unit, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
20
|
Ha T, Madoff DC, Li D. Symptomatic Fluid Drainage: Tunneled Peritoneal and Pleural Catheters. Semin Intervent Radiol 2017; 34:337-342. [PMID: 29249857 DOI: 10.1055/s-0037-1608704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Malignant ascites and pleural effusion are significant contributors to patient symptoms such as shortness of breath, abdominal distension, and nausea in the setting of cancer. There are numerous methods employed to control such symptoms such as serial drainages, pleurodesis, and tunneled drainage catheters. Tunneled drainage catheters are a safe, effective, and a minimally invasive procedure to palliate the symptoms of malignant ascites and pleural effusion.
Collapse
Affiliation(s)
- Tony Ha
- Division of Interventional Radiology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - David C Madoff
- Department of Radiology, Division of Interventional Radiology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| | - David Li
- Department of Radiology, Division of Interventional Radiology, New York Presbyterian Hospital/Weill Cornell Medical Center, New York, New York
| |
Collapse
|
21
|
|
22
|
Loss of PRP4K drives anoikis resistance in part by dysregulation of epidermal growth factor receptor endosomal trafficking. Oncogene 2017; 37:174-184. [PMID: 28892043 PMCID: PMC5770602 DOI: 10.1038/onc.2017.318] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2016] [Revised: 07/06/2017] [Accepted: 08/04/2017] [Indexed: 12/16/2022]
Abstract
Anoikis acts as a critical barrier to metastasis by inducing cell death upon cancer cell detachment from the extracellular matrix (ECM), thereby preventing tumor cell dissemination to secondary sites. The induction of anoikis requires the lysosomal-mediated downregulation of epidermal growth factor receptors (EGFRs) leading to termination of pro-survival signaling. In this study, we demonstrate that depletion of pre-mRNA splicing factor 4 kinase (PRP4K; also known as PRPF4B) causes dysregulation of EGFR trafficking and anoikis resistance. We also report a novel cytoplasmic localization of PRP4K at the late endosome, and demonstrate both nuclear and cytoplasmic localization in breast, lung and ovarian cancer tissue. Mechanistically, depletion of PRP4K leads to reduced EGFR degradation following cell detachment from the ECM and correlates with increased TrkB, vimentin and Zeb1 expression. As a result, PRP4K loss promotes sustained growth factor signaling and increased cellular resistance to anoikis in vitro and in a novel zebrafish xenotransplantation model of anoikis sensitivity, as well as increased metastasis in a mouse model of ovarian cancer. Thus, PRP4K may serve as a potential biomarker of anoikis sensitivity in ovarian and other epithelial cancers.
Collapse
|
23
|
Klymenko Y, Kim O, Stack MS. Complex Determinants of Epithelial: Mesenchymal Phenotypic Plasticity in Ovarian Cancer. Cancers (Basel) 2017; 9:cancers9080104. [PMID: 28792442 PMCID: PMC5575607 DOI: 10.3390/cancers9080104] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 08/02/2017] [Accepted: 08/06/2017] [Indexed: 02/07/2023] Open
Abstract
Unlike most epithelial malignancies which metastasize hematogenously, metastasis of epithelial ovarian cancer (EOC) occurs primarily via transcoelomic dissemination, characterized by exfoliation of cells from the primary tumor, avoidance of detachment-induced cell death (anoikis), movement throughout the peritoneal cavity as individual cells and multi-cellular aggregates (MCAs), adhesion to and disruption of the mesothelial lining of the peritoneum, and submesothelial matrix anchoring and proliferation to generate widely disseminated metastases. This exceptional microenvironment is highly permissive for phenotypic plasticity, enabling mesenchymal-to-epithelial (MET) and epithelial-to-mesenchymal (EMT) transitions. In this review, we summarize current knowledge on EOC heterogeneity in an EMT context, outline major regulators of EMT in ovarian cancer, address controversies in EMT and EOC chemoresistance, and highlight computational modeling approaches toward understanding EMT/MET in EOC.
Collapse
Affiliation(s)
- Yuliya Klymenko
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA.
- Medical Sciences Program, Indiana University School of Medicine, Bloomington, IN 47405, USA.
| | - Oleg Kim
- Department of Applied and Computational Mathematics and Statistics, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA.
- Department of Mathematics, University of California Riverside, Riverside, CA 92521, USA.
| | - M Sharon Stack
- Department of Chemistry and Biochemistry, Harper Cancer Research Institute, University of Notre Dame, Notre Dame, IN 46617, USA.
| |
Collapse
|
24
|
Abstract
Malignant ascites (MA) is a sign of advanced cancer and poor prognosis. MA can result in impairment in quality of life (QOL) and significant symptoms. As a supportive treatment, ascites can be drained by paracentesis (PC), percutaneously implanted catheters (tunneled, untunneled, central venous catheters), or peritoneal ports, or peritoneovenous shunts. The aim of this study was to evaluate the effectiveness, safety, and patient-reported outcomes (PRO) of different drainage methods for the management of MA. A systematic review of the literature was performed, and 32 original articles met the inclusion criteria. Patients selected for permanent drain insertion demonstrated symptoms related to MA and had undergone repeated PC. The primary focus of the reviewed articles was procedural safety issues. The rate of technical success of drainage device installation was 100%. Most patients experienced improvements in symptom control after ascites drainage. When analyzed together, 19.7% (255/1297) of patients experienced any complication and 6.2% (81/1297) experienced serious adverse events during MA drainage. Complications were reported for every drainage method; however, the least occurred after PC or central venous catheter, while the most serious occurred after peritoneovenous shunts. Adverse events were as follows: catheter obstruction: 4.4%, infection: 4.1%, leakage: 3.5%, catheter dislodgment: 2.3%, hypotension: 0.6%, injuries during device insertion: 0.6%, renal impairment: 0.5%, electrolyte imbalance: 0.2%, other: 3.6%. PRO and QOL endpoints were available for 12 studies. When PRO were measured using an interview, a significant improvement in symptom control and QOL was reported in almost all patients. Once standardized questionnaires were used, improvements in symptomatic scores and role functioning were observed. Deterioration was observed in cognitive and emotional subscales. MA drainage is a safe and effective method to control symptoms associated with ascites, and should be perceived as a supportive care, that can be applied for those who need it at any time of their cancer trajectory. Patient selection should be performed using a thorough assessment of symptoms and QOL, and should not be delayed.
Collapse
Affiliation(s)
- Maciej Stukan
- Department of Gynecologic Oncology, Gdynia Oncology Center, Szpitale Wojewodzkie w Gdyni Sp. z o.o., Gdynia, Poland
| |
Collapse
|
25
|
Kim S, Gwak H, Kim HS, Kim B, Dhanasekaran DN, Song YS. Malignant ascites enhances migratory and invasive properties of ovarian cancer cells with membrane bound IL-6R in vitro. Oncotarget 2016; 7:83148-83159. [PMID: 27825119 PMCID: PMC5347759 DOI: 10.18632/oncotarget.13074] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 10/11/2016] [Indexed: 12/16/2022] Open
Abstract
Transcoelomic route is the most common and the earliest route of metastasis, causing the ascites formation in advanced epithelial ovarian cancer (EOC). We demonstrated that interleukin 6 (IL-6) is enriched in the malignant ascites from patients with ovarian cancer, which enhanced invasive properties of EOC cells. Interestingly, the expression of IL-6R on cell membrane of EOC cells correlated with ascites-induced invasion. Selective knockdown of IL-6R or inhibition with IL-6 neutralizing antibody, suppressed the stimulatory effects of ascites on EOC invasion. Moreover, the ascites treatment induced the phosphorylation of JAK2-STAT3 and use of selective inhibitors of JAK2 and STAT3, blocked the expression of epithelial-mesenchymal transition related proteins in parallel with the suppression of EOC invasion. Thus, IL-6/IL-6R mediated JAK2-STAT3 signaling pathway could be a promising therapeutic target for anticancer therapy in ovarian cancer patients with ascites.
Collapse
MESH Headings
- Aged
- Antibodies, Neutralizing/pharmacology
- Antineoplastic Agents/pharmacology
- Ascites
- Ascitic Fluid/metabolism
- Carcinoma, Ovarian Epithelial
- Cell Line, Tumor
- Cell Membrane/drug effects
- Cell Membrane/metabolism
- Cell Movement/drug effects
- Epithelial-Mesenchymal Transition
- Female
- Humans
- Interleukin-6/metabolism
- Janus Kinase 2/antagonists & inhibitors
- Janus Kinase 2/metabolism
- Middle Aged
- Neoplasm Invasiveness
- Neoplasms, Glandular and Epithelial/drug therapy
- Neoplasms, Glandular and Epithelial/genetics
- Neoplasms, Glandular and Epithelial/metabolism
- Neoplasms, Glandular and Epithelial/pathology
- Ovarian Neoplasms/drug therapy
- Ovarian Neoplasms/genetics
- Ovarian Neoplasms/metabolism
- Ovarian Neoplasms/pathology
- Phosphorylation
- Protein Kinase Inhibitors/pharmacology
- RNA Interference
- Receptors, Interleukin-6/antagonists & inhibitors
- Receptors, Interleukin-6/genetics
- Receptors, Interleukin-6/metabolism
- STAT3 Transcription Factor/antagonists & inhibitors
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Soochi Kim
- Interdisciplinary Program in Cancer Biology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
| | - HyeRan Gwak
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
| | - Hee Seung Kim
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, Republic of Korea
| | - Boyun Kim
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Nano System Institute, Seoul National University, Seoul, Korea
| | | | - Yong Sang Song
- Interdisciplinary Program in Cancer Biology, College of Medicine, Seoul National University, Seoul, Republic of Korea
- Cancer Research Institute, Seoul National University, Seoul, Republic of Korea
- Biomodulation, Department of Agricultural Biotechnology, Seoul National University, Seoul, Republic of Korea
- Department of Obstetrics and Gynecology, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
26
|
Hicks AM, Chou J, Capanu M, Lowery MA, Yu KH, O'Reilly EM. Pancreas Adenocarcinoma: Ascites, Clinical Manifestations, and Management Implications. Clin Colorectal Cancer 2016; 15:360-368. [PMID: 27262896 PMCID: PMC5099112 DOI: 10.1016/j.clcc.2016.04.014] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2015] [Revised: 03/28/2016] [Accepted: 04/27/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND Ascites develops in a subset of patients with pancreatic adenocarcinoma (PAC) at presentation or as the disease advances. Limited data exist on the prognostic importance of malignant ascites in PAC. Our hypothesis is that this information will provide an understanding of the natural history and facilitate management decisions. METHODS We conducted a retrospective analysis of 180 patients treated at Memorial Sloan Kettering Cancer Center diagnosed between January 1, 2009 and December 31, 2014, with PAC and with ascites either at presentation or that developed during the disease course. RESULTS For the 180 patients, the overall survival was 15 months. The time from diagnosis to ascites presentation was 11 months, and the survival time after ascites development was 1.8 months (range, 1.6-2.3 months; 95% confidence interval). Of 62 patients (34%) who had ascitic fluid analyzed, 36 (58%) had positive cytology. Fifty-one (82%) patients had a serum ascites albumin gradient ≥ 1, and 11 (18%) had serum ascites albumin gradient < 1. Sixty-four (36%) patients had their ascites managed solely by serial paracenteses. A total of 116 patients required a catheter; of these, 108 (93%) had a Tenckhoff catheter, 4 (3%) a Pleurx catheter, 4 (3%) a pigtail catheter, and 1 (1%) a Denver catheter. Eight (7%) patients required 2 catheters to be placed, and in 6 (5%), Tenckhoff catheters had to be removed. The main observed complications were spontaneous bacterial peritonitis in 7 (11%) managed with paracenteses versus 26 (23%) who had a catheter placed, catheter malfunction in 8 (7%), and acute renal failure in 6 (3%). After ascites development, 79 (44%) patients received active anti-cancer therapy, and 101 (56%) patients were managed with supportive care alone. CONCLUSIONS In patients with PAC who presented with or developed ascites, serial paracenteses and indwelling catheters are common methods used for providing symptomatic relief. The complication rate was higher with indwelling catheters, primarily related to infection (eg, bacterial peritonitis). Overall, ascites has a significantly negative prognostic import with a short median survival.
Collapse
Affiliation(s)
- Angel Mier Hicks
- Department of Medicine, Icahn School of Medicine at Mount Sinai/St. Luke's Roosevelt Hospital Center Program, New York, NY
| | - Joanne Chou
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Marinela Capanu
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maeve A Lowery
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kenneth H Yu
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eileen M O'Reilly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY.
| |
Collapse
|
27
|
Pilanc KN, Ordu Ç, Akpnar H, Balc C, Başsülü N, Köksal Üİ, Elbüken F, Okutur K, Bülbül G, Sağlam S, Demir G. Dramatic Response to Catumaxomab Treatment for Malign Ascites Related to Renal Cell Carcinoma With Sarcomotoid Differentiation. Am J Ther 2016; 23:e1078-e1081. [PMID: 24732906 DOI: 10.1097/mjt.0000000000000064] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Refractory malignant ascites (MA) is a common complication in cancer patients. Renal cell carcinoma (RCC) is rarely present with peritoneal ascites, which is commonly associated with carcinomas of the gastrointestinal and female reproductive tracts; including especially ovarian high-grade serous carcinoma. Currently, chemotherapy and paracentesis represent the most widely used methods to relieve the symptoms. Recently, intraperitoneal therapy with catumaxomab-a trifunctional hybrid antibody-has been introduced for the treatment of MA. The benefit of this treatment has been demonstrated in patients with distinct abdominal malignancies. In this case report, we present the first case of successful catumaxomab treatment against MA in a patient with advanced RCC with sarcomatoid differentiation. After the second administration of catumaxomab, paracentesis became no longer necessary. Catumaxomab might represent a safe treatment option for MA in the course of metastatic RCC with sarcomatoid differentiation.
Collapse
Affiliation(s)
- Kezban Nur Pilanc
- Departments of 1Medical Oncology, 2Urology, 3Radiology, and 4Pathology, Faculty of Medicine, Bilim University; and 5Department of Radiology, Gayrettepe Florence Nightingale Hospital, Instanbul, Turkey
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Meyer L, Suidan R, Sun C, Westin S, Coleman RL, Mills GB. The management of malignant ascites and impact on quality of life outcomes in women with ovarian cancer. EXPERT REVIEW OF QUALITY OF LIFE IN CANCER CARE 2016; 1:231-238. [PMID: 30906877 PMCID: PMC6425954 DOI: 10.1080/23809000.2016.1185369] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Malignant ascites is one of the most common sequela of epithelial ovarian cancer. It causes significant symptoms and can have a detrimental impact on patient quality of life, especially in women with recurrent ovarian cancer. The management of symptomatic ascites consists of both mechanical treatments that aim to drain the peritoneal cavity, and medical therapies that prevent and diminish the development of ascites. Mechanical options include serial paracentesis, peritoneal catheters, and peritoneovenous shunts. Pharmaceutical treatments include diuretics, angiogenesis inhibitors, and other targeted agents. There is a perception, without formal analysis, that intractable ascites is less common in the taxane era of therapy. In this review paper, we highlight current and emerging therapeutic strategies, complications and contraindications, and their effects on patient quality of life.
Collapse
Affiliation(s)
- Larissa Meyer
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Rudy Suidan
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Charlotte Sun
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Shannon Westin
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Robert L Coleman
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| | - Gordon B Mills
- The University of Texas MD Anderson Cancer Center, Houston Texas, United States
| |
Collapse
|
29
|
Gu X, Zhang Y, Cheng M, Liu M, Zhang Z, Cheng W. Management of non-ovarian cancer malignant ascites through indwelling catheter drainage. BMC Palliat Care 2016; 15:44. [PMID: 27103467 PMCID: PMC4839130 DOI: 10.1186/s12904-016-0116-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 04/13/2016] [Indexed: 02/07/2023] Open
Abstract
Backgrounds Intra-abdominal placement of the Central Venous Catheter (CVC) was conducted to manage the ascites-related symptoms of non-ovarian cancer patients. The aim of this study is to document the efficacy of symptom relief and conduct survival analysis of non-ovarian cancer patients with malignant ascites who received paracentesis and indwelling catheter drainage. Methods Seventy eight patients received paracentesis and drainage. All patients who met the inclusion criteria were included in this study. The overall survival (OS) was defined as the interval between initial diagnosis and death. Since-paracentesis survival (SP-Survival) was defined as the interval between initial paracentesis and death. Results Hepatic cancer was the most frequent original cancer in this study. Peritoneal catheters remained in situ for a median of 13 days. No immediate complications, such as perforation of a viscus or excessive bleeding, were encountered during placement. All ascites-related symptoms improved after drainage compared with the baseline. There was a statistically significant improvement in the mean score for abdominal swelling (p < 0.001), anorexia (p = 0.023) and constipation (p = 0.045). Cancer type was shown to be an independent prognostic factor for overall survival length (p = 0.001). Serum albumin was an independent prognostic factor for SP-survival (p = 0.02). Conclusions Paracentesis and indwelling catheter drainage through CVC set is a useful method for improving painful symptom. Further research is needed to validate the findings.
Collapse
Affiliation(s)
- Xiaoli Gu
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yuanyuan Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Menglei Cheng
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Minghui Liu
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Zhe Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Wenwu Cheng
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China. .,, #270, DongAn Road, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
30
|
Jung M, Pützer S, Gevensleben H, Meller S, Kristiansen G, Dietrich D. Diagnostic and prognostic value of SHOX2 and SEPT9 DNA methylation and cytology in benign, paramalignant, and malignant ascites. Clin Epigenetics 2016; 8:24. [PMID: 26937257 PMCID: PMC4774089 DOI: 10.1186/s13148-016-0192-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/25/2016] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cytology remains the gold standard for the detection of malignant cells in ascites. However, its sensitivity is limited. The aim of this study was to evaluate DNA methylation biomarkers for the differential diagnosis of benign (ascites in patients without malignancy), malignant (ascites in cancer patients directly caused by malignancy), and paramalignant (ascites in cancer patients caused by comorbidities but not by malignancy) ascites. METHODS A cohort of 283 patients (134 cancer patients, 149 patients with benign diseases) presenting with ascites was prospectively enrolled. Ascites was evaluated by means of cytopathological investigation and DNA methylation of SHOX2 and SEPT9 in the cell-free and cellular fraction. DNA methylation in bisulfite-converted DNA was determined using quantitative methylation specific real-time PCR. Cytopathological and DNA methylation results were evaluated with regard to diagnosis and overall survival (OS). RESULTS Patients with positive DNA methylation had a poor overall survival compared to methylation-negative patients (hazard ratio: HR = 1.97, p = 0.001). In multivariate survival analysis, DNA methylation was an independent prognostic parameter (p = 0.003) together with age (HR = 1.03, p < 0.001) and the presence of malignant disease (HR = 1.87, p < 0.001). The combination of methylation with cytopathological analyses led to a 42 % increase in the detection rate of malignant ascites, resulting in 37 % positively diagnosed cancer patients and a specificity of 97 %. Among cancer patients, patients with DNA methylation-positive ascites showed an adverse clinical course (HR = 1.63, p = 0.039). CONCLUSIONS DNA methylation testing adds diagnostic and prognostic information and might constitute an effective ancillary method for the differential diagnosis of malignant, paramalignant, and benign ascites.
Collapse
Affiliation(s)
- Maria Jung
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Svenja Pützer
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Heidrun Gevensleben
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Sebastian Meller
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Glen Kristiansen
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany
| | - Dimo Dietrich
- Institute of Pathology, University Hospital Bonn, Sigmund-Freud-Str. 25, 53127 Bonn, Germany.,Department of Otolaryngology, Head and Neck Surgery, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
31
|
Qu C, Xing M, Ghodadra A, McCluskey KM, Santos E, Kim HS. The Impact of Tunneled Catheters for Ascites and Peritoneal Carcinomatosis on Patient Rehospitalizations. Cardiovasc Intervent Radiol 2015; 39:711-716. [PMID: 26662561 DOI: 10.1007/s00270-015-1258-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 11/01/2015] [Indexed: 01/15/2023]
Abstract
PURPOSE The aim of the study is to assess patient outcomes, complications, impact on rehospitalizations, and healthcare costs in patients with malignant ascites treated with tunneled catheters. MATERIALS AND METHODS A total of 84 patients with malignant ascites (mean age, 60 years) were treated with tunneled catheters. Patients with peritoneal carcinomatosis and malignant ascites treated with tunneled drain catheter placement over a 3-year period were studied. Overall survival from the time of ascites and catheter placement were stratified by primary cancer and analyzed using the Kaplan-Meier method. Complications were graded by the Common Terminology Criteria for Adverse Events v3.0 (CTCAE). The differences between pre- and post-catheter admissions, hospitalizations, and Emergency Department (ED) visits, as well as related inpatient expenses were compared using paired t tests. RESULTS There were no significant differences in gender, age, or race between different primary cancer subgroups. One patient (1%) developed bleeding (CTCAE-2). Four patients (5%) developed local cellulitis (CTCAE-2). Three patients (4%) had prolonged hospital stay (between 7 and 10 days) to manage ascites-related complications such as abdominal distention, discomfort, or pain. Comparison between pre- and post-catheter hospitalizations showed significantly lower admissions (-1.4/month, p < 0.001), hospital stays (-4.2/month, p = 0.003), and ED visits (-0.9/month, p = 0.002). The pre- and post-catheter treatment health care cost was estimated using MS-DRG IPPS payment system and it demonstrated significant cost savings from decreased inpatient admissions in post-treatment period (-$9535/month, p < 0.001). CONCLUSIONS Tunneled catheter treatment of malignant ascites is safe, feasible, well tolerated, and cost effective. Tunneled catheter treatment may play an important role in improving patients' quality of life and outcomes while controlling health care expenditures.
Collapse
Affiliation(s)
- Chuanxing Qu
- Division of Interventional Radiology, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Minzhi Xing
- Division of Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale Cancer Center, Yale University School of Medicine, 330 Cedar Street, TE 2-224, New Haven, CT, 06510, USA
| | - Anish Ghodadra
- Division of Interventional Radiology, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Kevin M McCluskey
- Division of Interventional Radiology, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ernesto Santos
- Division of Interventional Radiology, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hyun S Kim
- Division of Interventional Radiology, Department of Radiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Division of Interventional Radiology, Department of Radiology and Biomedical Imaging, Yale Cancer Center, Yale University School of Medicine, 330 Cedar Street, TE 2-224, New Haven, CT, 06510, USA.
- Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
32
|
The Mesothelial Origin of Carcinoma Associated-Fibroblasts in Peritoneal Metastasis. Cancers (Basel) 2015; 7:1994-2011. [PMID: 26426054 PMCID: PMC4695872 DOI: 10.3390/cancers7040872] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 09/14/2015] [Accepted: 09/23/2015] [Indexed: 01/15/2023] Open
Abstract
Solid tumors are complex and unstructured organs that, in addition to cancer cells, also contain other cell types. Carcinoma-associated fibroblasts (CAFs) represent an important population in the tumor microenviroment and participate in several stages of tumor progression, including cancer cell migration/invasion and metastasis. During peritoneal metastasis, cancer cells detach from the primary tumor, such as ovarian or gastrointestinal, disseminate through the peritoneal fluid and colonize the peritoneum. Tumor cells metastasize by attaching to and invading through the mesothelial cell (MC) monolayer that lines the peritoneal cavity, then colonizing the submesothelial compact zone where CAFs accumulate. CAFs may derive from different sources depending on the surrounding metastatic niche. In peritoneal metastasis, a sizeable subpopulation of CAFs originates from MCs through a mesothelial-to-mesenchymal transition (MMT), which promotes adhesion, invasion, vascularization and subsequent tumor growth. The bidirectional communication between cancer cells and MC-derived CAFs via secretion of a wide range of cytokines, growth factors and extracellular matrix components seems to be crucial for the establishment and progression of the metastasis in the peritoneum. This manuscript provides a comprehensive review of novel advances in understanding how peritoneal CAFs provide cancer cells with a supportive microenvironment, as well as the development of future therapeutic approaches by interfering with the MMT in the peritoneum.
Collapse
|
33
|
Wang L, Okubo T, Shinsaka M, Kobayashi A, Ogasawara M, Sakaguchi R, Nagai T, Seki H. Efficacy and safety of cell-free and concentrated ascites reinfusion therapy (CART) in gynecologic cancer patients with a large volume of ascites. J Obstet Gynaecol Res 2015; 41:1614-20. [PMID: 26177394 DOI: 10.1111/jog.12763] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 04/09/2015] [Accepted: 04/23/2015] [Indexed: 12/25/2022]
Abstract
AIM The aim of this study was to evaluate the efficacy and safety of cell-free concentrated ascites reinfusion therapy (CART) on a large amount of ascites. MATERIAL AND METHODS Fifty-eight CART procedures were performed in nine patients with ovarian, endometrial, or cervical cancer from February 2013 to September 2014. The medical records were retrospectively reviewed for the amount of collected ascites, vital signs, and laboratory results before and after CART. RESULTS No obvious change in the plasma protein and plasma albumin concentration was found after CART for < 5 L of ascites; however, obvious increases in both were observed in CART for ≥ 5 L of ascites (P < 0.001). The optimum cut-off value for obtaining a positive variant of plasma protein and plasma albumin after CART was 7.9 L. CART for ≥ 5 L of ascites did not increase the risk of transient water retention in the body (odds ratio = 2.2; 95% confidence interval: 0.35-13.83; P = 0.38); however, CART for ≥ 7.9 L of ascites increased the risk of water retention (odds ratio = 8.4; 95% confidence interval: 1.91-44.09; P = 0.004). The optimal cut-off value of ascites for predicting water retention due to CART was 9.2 L. CONCLUSION Massive ascites collection in CART < 9.2 L appears to be a safe and effective treatment for improving general condition, plasma protein, and electrolytes in gynecologic cancer patients.
Collapse
Affiliation(s)
- Liangcheng Wang
- Department of Obstetrics and Gynecology, Sekishindo Hospital, Saitama, Japan.,Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Takashi Okubo
- Department of Obstetrics and Gynecology, Sekishindo Hospital, Saitama, Japan
| | - Mamiko Shinsaka
- Department of Obstetrics and Gynecology, Sekishindo Hospital, Saitama, Japan
| | - Akiko Kobayashi
- Department of Obstetrics and Gynecology, Sekishindo Hospital, Saitama, Japan
| | - Miwa Ogasawara
- Department of Obstetrics and Gynecology, Sekishindo Hospital, Saitama, Japan
| | - Riko Sakaguchi
- Department of Obstetrics and Gynecology, Sekishindo Hospital, Saitama, Japan
| | - Tomonori Nagai
- Department of Obstetrics and Gynecology, Sekishindo Hospital, Saitama, Japan.,Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Hiroyuki Seki
- Department of Obstetrics and Gynecology, Sekishindo Hospital, Saitama, Japan.,Department of Obstetrics and Gynecology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| |
Collapse
|
34
|
Jang M, Yew PY, Hasegawa K, Ikeda Y, Fujiwara K, Fleming GF, Nakamura Y, Park JH. Characterization of T cell repertoire of blood, tumor, and ascites in ovarian cancer patients using next generation sequencing. Oncoimmunology 2015; 4:e1030561. [PMID: 26451311 PMCID: PMC4589054 DOI: 10.1080/2162402x.2015.1030561] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/10/2015] [Accepted: 03/13/2015] [Indexed: 12/31/2022] Open
Abstract
Tumor-infiltrating lymphocytes (TILs) play an important role in regulating the host immune response and are one of key factors in defining tumor microenvironment. Some studies have indicated that T cell infiltration in malignant ascites is associated with clinical outcome, but few studies have performed detailed characterization of T cell diversity or clonality in malignant effusions. We have applied a next generation sequencing method to characterize T cell repertoire of a set of primary cancers, ascites, and blood from 12 ovarian cancer patients and also analyzed the T cell subtype populations in malignant fluids from 3 ovarian cancer patients. We observed enrichment of certain T cells in tumors and ascites, but most of the enriched T cell receptor (TCR) sequences in tumors and ascites were not common. Moreover, we analyzed TCR sequences of T cell subtypes (CD4+, CD8+, and regulatory T cells) isolated from malignant effusions and also found clonal expansion of certain T cell populations, but the TCR sequences were almost mutually exclusive among the three subgroups. Although functional studies of clonally expanded T cell populations are definitely required, our approach offers a detailed characterization of T cell immune microenvironment in tumors and ascites that might differently affect antitumor immune response.
Collapse
Affiliation(s)
- Miran Jang
- Department of Medicine; The University of Chicago ; Chicago, IL USA
| | - Poh-Yin Yew
- Department of Medicine; The University of Chicago ; Chicago, IL USA
| | - Kosei Hasegawa
- Department of Gynecologic Oncology; Saitama Medical University International Medical Center ; Hidaka, Saitama, Japan
| | - Yuji Ikeda
- Department of Medicine; The University of Chicago ; Chicago, IL USA
| | - Keiichi Fujiwara
- Department of Gynecologic Oncology; Saitama Medical University International Medical Center ; Hidaka, Saitama, Japan
| | - Gini F Fleming
- Department of Medicine; The University of Chicago ; Chicago, IL USA
| | - Yusuke Nakamura
- Department of Medicine; The University of Chicago ; Chicago, IL USA ; Department of Surgery; The University of Chicago ; Chicago, IL USA
| | - Jae-Hyun Park
- Department of Medicine; The University of Chicago ; Chicago, IL USA
| |
Collapse
|
35
|
Stukan M, Leśniewski-Kmak K, Wróblewska M, Dudziak M. Management of symptomatic ascites and post-operative lymphocysts with an easy-to-use, patient-controlled, vascular catheter. Gynecol Oncol 2015; 136:466-71. [DOI: 10.1016/j.ygyno.2014.11.073] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 11/17/2014] [Accepted: 11/19/2014] [Indexed: 10/24/2022]
|
36
|
Intraperitoneal administration of cisplatin plus bevacizumab for the management of malignant ascites in ovarian epithelial cancer: results of a phase III clinical trial. Med Oncol 2015; 32:292. [PMID: 25609006 DOI: 10.1007/s12032-014-0292-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Accepted: 10/13/2014] [Indexed: 10/24/2022]
Abstract
Bevacizumab is a humanized antihuman VEGF-A monoclonal antibody. This study aims to evaluate the efficacy and safety of intraperitoneal administration of cisplatin plus bevacizumab (Avastin) in the management of malignant ascites in ovarian epithelial cancer. Fifty-eight ovarian epithelial cancer patients with malignant ascites were randomly assigned to receive either intraperitoneal administration of cisplatin only (control group, n = 27, cisplatin: 40 mg/m(2) every 2 weeks, for 6 weeks) or cisplatin plus bevacizumab (study group, n = 31, cisplatin: 40 mg/m(2), bevacizumab: 300 mg, every 2 weeks for 6 weeks). All patients regularly received TC regimen (paclitaxel 135 mg/m(2) d1 + carboplatin AUC 5 d1) every 3 weeks. The outcome, quality of life (QoL) and adverse effect of the treatment were analyzed, and VEGF and CA-125 level in ascites were detected by ELISA. After treatment with cisplatin plus bevacizumab, VEGF level in ascites was significantly decreased compared to baseline (P < 0.05). Meanwhile, ascites VEGF level of study group was significantly lower than that of control group (P < 0.05). The overall response rate (ORR) of study group was significantly higher than that of control group (ORR 90.32 vs. 59.26 %, P < 0.05). QoL improvement rate of study group was also significantly higher than that of control group (93.55 vs. 48.15 %, P < 0.05). All patients were well tolerated, and no serious adverse effect occurred. Intraperitoneal administration of cisplatin plus bevacizumab is effective and safe for the management of malignant ascites in ovarian epithelial cancer.
Collapse
|
37
|
A survey of treatment approaches of malignant ascites in Germany and Austria. Support Care Cancer 2014; 23:2073-8. [PMID: 25528551 DOI: 10.1007/s00520-014-2557-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2014] [Accepted: 12/07/2014] [Indexed: 01/26/2023]
Abstract
BACKGROUND Malignant ascites (MA) is a common manifestation of advanced cancer. Currently, there are no evidence-based guidelines for the management of MA. We conducted a survey with physicians throughout Germany and Austria, to get an overview of current approaches and opinions in the treatment of MA. METHODS One hundred and twenty-eight medical oncologists (MO), gastroenterologists (GE), and gynecologists (GYN) completed an electronic questionnaire consisting of 33 questions. RESULTS Ninety percent of the physicians were from Germany and 10% from Austria; 48% of those were MO, 30% were GYN, and 14% were GE. Most physicians treated an average of 34 patients (pts)/year with MA. Twenty-six percent of these pts suffered from ovarian, 20% from pancreatic, 17% from gastric, and 14% from colorectal cancer. The majority of the physicians associated MA with poor prognosis (92%) and significant reduction in quality of life (87%). One third felt that MA was a contraindication for full dosing of systemic chemotherapy. Paracentesis (PC) was performed in 70% of pts with symptom relieve and quality of life being the main reasons. Almost half of the pts required 3-5 PC, 50% even more than 5 PC during the course of their disease. Only 15% of pts needed multiple PC per week; the majority (79%) needed the procedure either once a week or every 14 days. In 61% of pts, 3-5 L of ascites fluid was drained. Only in 8%, 5 L and more were removed. Volume substitution with IV albumin was performed in 40% of pts. Most pts (55%) had to stay 1-3 h in a healthcare facility for the procedure. However, 21% had to stay ≥1 day. While almost all physicians (89%) performed a PC at some point in the treatment of MA, 75% felt that a systemic chemotherapy and 55% thought a concomitant diuretic therapy were a necessary adjunct. Seven percent of the pts received a targeted treatment with catumaxomab. CONCLUSIONS Repeated PC is the main pillar of treatment of MA; its effect is only temporary and requires significant hospital resources. Further treatment strategies of MA have to be evaluated in prospective studies. Targeted therapies like catumaxomab and VEGF inhibitors should be integrated into these.
Collapse
|
38
|
Matte I, Lane D, Laplante C, Garde-Granger P, Rancourt C, Piché A. Ovarian cancer ascites enhance the migration of patient-derived peritoneal mesothelial cells via cMet pathway through HGF-dependent and -independent mechanisms. Int J Cancer 2014; 137:289-98. [PMID: 25482018 DOI: 10.1002/ijc.29385] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2014] [Revised: 11/14/2014] [Accepted: 11/24/2014] [Indexed: 12/11/2022]
Abstract
Ovarian cancer ascites consist of a proinflammatory environment that is characterized by the presence of abundant human peritoneal mesothelial cells (HPMCs). Cytokines and growth factors in ascites modulate cell activities of tumor cells. The expression of proinflammatory cytokines in ascites is associated with a more aggressive tumor phenotype. The effect of ascites on HPMCs is for the most part unknown but this interplay is thought to be important for epithelial ovarian cancer (EOC) progression. Here, we examine the components of ascites, which stimulate patient-derived HPMC migration, from women with advanced EOC. We show that ovarian cancer ascites enhanced the migration of HPMCs. This effect was inhibited by heat treatment, hepatocyte growth factor (HGF) blocking antibodies and a HGF receptor (cMet) inhibitor. In ovarian cancer ascites, HGF is present at high concentration compared to benign fluids. Ascites-mediated activation of cMet was associated with Akt and EKR1/2 phosphorylation. This response was partly inhibited by heat treatment and cMet inhibitor. Ascites-induced migration and a cMet phosphorylation were strongly inhibited by epidermal growth factor receptor (EGFR) inhibitor PD153035, suggesting the transactivation of cMet by EGFR. Our study suggests that HGF and ligands of EGFR are factors that mediate ovarian cancer ascites-mediated migration of HPMCs by activating cMet and possibly downstream ERK1/2 and Akt pathways. The study provides evidence for the first time that ascites not only support tumor growth but also enhance the migratory potential of cancer-associated mesothelial cells, which in turn may support cancer progression.
Collapse
Affiliation(s)
- Isabelle Matte
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Canada
| | - Denis Lane
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Canada
| | - Claude Laplante
- Département de Pathologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Canada
| | - Perrine Garde-Granger
- Département de Pathologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Canada
| | - Claudine Rancourt
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Canada
| | - Alain Piché
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke, Sherbrooke, Canada
| |
Collapse
|
39
|
Narayanan G, Pezeshkmehr A, Venkat S, Guerrero G, Barbery K. Safety and efficacy of the PleurX catheter for the treatment of malignant ascites. J Palliat Med 2014; 17:906-12. [PMID: 24885753 DOI: 10.1089/jpm.2013.0427] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Malignant ascites is a common complication seen in association with various types of neoplastic processes. Due to high recurrence rates, patients may require multiple paracenteses, which have associated complications such as increased risk of bleeding, infection, pain, and volume and electrolyte depletion. OBJECTIVE This study evaluated the management of malignant ascites by placement of the PleurX® tunneled catheter system at a single center. METHODS This was a retrospective study of 38 patients who underwent PleurX catheter placement for refractory malignant ascites between February 2006 and March 2012 at our institution. Pretreatment characteristics and outcome measures were reported using descriptive statistics. RESULTS The population included 21 males and 17 females with a mean age of 60.6 years (range, 36-79 years) diagnosed with metastatic disease from a variety of primary malignancies, the most common of which was pancreatic cancer (10 patients). In 84% of patients (32/38) who were not lost to follow-up, mean survival time was 40.7 days (range 4-434 days). Technical success rate of catheter placement was 100%. CONCLUSIONS The PleurX catheter can be used to manage malignant ascites in severely ill patients with metastatic cancer, with a high rate of procedural success and a low incidence of potentially serious adverse events, infections, or catheter-related complications.
Collapse
Affiliation(s)
- Govindarajan Narayanan
- Department of Radiology, Vascular and Interventional Radiology, University of Miami , Miller School of Medicine, Miami, Florida
| | | | | | | | | |
Collapse
|
40
|
L’ascite non liée à la cirrhose : physiopathologie, diagnostic et étiologies. Rev Med Interne 2014; 35:365-71. [DOI: 10.1016/j.revmed.2013.12.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Revised: 10/03/2013] [Accepted: 12/02/2013] [Indexed: 12/31/2022]
|
41
|
Abdel Ghaffar MK, Hassan MS, Mostafa MY. Value of implantable peritoneal ports in managing recurrent malignant ascites. THE EGYPTIAN JOURNAL OF RADIOLOGY AND NUCLEAR MEDICINE 2014. [DOI: 10.1016/j.ejrnm.2014.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
42
|
Matte I, Lane D, Bachvarov D, Rancourt C, Piché A. Role of malignant ascites on human mesothelial cells and their gene expression profiles. BMC Cancer 2014; 14:288. [PMID: 24761768 PMCID: PMC4008408 DOI: 10.1186/1471-2407-14-288] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 04/03/2014] [Indexed: 12/12/2022] Open
Abstract
Background Malignant ascites is often present at diagnostic in women with advanced ovarian cancer (OC) and its presence is associated with a worse outcome. Human peritoneal mesothelial cells (HPMCs) are key components of malignant ascites. Although the interplay between HPMCs and OC cells is believed to be critical for tumor progression, it has not been well characterized. The purpose of this study was to assess the effect of ascites on HPMCs and clarify the role of HPMCs in OC progression. Methods Human OC ascites and benign peritoneal fluids were assessed for their ability to stimulate HPMC proliferation. Conditioned medium from ascites- and benign fluid-stimulated HPMCs were compared for their ability to attenuate apoptosis induced by TNF-related apoptosis-inducing ligand (TRAIL). We conducted a comparative analysis of global expression changes in ascites-stimulated HPMCs using Agilent oligonucleotide microarrays. Results As compared to benign peritoneal fluids, malignant ascites stimulated the proliferation of HPMCs. TRAIL-induced apoptosis was attenuated in OC cells exposed to conditioned medium from ascites-stimulated HPMCs as compared to OC cells exposed to conditioned medium from benign fluid-stimulated HPMCs. A total of 649 genes were differentially expressed in ascites-stimulated HPMCs. Based on a ratio of more than 1.5-fold and a P < 0.05, 484 genes were up-regulated and 165 genes were down-regulated in ascites-exposed HPMCs. Stimulation of HPMCs with OC ascites resulted in differential expression of genes mainly associated with the regulation of cell growth and proliferation, cell death, cell cycle and cell assembly and organization, compared to benign peritoneal fluids. Top networks up-regulated by OC ascites included Akt and NF-κB survival pathways whereas vascular endothelial growth factor (VEGF) pathway was down-regulated. Conclusions The results of this study not only provide evidence supporting the importance of the interplay between cancer cells and HPMCs but also define the role that the tumor environment plays in these interactions.
Collapse
Affiliation(s)
| | | | | | | | - Alain Piché
- Département de Microbiologie et Infectiologie, Faculté de Médecine, Université de Sherbrooke, 3001, 12ième Avenue Nord, Sherbrooke, Québec, J1H 5N4, Canada.
| |
Collapse
|
43
|
Clinicopathological characteristics and prognosis of gastric cancer with malignant ascites. Tumour Biol 2013; 35:3261-8. [DOI: 10.1007/s13277-013-1426-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/13/2013] [Indexed: 12/17/2022] Open
|
44
|
Zhang HQ, Xie M, He B, Lu S, Wan YY, Song RF. Clinicopathological features and prognostic factors of gastric cancer patients with malignant ascites: Analysis of 223 cases. Shijie Huaren Xiaohua Zazhi 2013; 21:2826-2831. [DOI: 10.11569/wcjd.v21.i27.2826] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To analyze the clinicopathological features and prognostic factors of gastric cancer patients with malignant ascites.
METHODS: Clinicopathological data for 223 gastric cancer patients with malignant ascites treated at the Jiangxi Provincial Tumor Hospital between January 2008 and December 2012 were retrospectively analyzed. Survival analysis was conducted using Kaplan-Meier method. Factors influencing survival were analyzed using univariate (Log-rank) and multivariate (Cox) models.
RESULTS: A total of 223 patients (95 females and 128 males, median age, 52 years) were included, accounting for 6.2% of all gastric cancer cases treated at our hospital during the same period. Among them, 50 (22.4%) were young, 38 (17.0%) were senile, 122 (54.7%) underwent curative gastric resection or cytoreductive surgery, 97 (43.5%) presented with malignant ascites at the initial diagnosis of gastric cancer, and 148 (66.4%) received systematic and/or intraperitoneal chemotherapy. Proximal gastric cancer was diagnosed in 22 cases (9.9%) and whole stomach cancer in 12 cases (5.4%). Approximately 79.4% (177/223) of cases were diagnosed with poorly differentiated adenocarcinoma or undifferentiated adenocarcinoma. After a median follow-up period of 9.7 months, it was found that the median survival following diagnosis of malignant ascites was 4.9 months, and the overall one-year survival rate was 12.6%. Univariate analysis revealed that ECOG score, presence of malignant ascites at the initial diagnosis of gastric cancer, history of gastric cancer surgery, total bilirubin level, presence of metastasis in other site(s), and chemotherapy were significant factors affecting the survival (all P < 0.05). Multivariate analysis showed that ECOG score (P = 0.01), presence of malignant ascites at the initial diagnosis of gastric cancer (P = 0.001), presence of metastasis in other site(s) (P = 0.002) and chemotherapy (P < 0.001) were independent prognostic factors.
CONCLUSION: The survival of gastric cancer patients with malignant ascites is relatively short. Worse ECOG score, presence of malignant ascites at the initial diagnosis and presence of metastasis in other site(s) are associated with poor prognosis in gastric cancer patients with malignant ascites, and the survival time could be prolonged by systematic and intraperitoneal chemotherapy.
Collapse
|
45
|
Kipps E, Tan DSP, Kaye SB. Meeting the challenge of ascites in ovarian cancer: new avenues for therapy and research. Nat Rev Cancer 2013; 13:273-82. [PMID: 23426401 PMCID: PMC4673904 DOI: 10.1038/nrc3432] [Citation(s) in RCA: 412] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Malignant ascites presents a considerable clinical challenge to the management of ovarian cancer, but also provides a wealth of opportunities for translational research. The accessibility of ascitic fluid and its cellular components make it an excellent source of tumour tissue for the investigation of prognostic and predictive biomarkers, pharmacodynamic markers and for molecular profiling analysis. In this Opinion article, we discuss recent advances in our understanding of its pathophysiology, the development of new methods to characterize its molecular features and how these findings can be used to improve the treatment of malignant ascites, particularly in the context of ovarian cancer.
Collapse
Affiliation(s)
- Emma Kipps
- The Institute of Cancer Research/Royal Marsden Hospital, Medicine, Downs Road, Sutton SM2 5PT, UK
| | | | | |
Collapse
|
46
|
Abstract
Catumaxomab is a rat/murine hybrid, trifunctional, bispecific (anti-human epithelial cell adhesion molecule [EpCAM] × anti-CD3) monoclonal antibody. Compared with paracentesis alone, paracentesis followed by catumaxomab therapy was associated with significant prolongation of paracentesis-free survival and time to repeat paracentesis in a randomized, open-label, multicentre, pivotal phase II/III trial in patients with recurrent symptomatic malignant ascites due to EpCAM-positive tumours who were resistant to conventional chemotherapy. The benefits of catumaxomab were seen across a broad range of epithelial ovarian and nonovarian cancers, and irrespective of whether or not catumaxomab recipients developed human anti-mouse antibodies. Combining catumaxomab with paracentesis also resulted in more pronounced and prolonged reductions in ascites signs and symptoms and a delayed deterioration in health-related quality of life compared with paracentesis alone. Despite the study not being designed or powered to evaluate overall survival, significant differences favouring the addition of catumaxomab to paracentesis were seen in analyses of the safety population and the subpopulation of patients with gastric cancer. Catumaxomab was generally well tolerated in the pivotal phase II/III trial. The most frequent adverse events attributed to catumaxomab treatment included cytokine-release-related symptoms, which were mostly of mild to moderate severity and manageable with standard symptomatic treatment.
Collapse
|
47
|
Crawford B, Piault E, Gotlieb W, Joulain F. Development and validation of the self-completed ascites impact measure to understand patient motivation for requesting a paracentesis. PATIENT-RELATED OUTCOME MEASURES 2012; 3:21-30. [PMID: 22915981 PMCID: PMC3417936 DOI: 10.2147/prom.s28179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2012] [Indexed: 11/23/2022]
Abstract
Background: The Ascites Impact Measure (AIM) was developed to record patients’ daily experiences of symptoms that trigger a request for a paracentesis. Methods: Development of the AIM followed a rigorous step-wise approach, including a review of the literature, expert opinions, and qualitative research involving patients who experience symptomatic malignant ascites. The AIM’s measurement properties were assessed using data from two international trials, including 59 ovarian cancer patients with symptomatic malignant ascites. Results: Following the literature review and expert discussions to develop the conceptual model, ten patients with symptomatic malignant ascites were interviewed in the item elicitation phase, resulting in a draft questionnaire with four questions. Validation analyses consisted of 59 patients pooled from two international trials. Inter-items correlations for the AIM were good (r > 0.60), except for the Pain item. Internal consistency reliability (α = 0.89) improved after removing the Pain item from the Total Symptom score (TSS). Test-retest reliability was sufficient. Scores significantly improved after paracentesis except for the Pain item. Preliminary estimates indicate that a two-point improvement on the three-item TSS (without the Abdominal Pain item) could be interpreted as clinically meaningful. Conclusion: The Abdominal Pain item appears to behave differently than the other three items, and could be more related to cancer. While the validity of the AIM TSS (four-item) is acceptable, removing the Pain item from the TSS scoring algorithm demonstrated better construct validity. In addition, test-retest reliability and responsiveness were found to be similar to the results for the four-item AIM TSS. The Pain item should be used as a supplemental item to the three-item AIM TSS, as it provides additional information about the underlying cancer state.
Collapse
|
48
|
Masoumi Moghaddam S, Amini A, Morris DL, Pourgholami MH. Significance of vascular endothelial growth factor in growth and peritoneal dissemination of ovarian cancer. Cancer Metastasis Rev 2012; 31:143-162. [PMID: 22101807 PMCID: PMC3350632 DOI: 10.1007/s10555-011-9337-5] [Citation(s) in RCA: 151] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a key regulator of angiogenesis which drives endothelial cell survival, proliferation, and migration while increasing vascular permeability. Playing an important role in the physiology of normal ovaries, VEGF has also been implicated in the pathogenesis of ovarian cancer. Essentially by promoting tumor angiogenesis and enhancing vascular permeability, VEGF contributes to the development of peritoneal carcinomatosis associated with malignant ascites formation, the characteristic feature of advanced ovarian cancer at diagnosis. In both experimental and clinical studies, VEGF levels have been inversely correlated with survival. Moreover, VEGF inhibition has been shown to inhibit tumor growth and ascites production and to suppress tumor invasion and metastasis. These findings have laid the basis for the clinical evaluation of agents targeting VEGF signaling pathway in patients with ovarian cancer. In this review, we will focus on VEGF involvement in the pathophysiology of ovarian cancer and its contribution to the disease progression and dissemination.
Collapse
Affiliation(s)
- Samar Masoumi Moghaddam
- Cancer Research Laboratories, Department of Surgery, St George Hospital, University of New South Wales, Sydney, NSW 2217 Australia
| | - Afshin Amini
- Cancer Research Laboratories, Department of Surgery, St George Hospital, University of New South Wales, Sydney, NSW 2217 Australia
| | - David L. Morris
- Department of Surgery, St George Hospital, University of New South Wales, Sydney, NSW 2217 Australia
| | - Mohammad H. Pourgholami
- Cancer Research Laboratories, Department of Surgery, St George Hospital, University of New South Wales, Sydney, NSW 2217 Australia
| |
Collapse
|
49
|
Targeted endostatin-cytosine deaminase fusion gene therapy plus 5-fluorocytosine suppresses ovarian tumor growth. Oncogene 2012; 32:1082-90. [PMID: 22562248 DOI: 10.1038/onc.2012.134] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
There are currently no effective therapies for cancer patients with advanced ovarian cancer, therefore developing an efficient and safe strategy is urgent. To ensure cancer-specific targeting, efficient delivery, and efficacy, we developed an ovarian cancer-specific construct (Survivin-VISA-hEndoyCD) composed of the cancer specific promoter survivin in a transgene amplification vector (VISA; VP16-GAL4-WPRE integrated systemic amplifier) to express a secreted human endostatin-yeast cytosine deaminase fusion protein (hEndoyCD) for advanced ovarian cancer treatment. hEndoyCD contains an endostatin domain that has tumor-targeting ability for anti-angiogenesis and a cytosine deaminase domain that converts the prodrug 5-fluorocytosine (5-FC) into the chemotherapeutic drug, 5-fluorouracil. Survivin-VISA-hEndoyCD was found to be highly specific, selectively express secreted hEndoyCD from ovarian cancer cells, and induce cancer-cell killing in vitro and in vivo in the presence of 5-FC without affecting normal cells. In addition, Survivin-VISA-hEndoyCD plus 5-FC showed strong synergistic effects in combination with cisplatin in ovarian cancer cell lines. Intraperitoneal (i.p.) treatment with Survivin-VISA-hEndoyCD coupled with liposome attenuated tumor growth and prolonged survival in mice bearing advanced ovarian tumors. Importantly, there was virtually no severe toxicity when hEndoyCD is expressed by Survivin-VISA plus 5-FC compared with CMV plus 5-FC. Thus, the current study demonstrates an effective cancer-targeted gene therapy that is worthy of development in clinical trials for treating advanced ovarian cancer.
Collapse
|
50
|
Hess J, Ruf P, Lindhofer H. Cancer therapy with trifunctional antibodies: linking innate and adaptive immunity. Future Oncol 2012; 8:73-85. [PMID: 22149036 DOI: 10.2217/fon.11.138] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Trifunctional antibodies (trAbs) are promising novel anticancer biologics with a particular mode of action capable of linking innate with adaptive immunity. Based on their unique structure, trifunctional IgG-like heterodimeric antibodies, consisting of nonhuman mouse and rat immunoglobulin halves are able to redirect T lymphocytes, as well as accessory cells, to the tumor site. This recruitment of immune cells is accompanied by cellular activation events elicited by anti-CD3, as well as Fcγ-receptor engagement of trAbs supported by a proinflammatory Th1-biased cytokine milieu. All necessary immunological factors required for long-term vaccination-like effects are stimulated along trAb-mediated therapeutic interventions. Thus, the concerted interplay of antibody-dependent cellular cytotoxicity plus the polyclonal T-cell cytotoxicity and Fcγ-receptor-driven induction of long-lasting immune responses after the initial tumor cell elimination represent the major hallmarks of trAb-mediated treatment of malignant diseases.
Collapse
Affiliation(s)
- Juergen Hess
- TRION Pharma GmbH, Frankfurter Ring 193a, 80807 Munich, Germany
| | | | | |
Collapse
|